SEARCH RESULTS FOR: Inflammation

Myocardial Infarction: Findings on History

Yu Yan - MI Findings on History - FINAL.pptx - 
Myocardial Infarction: Findings on HistoryLegend:Published January 30, 2013 on www.thecalgaryguide.comMechanismPathophysiologySign/Symptom/Lab FindingComplicationsAuthor:  Yan YuReviewers:Sean SpenceTristan JonesNanette Alvarez** MD at time of publication Systolic function(necrotic myocardium cannot contract as well)Reflexive ? in sympathetic activity (to try to maintain CO)Clammy skin? stroke volume (SV), ? cardiac output (CO)Myocardial infarction (tissue necrosis)Note: Myocardial ischemic pain may differ between patients, but recurrences usually feel the same in any given patient.Generalized vasoconstrictionVasoconstriction of skin arteriolesCool skinLocal myocardial inflammationIrritation of T1-T4 sympathetic afferentsIrritation of cardiac branches of vagus nerveSignals enter spinal cord, mixes with T1-T4 dermatomesCrushing, Diffuse L).(Onset: often at rest; crescendo)Activation of reflexive vagal responses (listed below)Weakness, dizziness, nausea, vomitingInflammatory mediators irritates nerves innervating the heart (the cardiac plexus)Cytokines act on hypothalamic T0 regulatorMild fever? Sweating (diaphoresis)Inflammatory cytokines can spread systemicallyBrain perceives nerve irritation as pain coming from T1-T4 dermatomesBlood backs up from the LV, into the left atrium and eventually accumulates in the pulmonary vasculatureHigh pulmonary venous blood pressure forces fluid out of capillaries, into pulmonary interstitium & alveoliRespiratory muscles work harder to ventilate lungsSoggier lung interstitium ? lung complianceDyspnea(Shortness of breath)Fluid compresses airways, ? resistance to airflow 102 kB / 204 words" title="Yu Yan - MI Findings on History - FINAL.pptx - Myocardial Infarction: Findings on HistoryLegend:Published January 30, 2013 on www.thecalgaryguide.comMechanismPathophysiologySign/Symptom/Lab FindingComplicationsAuthor: Yan YuReviewers:Sean SpenceTristan JonesNanette Alvarez** MD at time of publication Systolic function(necrotic myocardium cannot contract as well)Reflexive ? in sympathetic activity (to try to maintain CO)Clammy skin? stroke volume (SV), ? cardiac output (CO)Myocardial infarction (tissue necrosis)Note: Myocardial ischemic pain may differ between patients, but recurrences usually feel the same in any given patient.Generalized vasoconstrictionVasoconstriction of skin arteriolesCool skinLocal myocardial inflammationIrritation of T1-T4 sympathetic afferentsIrritation of cardiac branches of vagus nerveSignals enter spinal cord, mixes with T1-T4 dermatomesCrushing, Diffuse "Pain" or "tightness": Often retrosternal, with radiation to shoulder, neck, and inner aspect of both arms (R > L).(Onset: often at rest; crescendo)Activation of reflexive vagal responses (listed below)Weakness, dizziness, nausea, vomitingInflammatory mediators irritates nerves innervating the heart (the cardiac plexus)Cytokines act on hypothalamic T0 regulatorMild fever? Sweating (diaphoresis)Inflammatory cytokines can spread systemicallyBrain perceives nerve irritation as pain coming from T1-T4 dermatomesBlood backs up from the LV, into the left atrium and eventually accumulates in the pulmonary vasculatureHigh pulmonary venous blood pressure forces fluid out of capillaries, into pulmonary interstitium & alveoliRespiratory muscles work harder to ventilate lungsSoggier lung interstitium ? lung complianceDyspnea(Shortness of breath)Fluid compresses airways, ? resistance to airflow 102 kB / 204 words" />

myocardial-infarction-findings-on-investigations

Yu Yan - MI Findings on Investigations - FINAL.pptx
Myocardial Infarction: Findings on InvestigationsAuthor:  Yan YuReviewers:Sean SpenceTristan JonesNanette Alvarez** MD at time of publicationTissue ischemia disrupts normal cardiac electrical conduction(detected on serial ECG)Acute, trans-mural myocardial ischemiaIschemia of sub-endocardial myocardiumMyocardial infarctionNote: Both types of ST-segment changes are non-specific: they can indicate Myocardial Infarctions , but can also be false positives (i.e. caused by left ventricular hypertrophy, bundle branch blocks, and other non-myocardial ischemic causes)If ischemia progresses to tissue infarctionPathologic Q-waves (localizes to site of ischemia)Tissue necrosis ? Local myocardial inflammation2-4 hours after MI: troponin proteins released into blood3-8 hours after MI:Creatinine-kinase MB-isozymes released into blood? serum Cardiac Troponins: cTnT, cTnI(Sensitive and most specific serum marker for myocardial necrosis)Relatively faster clearance from circulation? serum CK-MB(less sensitive and specific for myocardial necrosis than Troponins)ST-segment depression(non-localizing)Inflammatory cytokines can spread systemicallyStimulation of neutrophil and monocyte migration towards area of inflammation? WBC count (on CBC)? C-Reactive Protein (CRP)Dead, damaged cardiac myocytes release inner contents into the bloodRelatively slower clearance from circulationSerum CK-MB levels normalize within 3 daysSerum Troponin levels normalize within 14 daysNote: Measuring both CK-MB and Troponins gives a timeline to the MI. For instance, if CK-MB is normal but Troponins are high, it means the MI happened >3 days but <14 days ago.ST-segment elevation(localizes to site of ischemia)Legend:Published January 30, 2013 on www.thecalgaryguide.comMechanismPathophysiologySign/Symptom/Lab FindingComplications
104 kB / 212 words

systemic-lupus-erythematosus-gastrointestinal-manifestations

Systemic Lupus Erythematosus: Gastrointestinal Manifestations 
Abbreviations: • APLA Phospholipid — Anti-Antibodies 1— Production of auto-antibodies APLA in circulation promotes blood coagulation —01• Thrombosis of vessels in the pancreas Micro-thrombi of vessels in the liver Liver infarcts Thrombosis of vessels in the small intestine Acute Pancreatitis Death of liver cells release contents into blood stream Thrombus in the hepatic vein • Vascular Damage Mesenteric Vasculitis/Ischemia Budd Chiari Syndrome Damage to Auerbach's Plexus 11` Liver Enzymes 1` in capillary permeability 
Abnormal release of 
inflammatory cytokines 
1 
Inflammation in esophageal muscles 
Protein-Losing Enteropathy 
Ascites 
Note: The gastrointestinal manifestations of systemic lupus erythematosus are due to multifactorial and complex causes, some of which are unknown. 
Legend: Pathophysiology 
Mechanism 
Impaired motor innervation 
Authors: Joseph Tropiano Reviewers: Zaini Sarwar Harjot Atwal Liam Martin* * MD at time of publication 
Abnormal production of immune complexes 
Immune complex deposition in blood vessels 
Immune complex deposition in smooth muscle 
Inflammatory reaction in the GI tract 
Immune complex mediated vasculitis 
Chronic ischemia of bowel smooth muscle 
Muscular damage 

Myopathy or neurogenic pathology of the GI muscles 
GI muscle damage not severe enough to inhibit peristalsis completely 
Esophageal Motility Disorders 
Dysphagia 
Sign/Symptom/Lab Finding 
Complications 

Hypomotility 
Dysmotility

esophageal-gastric-varices

Esophageal/Gastric Varices: Pathogenesis and clinical findings 
Schistosomiasis 
Schistosoma species enter the body through the skin and circulate to liver 
Eggs lodge in terminal portal venules causing inflammation and fibrosis • 1` resistance through fibrosed and inflamed sinusoids 
Cirrhosis Liver disease activates hepatic stellate cells causing hepatic fibrosis • I` resistance through fibrosed and distorted sinusoids • 1` portal inflow due to splanchnic vasodilation 
Veno-Occlusive Disease Budd-Chiari Syndrome Endothelial damage Hypercoagulable in the sinusoids leads to clotting states* cause factor deposition in thrombosis of hepatic sinusoids hepatic veins 1` resistance t resistance through through hepatic occluded distal veins occluded sinusoids by thrombus 

► Intra Hepatic Portal Hypertension 
Post Hepatic Portal 
Portal Vein Thrombosis Hypercoagulable states* cause thrombosis of portal vein 
Infiltrative Lesion 
• Primary or secondary malignancy localized to the portal vein 
Splenic Vein Thrombosis 
Pancreatitis leads to inflammation and thrombosis of the splenic vein 
1 resistance through '1' resistance through 1` resistance through portal vein occluded portal vein occluded by splenic vein occluded by thrombus malignancy by thrombus 

Pre Hepatic Portal 
Hypertension Hypertension 
*Hypercoagulable states such as thrombophilia, malignancy, or connective tissue disease Portal esophageal/gastric Esophageal/Gastric blood flow backed anastomoses up into Varices As variceal pressure 1` vessels swell 4— Blood loss from circulation 1 vessel J, wall thickness 1 vessel size tension Dilation of veins in submucosa Blood oxidized and vomited or passed through GI Authors: Bigger Varices  Variceal rupture Upper GI bleed Gabriel Burke Reviewers: Vadim lablokov Laura Byford-Richardson Meredith Borman* * MD at time of publication • Red Wale Mark or Cherry Red Spot Blood loss too rapid to be oxidized before emesis or passage of GI (visualized on endoscopy)  
Legend: 
Pathophysiology Mechanism 
Sign/Symptom/Lab Finding 
Complications 
• Venous drainage of spleen backed up into gastric anastomoses 
Tachycardia and hypotension 
Anemia Death Melena  Coffee ground emesis Hematemesis  Bright red blood per rectum

central-retinal-artery-occlusion-pathogenesis-and-clinical-findings

Central Retinal Artery Occlusion: Pathogenesis and clinical findings 
Inflammatory Disease: Cardiogenic Embolism: Hypercoagulable state: Hematologic Disease: (i.e. GCA, SLE, GPA) (i.e. Valvular, arrhythmias, congenital defects) (i.e. OCP, Protein C&S deficiency, ATIII deficiency) (i.e. leukemia/lymphoma, sickle cell, polycythemia) Endothelial cell damage Abnormal blood flow 1` coagulation and/or 1 blood viscosity and creates hypercoagulable state causing localized stasis 4, anti-coagulation inflammation 
Abbreviations: • GCA — Giant Cell Arteritis • SLE — Systemic Lupus Erythematosus • GPA — granulomatosis with polyangitis • OCP — Oral contraceptive pill • ATIII — Anti-thrombin Ill 
Thrombus formation 
Blockage of central retinal artery 
Central Retinal Artery Occlusion (CRAO) 
Authors: Graeme Prosperi-Porta Reviewers: Stephanie Cote Usama Malik Johnathan Wong* * MD at time of publication Carotid Artery Atherosclerosis 
Atherosclerotic plaque dislodges from carotid artery 
The retina becomes pale 4, perfusion of retinal Slow retinal artery blood Acute retinal edema Ganglion cells and axons from NI, perfusion arterioles due to upstream flow allows for caused by ischemia results death due to ischemia CRAO segmentation of the blood column in a blurred appearance of the retina results in disc pallor seen months after CRAO The choroidal vessels supplying the macula via the posterior ciliary artery become more prominent within a background of retinal pallor

infectious-esophagitis-pathogenesis-and-clinical-findings

Infectious Esophagitis: Pathogenesis and clinical findings 
HIV/AIDS Radiation therapy Chemotherapy Organ Transplant Antibiotics I/Esophageal motility Tir CD4+ T cells 4,Monocyte and Corticosteroid and granulocyte precursors anti-TNF therapy • • 
Immunosuppression 
Note: Bacterial causes of infectious esophagitis are difficult to isolate as they are often polymicrobial in nature and derived from normal oral flora. 
Cytomegalovirus Infection of endothelial cells and fibroblasts 
I, Protective flora 4, Pathogen clearance 
Authors: David Deng Reviewers: Peter Bishay Vadim lablokov Kirles Bishay* MD at time of publication 
Mechanical stricture Inflammation Ulceration ,..,4 Dysphagia Infectious Viral Bacterial infection infection esophagitis • Fungal infection Odynophagia (i.e. Candida) 
Herpes Simplex Infection of squamous cells and macrophages 
Colonization facilitated by use of antacid therapy 
Nuclear Large Superficial Squamous Macrophage inclusion bodies esophageal ulceration ulcers cell inclusion bodies aggregation Legend: Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications 
Spores and pseudohyphae seen on biopsy 
• Invas.on of underlying blood vessels 
• White plaques over erythematous base 
'1' neutrophils due to inflammatory response

Primary Spontaneous Pneumothorax: Pathogenesis and clinical findings

Primary Spontaneous Pneumothorax: Pathogenesis and clinical findings 
Thoracic Tall, thin endometriosis males 
Genetic Factors (i.e. FLCN mutations, HCY, MFS, CTD) 
Malnutrition Smoking 
Structurally compromised lung parenchyma 
Notes: • PSPs usually occur at rest • Respiratory symptoms vary in severity • Suspect thoracic endometriosis in young women with recurrent PSPs that coincide with menstruation • *Pathophysiology of tension pneumothorax is described in a separate slide 
Air leaks into the subcutaneous tissue 
Subcutaneous emphysema 
Authors: Lauren Hampton Reviewers: Kening (Midas) Kang Natalie Morgunov Sadie Kutz Usama Malik Leila Barss* * MD at time of publication 
Thoracic Ischemia endometriosis 
Mechanical forces of respiration create blebs Inflammation disrupts mesotheial and/or bullae ~ cell layer of the visceral pleura 
47 
Spontaneous rupture of blebs or bullae 
47 
Sudden onset pleuritic chest pain 
71r 
Primary spontaneous pneumothorax: Presence or introduction of air in the pleural space in a patient WITHOUT diagnosed or clinically apparent lung disease Tachycardia Abbreviations: Communication occurs between the alveoli and pleural space • FLCN- Folliculin gene • HCY- Homocystinuria • MFS- Marfan syndrome Alveolar pressure > pleural pressure • CTD- Connective tissue disease • PSP- Primary spontaneous Air from the lungs enters the pleural space pneumothorax • V/Q- ventilation/perfusion Air separates the chest from /1` intrapleural pressure • Sp02- oxygen saturation the lung parenchyma Small areas of lung collapse Blood flow to areas of On affected side: under un-opposed intrinsic atelectasis is maintained -• Si, chest wall expansion elastic recoil while ventilation 4, t resonance to percussion Si, or absent tactile fremitus Si, or absent breath sounds 4, lung compliance Shunting and V/Q mismatch Pleural line on chest x-ray 1` work of breathing Tension pneumothorax* Accessory muscle use, 4• Sp02, Sudden onset dyspnea, Tachycardia Hypotension, Juglar venous Tachypnea  distension, Pulsus paradoxus

Asthma Acute Exacerbation: Pathogenesis and Treatment

Asthma Acute Exacerbation: Pathogenesis and Treatment 
Viral URI 
Allergen 
Pollution 
Other Triggers 
Activation of immune system: Epithelial chemokine activation, lymphocyte activation, macrophage activation, t leukotriene production 
Inflammation of lower airway 
• Dyspnea 
Air flows past inflamed airways causes t irritation 
Cough and wheezing 
Release of inflammatory mediators 
Mucosal edema causing turbulent air flow 7Ir Wheezing 
Notes • Asthma: Airway hyper-responsiveness causing airflow obstructions • Acute Exacerbation (Asthma): An episode of increased symptoms due to decreases in airflow 
Abbreviations • PCO2: Partial pressure of CO, in arterial blood • PEF: Peak expiratory flow • SABA: Short-acting beta-2 agonists • Sp02 : Blood oxygen saturation level 
Mild to moderate  exacerbation: PEF 50% of predicted 
Titrate O2 toSpO2, 92%, give SABA & steroids ■  
Good response:  symptoms  resolved, PEF > 80% 

[Treat at home with SABA as needed and steroids 
Dyspnea 
Bronchoconstriction 
1` Residual volume and 1` PCO2 

Respiratory failure 
1` Air trapping causes '1' intra-alveolar pressure 
Severe exacerbation: PEF 50% of predicted Educate patient regarding medications, Loss of Pulsus inhaler technique & [consciousness paradoxus follow up with primary care provider  I 
Legend: Pathophysiology Mechanism 
Titrate O2 to402 93%, give SABA, steroids & magnesium sulfate 
Sign/Symptom/Lab Finding 
{Worsening symptoms and/or respiratory failure: Do not delay intubation, send to ICU, give SABA, steroids & magnesium sulfate 
Authors: Luke Gagnon Reviewers: Midas (Kening) Kang Usama Malik Lian Szabo* * MD at time of publication 
4, Delivery of oxygen rich air to alveoli 4, Oxygenation of blood 
Drowsy and confused  
Central  cyanosis 
• Tachycardia 
Pneumothorax 
[Depending on 1 severity: Observation or place chest tube

Bronchiectasis Pathogenesis and clinical findings

Bronchiectasis: Pathogenesis and clinical findings 
Acquired immunodeficiency Lymphoma, HIV, transplant 
Autoimmune Lupus, inflammatory bowel disease, rheumatoid arthritis 
Congenital/Genetic Cystic fibrosis, A1AT deficiency, Marfan, immunoglobulin deficiency, Kartagener syndrome, Young syndrome 
Endobronchial obstruction Neoplasm, foreign body, lymph node compression 
Other Inhalation exposure (smoke, ammonia), MAC complex infection, COPD, allergic bronchopulmonary aspergillosis, chronic infections 
Irreversibly dilated bronchi 
Chronic bronchial infection and inflammation 
1 
Easily collapsible airways 
 I Bronchiectasis (persistent and progressive damage to lungs) 
Chronic cough  (mucopurulent) 
Defect in immunity and/or mucus clearance 
Persistent bacteria in airway (commonly Pseudomonas/Staph aureus) 
Inflammatory response 
Rhinosinusitis 
Abbreviations: • A1AT — Alpha-1-antitrypsin • COPD — Chronic Obstructive Pulmonary Disease • HIV — Human Immunodeficiency Virus • MAC — Membrane Attack Complex • VQ— Ventilation/Perfusion ratio 
Legend: 
Pathophysiology Mechanism 
Fever 
Sign/Symptom/Lab Finding 
Failure to thrive (children)  

Authors: Rebecca (Becky) Phillips Reviewers: Midas (Kening) Kang Usama Malik Eric Leung* * MD at time of publication 
Notes: • Can be focal (single lobe/segment) or diffuse (both lungs) • Mainly in elderly • 1% prevalence in children 
Tissue damage 
Epithelial destruction of airways 
Further impairment of bacterial clearance 
Persistent inspiratory adventitious sounds  (crackles > wheezing)  
Complications 
Structural damage to bronchial walls 
Obstructive pulmonary function tests  
Hemoptysis 
Chest pain 
VQ mismatch and 4, gas exchange 
4, oxygenation 

Digital  clubbing (rare)  
Fatigue Dyspnea  
Cyanosis  (uncommon)

Benign Prostatic Hyperplasia: Pathogenesis and medications

Benign Prostatic Hyperplasia: Pathogenesis and medications 
Aging 
Testosterone 
Testosterone metabolized into DHT by type II 5- a-reductase in prostate 
DHT binds to androgen receptor in prostate cell nuclei 
Hyperplasia of the prostate 
Prostate encapsulated by fibromuscular tissue, therefore grows inwards 
Prostatic urethral compression and bladder outlet obstruction 
Legend: 
a-1 blockers (e.g. tamsulosin) 
Note: MoA not fully established 
PDE-5 inhibitors (e.g. tadalafil) 
Bladder and prostate smooth-muscle a-1 receptor antagonism 
—110. 
Relaxation of bladder outlet and prostate smooth-muscle 
Authors: Michael Korostensky Reviewers: Alex Tang Usama Malik Dr. Jay Lee* * MD at time of publication 
Acronyms: 5-ARI = 5-a reductase inhibitors COX = cyclooxygenase DHT = dihydrotestosterone GnRH = gonadotropin-releasing hormone LUTS = lower urinary tract symptoms PDE-5-mediated cGMP degradation in prostate smooth-Improved urinary outflow muscle and associated vascular supply Relaxation of prostate smooth-muscle MoA = mechanism of action NSAID = nonsteroidal anti-inflammatory drugs PDE-5 = phosphodiesterase-5 -NO 
5-ARIs (e.g. dutasteride) 
LHRH receptor antagonists (e.g. cetrorel ix) 
P3-adrenergic agonists (e.g. mirabegron) 
anticholinergics (e.g. oxybutynin) 
NSAIDs 
1` Bladder pressures 
Pathophysiology Mechanism 
5-a-reductase activity 
1, Conversion of testosterone into DHT 
4, Progression of LUTS 
1, Testosterone secretion from testicular Leydig cells 1, LH secretion from pituitary GnRH antagonism DHT production Relaxation of detrusor Bladder muscle 1` capacity Improved LUTS  
Acetylcholine antagonism at muscarinic receptors Relaxation of bladder outlet smooth-muscle 1` volume to first detrusor contraction 4, Prostaglandin release Analgesia and 4, Prostatic ,f, COX activity ► inflammation  —110. Bladder smooth-muscle hyperplasia (detrusor thickening) /1` Sensitivity (i.e. overactive detrusor) -1110. 1, Volume to first detrusor contraction LUTS

Mixed Urinary Incontinence Pathogenesis and clinical findings

Mixed Urinary Incontinence: Pathogenesis and clinical findings 
Abbreviations: • BOO — Bladder Outlet Obstruction • BPH — Benign Prostatic Hyperplasia • CNS — Central Nervous System • IAP — Intra-abdominal pressure • OAB — Over-Active Bladder • PVR — Post Void Residual • SUI —Stress Urinary Incontinence • UTI — Urinary Tract Infection • UUI — Urge Urinary Incontinence 
Mixed Urinary Incontinence 47 
Urinary leakage accompanied by both urgency and t intra-abdominal pressure 
Urgency Urinary Incontinence (UUI) 4, Urinary leakage preceded by a sudden, strong urge to void 
Overflow Incontinence vir Overfilling of the bladder from obstruction; BOO (tumour, stone, BPH, urethral or bladder neck stricture) 
Detrusor Overactivity Ilr OAB (idiopathic), CNS lesion (neurogenic), inflammation/ infection (cystitis, UTI), diabetes mellitus 
4. Bladder Wall Compliance 
Progressive t in intravesicle pressure during bladder filling pushing urine from the bladder 
Authors: Braden Millan Reviewers: Alex Tang Usama Malik Jay C. Lee* * MD at time of publication 
Stress Urinary Incontinence (SUI) + Episodic involuntary urinary leakage with sudden l• in intra-abdominal pressure 
4. 
Urethral hypermobility, intrinsic sphincter deficiency, or a poorly coapting urethra 
4, 
4, Pelvic floor muscle and ligament strength causing 4. tone of vesicoureteral sphincter unit; 4, urethral strength and associated striated and smooth muscle; iatrogenic 
Legend: 
Failure to Void  Weak Stream (+ dribbling), Intermittent, Straining, '1` PVR if a complication of urinary retention; obstruction visible on cystoscopy 
Failure to Store  Frequency, Urgency, Nocturia, Dysuria if SUI or UUI not caused by obstruction 
Pathophysiology Mechanism 
Urodynamic Studies  SUI — 4, urethral closure pressure with 11` IAP/Bladder Volume and urinary leakage UUI — involuntary detrusor contraction and/or detrusor sphincter dyssynergia 

Incontinence, 4, Quality of Life, UTI's

Celiac Disease: Pathogenesis and clinical findings

Celiac Disease: Pathogenesis and clinical findings 
Associated with other  autoimmune disorders  (i.e. DM1, thyroiditis, RA, SLE, Addison's) 
Genetic predisposition -■ (Northern European, Down's syndrome, Associated with HLA DQ 2,8) 
Note: *The anti-TTG antibody is an IgA anti-body, therefore if the patient is IgA-deficient, absence of anti-TTG does not rule out celiac 
Anti-TTG in serum*  

Anti-TTG reacts with TTG in skin 
Deposition of anti-TTG in renal glomeruli 

Dermatitis herpetiformis 
Chronic Kidney Disease 
Small Bowel Biopsy:  Crypts of bowel become enlarged (hyperplasia) with architectural change, villous shortening 
Legend: Pathophysiology 
Mechanism 
Exposure to prolamins (proteins found in wheat, rye, oats, barley) 
TTG alters prolamin Altered protein fits more easily into HLA 
HLA activates adaptive immunity 
IgA generated against prolamin-TTG 
Wheat prolamin (gliandin) interacts with and activates zonulin signalling 
Gut epithelium becomes more porous 
Large dietary proteins in epithelium disrupt tight junctions 
Author: Matthew Harding Yan Yu Peter Bishay Reviewers: Dean Percy Jason Baserman Usama Malik Kerri Novak* * MD at time of publication 
Inflammation of Intestinal epithelium Inflammation disrupts structure of bowel mucosa Mechanism Unknown Lymphocytes migrate to site of inflammation Extraintestinal Complications: Arthropathy Ataxia (gluten associated) Infertility Mild Hepatitis Villi of intestine atrophy Risk of Microscopic Colitis (50x) Malabsorption Extraintestinal manifestations: Chronic watery Fatigue Failure to thrive, weight loss Anemia (Fe, B12, folate) Peripheral neuropathy (B12, Ca) Ataxia (Ca) Dysrhythmia (Ca, K) Osteoporosis (Vit D, Ca) diarrhea + Intestinal manifestations steatorrhea: Pale, foul-smelling Abdominal bloating Steatorrhea (fat in stool) Diarrhea

Orbital Cellulitis: Pathogenesis and clinical findings

Orbital Cellulitis: Pathogenesis and clinical findings
Authors: Amanda Marchak Reviewers: Jaimie Bird Dr. Rupesh Chawla* * MD at time of publication
Staphylococcus aureus, Streptococcus pyogenes
Note:
Orbital cellulitis is an extremely serious infection. If not caught and treated early, it can lead to death. CT should be performed if suspected.
Involves the orbit
Panopthalmitisb Endopthalmitisc Blindness
 Streptococcus pneumoniae, Moraxella catarrhalis, Haemophilus influenza
Local infection or break in skin
      Eye surgery or trauma
Direct inoculation
Sinusitis (more common)       Periorbital cellulitis1,2
       Hematogenous spread
Contiguous spread of infection
  Pathogens reach orbital tissue (posterior to the orbital septum)
        Spreads to periorbital tissue (anterior to the orbital septum)
Localized inflammation
Conjunctival chemosisa
Eyelid and periorbital edema
Pain on palpation
Induration
Warmth
Orbital Cellulitis Inflammation of orbital tissue       Proptosis
Spreads to surrounding structures
Subperiosteal abscess Brain abscess Cavernous sinus thrombosis Meningitis Subdural empyema Orbital abscess
Notes:
        Impinges on ocular muscles
Impaired extra- ocular movements
Pain with eye
movement or opthalmoplegia
Definitions:
Impinges on nerves
Afferent pupillary defect
Decreased visual acuity
Exposes cornea
Corneal drying and scarring
                         a. Chemosis: Edema of the bulbar conjunctiva
b. Panopthalmitis: inflammation of all coats of the eye including intraocular structures.
c. Endopthalmitis: inflammation of the interior of the eye.
1. See slide on Periorbital Cellulitis for how sinusitis can lead to the development of periorbital cellulitis
2. The micro-organism responsible for periorbital cellulitis varies depending on how the pathogen was introduced to the system.
  Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published November 5, 2018 on www.thecalgaryguide.com

Periorbital Cellulitis: Pathogenesis and Clinical Findings

Periorbital Cellulitis: Pathogenesis and Clinical Findings
Authors: Amanda Marchak Reviewers: Jaimie Bird Dr. Rupesh Chawla* * MD at time of publication
Staphylococcus aureus, Streptococcus pyogenes (most common organisms)
 Note: Also referred to as preseptal cellulitis
      Dacryoadenitisa Conjunctivitisb
Acute chalazionc
Dacryocystitisd Hordeolume
Streptococcus pneumoniae, Moraxella catarrhalis, non-typable Haemophilus influenza (most common organisms)
Abrasion Insect bite
Burns Trauma
             Local infection
Contiguous spread of infection
Sinusitis
Otitis media Hematogenous spread
Local break in skin Micro-organisms enter
Definitions:
              Note:
Eye exam should reveal normal:
- extra-ocular
movements and globe
position
- pupillary reflex and
visual acuity
If any are abnormal, the presentation is no longer considered periorbital cellulitis, as the infection has likely spread beyond the preseptal compartment/orbital septum.
If the eye cannot be assessed, the patient NEEDS a CT scan.
Pathogens reach dermis and subcutaneous periorbital tissue
Periorbital Cellulitis
a. Dacryoadenitis: infection of the lacrimal glands
b. Conjunctivitis: inflammation of the conjunctiva
c. Chalazion: a benign, painless bump or nodule inside the upper or lower eyelid which results from healed internal hordeolums that are no longer infectious.
d. Dacryocystitis: an infection of the lacrimal sac, secondary to obstruction of the nasolacrimal duct at the junction of lacrimal sac.
e. Hordeolum: localized infection or inflammation of the eyelid margin involving hair follicles of the eyelashes or meibomian glands.
   Spreads beyond preseptal compartment/orbital septum
Involves the orbit Orbital cellulitis
See slide on Orbital Cellulitis: Pathogenesis and clinical findings
Localized inflammation
Pain on palpation
Induration
Warmth
Eyelid and periorbital edema
           Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published November 5, 2018 on www.thecalgaryguide.com

Mastoiditis: Pathogenesis and clinical findings

Mastoiditis: Pathogenesis and clinical findings
Authors:
Amanda Marchak
Reviewers:
Nicola Adderley Jim Rogers Emily Ryznar Danielle Nelson* * MD at time of publication
 Acute Otitis Media (AOM)
Distal middle ear is physically connected to mastoid air spaces
Pathogens spread from middle ear to the mastoid air spaces
Mucosa lining the mastoid becomes inflamed
Mastoiditis 1
Infection persists
Accumulation of pus in mastoid cavities
↑ pressure Formation of abscess cavities
Dissection of pus into adjacent areas
Infection spreads
Into intracranial compartment
See slide on Acute Otitis Media (AOM): Pathogenesis and Clinical Findings in Children
         Post- operation
Trauma Infection
Notes:
1. Most common suppurative complication of AOM
Tenderness, erythema, swelling and fluctuance over the mastoid process
Inflammation spreads to external auditory canal
Cranial Nerve VII anatomically near mastoid
Cranial Nerve VIII anatomically near mastoid air space
Destroys bony septae b/t air cells (visible on CT)
Mastoid abscess
Swelling of external auditory canal
Mastoid inflammation disrupts nerve
Mastoid inflammation disrupts nerve
Petrositis
Facial nerve palsy
Sensorineural hearing loss Labyrinthitis
                                         Osteomyelitis of the calvaria
 Into adjacent bones
Underneath the periosteum     Subperiosteal abscess   Pinna is pushed out and
      of the temporal bone
Into the neck beneath the attachment of the sternocleidomastoid and digastric muscles
forward
       Dural venous thrombosis Temporal lobe abscess Meningitis
Epidural abscess Subdural abscess Cerebellar abscess
Bezold abscess
      Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published November 5, 2018 on www.thecalgaryguide.com

Sinusitis: Pathogenesis and clinical findings

Sinusitis: Pathogenesis and clinical findings
Authors: Amanda Marchak Reviewers: Nicola Adderley Jim Rogers Danielle Nelson* * MD at time of publication
Abbreviations
URTI – Upper respiratory tract infection
Nasal obstruction/ congestion
Hyposmia
Headache
Facial pain/pressure
Maxillary tooth pain
Ear pain/ fullness
Osteomyelitis of frontal bone
          Chemical irritants
Cystic Fibrosis
Direct toxic effect on cilia
Viral URTI Allergies
Inflammation of paranasal sinuses
Edematous passageways
Septal deviation Adenoid hypertrophy Polyps
Turbinate hypertrophy Tumors Foreign body
      Dysfunctional cilia
Congenital and/or craniofacial abnormality Obstruct sinus ostia
       Cilia unable to clear mucus from sinuses
     Mucus unable to drain through ostia
   Post-nasal drip       Mucus overflows from the sinuses Cough
Mucus accumulates in sinuses
Occupies a larger volume
Applies ↑ pressure to sinus walls
Mucopurulent discharge
Bacterial1 overgrowth in sinuses Bacterial infection spreads to adjacent structures
          Halitosis Pharyngitis Throat clearing
Dental root infection
Immunodeficiency
Note:
Irritates the back of the throat
              Perforation of the Schneiderian membrane2
Passage of bacteria into the sinuses
Fever
Fatigue
Subperiosteal orbital abscess
Orbital abscess Orbital edema
            ↑ susceptibility to bacteria
     1. The most common bacteria are Streptococcus pneumoniae, Haemophilus influenza, and Moraxella catarrhalis. Staphylococcus aureus and Group A Streptococcus may be seen, but are less common. However, in cases of dental root infection, oral anaerobes become more common, while Pseudomonas species are associated with foreign bodies.
2. The Schneiderian membrane is the membranous lining of the maxillary cavity.
Cavernous sinus thrombosis
Meningitis Cerebral abscess
Subdural abscess Epidural abscess
Periorbital or orbital cellulitis
             Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published November 5, 2018 on www.thecalgaryguide.com

Boutonniere Deformity: Pathogenesis and Complications

Boutonniere Deformity: Pathogenesis and Complications Laceration of extensor surface of PIP
Authors: William F Hill Marshall Thibedeau Reviewers: Emily Ryznar Brett Byers* *MD at time of publication
Rheumatoid Arthritis
            Hand trauma
Hyperflexion of PIP
Dislocation of PIP
Central Slip Rupture
Unopposed lumbrical and interossei pull on lateral bands
Stretching of triangular ligament
Volar subluxation of lateral bands
Lateral band contraction
Hyperextension of DIP
Erosion of connective tissue
Synovial inflammation
   Avulsion of central slip insertion
          Triangular ligament prevent bowstringing of the lateral bands during finger flexion
EDC inserts dorsally on to extensor aponeurosis
Interossei and lumbricals travel from volar aspect of palm with action dorsal to the axis of rotation
Aponeurosis gives rise to central slip and lateral bands
Elson test for central slip rupture:
• Flexion of PIP 90 degrees over edge of table • Extend affected phalanx against resistance • Positive if DIP becomes extended and rigid
• From exaggerated lateral band action
               Abbreviations:
PIP proximal interphalangeal joint EDC: extensor digitorum communis IPJ: interphalangeal joint
DIP: distal interphalangeal joint ROM: range of motion
Flexion of PIP
  Boutonniere Deformity
Shortening of collateral ligaments and volar plate of PIP joint
Premanent contracture and fibrosis of IPJs
        Permanent deformity
↓ ROM in PIP extension
Osteoarthritis
 Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
  Complications
Published December 4, 2018 on www.thecalgaryguide.com

Complex Regional Pain Syndrome: Pathogenesis and clinical findings

Complex Regional Pain Syndrome: Pathogenesis and clinical findings
Authors: Calvin Howard Reviewers: Sina Marzoughi Scott Jarvis* * MD at time of publication
 Central nervous system lesion Unknown Mechanism
ACE inhibitors
Trauma with at most minor nerve lesion
i.e. surgery, nerve compression, fracture, tissue trauma, ischemia, sprain
           Neurogenic inflammation
Sympathetic dysregulation
Acute Phase
Proinflammatory cytokine profile
Central and peripheral nociceptive sensitization
Chronic Phase
Central sensorimotor dysregulation
↓ Perfusion of cortex & ↓ cortical grey matter volume of limbic and sensorimotor areas
↑ Perfusion to motor cortex
↓ Grey matter volume in cortical pain regions
↓ Connectivity of sensorimotor planning/control regions
             Complex Regional Pain Syndrome 1
(Formerly Reflex Sympathetic Dystrophy)
   Sensory: Hyperalgesia or allodynia
Sudomotor: Edema, sweating changes, asymmetric sweating Vasomotor: Temperature asymmetry, altered skin colour
Motor/trophic: Decreased range of motion, motor dysfunction, trophic changes
Definitions:
• Hyperalgesia: Heightened sensation of pain
• Allodynia: Pain caused by normally non-painful stimuli • Sudomotor: Relating to the sweat glands
• Vasomotor: Relating to the muscle within vasculature • Trophic: Relating to growth and atrophy of cells
• Cytokine: Molecules that recruit inflammatory cells
     Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published September 20, 2018 on www.thecalgaryguide.com

Venous insufficiency- Signs and symptoms

Venous insufficiency- Signs and symptoms vein veins blood flow interruption venous system valve incompetence reflux venous obstruction venous return backflow blood hypertension hydrostatic transmural pressure in postcapillary vessels capillary dilatation vessel permeability extravasation RBCs interstitial hemoglobin released degraded hemosiderin deposits hemosiderosis brown discolouration high pressure superficial venous circulation varicose veins tortuous dilated superficial veins distended veins compress somatic nerves achy pain protein-rich exudate edema peripheral ankle edema tissue perfusion metabolic exchange stasis dermatitis dermal fibrosis lipodermatosclerosis fibrotic thickened skin pruritus chronic inflammation risk of thrombus local ischemia embolus skin integrity venous ulcer risk of infection Adderley gagnon Waechter

Reactive Neutrophilia- Pathogenesis and Clinical Findings

reactive neutrophilic pathogenesis clinical findings infection inflammation malignancy drugs emotional stimuli stress smoking hyposplenism asplenia rheumatoid arthritis Crohn's epinephrine retinoic acid glucocorticoids anxiety exercise heat stroke surgery neutrophils neutrophil bone marrow demarginalization splenic sequestration neutrophilic ANC peripheral blood smear absolute neutrophil count left shift bands metamyelocytes myelocytes toxic granulations dohle bodies brenneis Siddique savoie ryznar

Primary Myelofibrosis pathogenesis and clinical findings

Primary Myelofibrosis: Pathogenesis and clinical findings
Legend: Published March 30, 2019 on www.Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications thecalgaryguide.com
Author:
Tony Gu
Reviewers:
Naman Siddique
Sonia Cerquozzi*
Man-Chiu Poon*
* MD at time of publication
Definitions:
Constitutive activation – Constant
expression of gene
Extramedullary hematopoeisis – red
blood cell production outside of bone
marrow
Somatic mutations in
genes that drive
cancerous replication
(e.g., JAK2, CALR, MPL)
within hematopoietic
stem cells
Non-driver mutations in
other myeloid genes
(e.g., LNK, CBL, TET2,
ASXL1, IDH)
Constitutive activation of
cellular proliferation
pathways
↑ cell signaling
↑ gene transcription and
expression
Cellular proliferation and
resistance to apoptosis
Proliferation of abnormal
megakaryocytes
↑ neutrophil engulfment
by megakaryocytes
↑ growth factor release
by megakaryocytes
Stimulation of
fibroblasts
Stimulation of
endothelial cells
New blood vessel formation
↑ osteoprotegerin Unbalanced osteoblast
proliferation Osteosclerosis
Fibrosis of the
bone marrow
Anemia
Bleeding and
bruising
Infections
Fatigue and pallor
Bone pain
Increased cell
turnover
Tumor lysis
syndrome
Cachexia, night sweats,
fever/chills, malaise
Expanding
marrow pushing
against bone
Extramedullary
hematopoiesis Hepatomegaly
Portal
hypertension
Splenomegaly
↑ LDH
Thrombocytosis
Leukocytosis
Secretion of
coagulation
inducing cytokines
Arterial and
venous
thromboembolism
↓ blood cell
production
&
leukoerythroblastosis
Thrombocytopenia
Leukopenia
↑ K+, PO4
2-, uric acid
↓ Ca2+
Bone pain
Periostitis
Immature granulocyte and
erythroid precursors with blasts
↑ cytokine
production
(+)
↑ sequestration of blood cells
Disseminated
intervascular
coagulation
(see MAHA slide)
Definitions:
Periostitis – Inflammation of the
membrane surrounding bone
Osteosclerosis – Abnormal hardening
and increased in density of bone

Hemorrhoids - Pathogenesis and Clinical Findings

INTERNAL Hemorrhoids
- Found proximal to the dentate line
- Visceral innervation
Behavioural or Genetic Predisposition
I.e. hereditary bowel/rectal problems or
shared habits and practices (unclear mechanism)
Increased Intra-Abdominal Pressure
I.e. pregnancy, constipation, chronic straining,
lifting, cirrhosis
Hemorrhoids: Pathogenesis and clinical findings
Dilations originate from inferior
hemorrhoidal venous plexus
Vascular cushions engorge
along anal canal
Legend: Published March 30, 2019 on www.Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications thecalgaryguide.com
Authors:
Aleeza Manucot
Reviewers:
Yoyo Chan
Sean Doherty
Dr. Sylvain Coderre*
* MD at time of publication
Supporting tissues of anal cushions weaken,
disintegrate, or deteriorate
Inflammatory reaction
occurs, involving vascular
wall and connective tissue
Thrombosis
Pain
↑ mucus secretions or fecal
soiling of prolapsing
hemorrhoids
Cushion epithelium erodes via
damage from compression
Painless
rectal
bleeding
Bleeding without prolapse
Prolapse with spontaneous
reduction
Prolapse requiring manual
reduction
Irreducible
1st degree
2nd degree
3rd degree
4th degree
Infarction and thrombosis
Acute severe pain
Anal cushions prolapse (downwardly slide)
into rectum or open space
Dentate line: divides
the upper two thirds
and lower third
of the anal canal
EXTERNAL Hemorrhoids
- Found distal to the dentate line
- Somatic innervation
Somatic nerve
receptors activated
Sebaceous glands
↑ secretions around
area of hemorrhoid
Itching Perianal
irritation
Swelling
Inflammation creates
prothrombotic state
Hemorrhoids

gastroesophageal-reflux-disease-gerd-complications

Gastroesophageal Reflux Disease (GERD): Complications
Esophageal stricture
disease
Esophagitis
Esophageal
adenocarcinoma
Barrett’s esophagus
GERD
Reflux of gastric content into distal esophagus
Damage to squamous
esophageal epithelium
Legend: Published March 30, 2019 on www.Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications thecalgaryguide.com
Authors:
Wendy Wang
Reviewers:
Yoyo Chan
Sean Doherty
Dr. Sylvain Coderre*
* MD at time of publication
Squamous esophageal
epithelium undergoes
metaplasia to become
columnar epithelium
This predisposes cells to
premalignant changes
(dysplasia)
Collagen is deposited
where ulcers heal
Asthma/Chronic Cough
Chronic Laryngitis
Laryngeal and
Tracheal Stenosis
Extra-esophageal Complications Esophageal Complications
Airway becomes
irritated
Fibroblasts proliferate
and deposit granulation
tissue in airway
Tissue deposition
leads to narrowing of
laryngeal and
tracheal space
Damage to pharyngeal
lining and airway
Esophageal tissue repeatedly
exposed to stomach acid
Pro-inflammatory cells and cytokines
are recruited to the area
Definitions:
• Metaplasia: abnormal change in the
nature of a tissue
• Pro-inflammatory cells and cytokines:
Mediators of inflammation. Examples
of cells include macrophages and T
cells, cytokines include IL-17, IL-2, IL-4
Over time, collagen fibers
contract
Bronchoconstriction
↑ vagal
tone
↑ bronchial
reactivity
Cough sensory
nerve endings are
stimulated
Vagal reflex
is activated
Activation of
cough center in
brainstem
↑ inflammation of
squamous epithelium
Ulcers form in esophagus

Polyarteritis Nodosa (PAN): Pathogenesis and Clinical Findings

Polyarteritis Nodosa (PAN): Pathogenesis and clinical findings
   Environmental triggers
Infectious/viral agents (commonly Hepatitis B)
Medical Comorbidities Malignancies (most commonly hairy-cell leukemia)
Immunogenetic Predisposition: patient is genetically predisposed to a dysregulated immune response
Fever
↑ ESR and CRP
        Postulate 1
Viral antigen-antibody complexes deposit in vasculature, causing lesions and activating cellular inflammatory response
Authors: Nela Cosic, Yan Yu* Reviewers: Sean Doherty Martin Atkinson*
* MD at time of publication
Palpable or necrotic purpura
Malignant Hypertension
Renal Insufficiency
Myocardial ischemia
Heart failure Diffuse myalgias
Postulate 2
Viral replication causes direct injury to vascular endothelial cells
↑ Anti- endothelial cell autoantibodies (AECA)
Altered cytokine profile (↑TNF-α, IL-1β, IFN-α, IL- 2)à↑ T-cell mediated immune response
Weight metabolism Loss
Autoimmune attack on various areas of the body
Malaise and/or Arthralgias (knees, ankles, elbows, wrists)
Orchitis: Testicular pain, erythema and/or swelling
Small intestine perforation GI Manifestations
Non-specific abdo pain
GI hemorrhage
Peripheral sensory changes: Distal mononeuropathy
multiplex
    Polyarteritis Nodosa (PAN)
Focal segmental necrotizing leukocytoclastic vasculitis of medium or small-sized arteries
Inflammation of arteries damages the vascular endothelium of those arteries
Inflammation predisposes formation of arterial thromboses
Blockage of arteriesà tissue ischemia and possible necrosis (tissue cell death)
↑ basal
         Arterial aneurysms
Inflamed subcutaneous arteries
Inflamed renal artery
àluminal narrowing and reduced blood flow to kidneys
Inflamed coronary artery à luminal narrowing, occlusion, thromboses
Segmental inflammation of muscular arteries, stimulating surrounding nociceptors. Muscle ischemia develops long-term.
Ischemia/necrosis of the testicles
Ischemia/necrosis of the small intestine
                                    Ischemic vasculitic nerve damage: Immune complex deposition within vessel walls of arteries traveling with nerves leads to persistent vascular inflammation and ischemia of associated nerve
     Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published April 18, 2019 on www.thecalgaryguide.com

dermatomyositis-dm-and-polymyositis-pm-pathogenesis-and-clinical-findings

Dermatomyositis (DM) and Polymyositis (PM):
Authors: Merna Adly, Yan Yu* Reviewers: Nela Cosic Sean Doherty Martin Atkinson* * MD at time of publication
Pathogenesis and clinical findings
   Immunogenetic and Cellular Predisposition
Genetic polymorphisms cause dysregulated immune response, cytokine profile, and protein expression in muscle cells
Demographics
F:M, 2:1
Bimodal age distribution:
- Juvenile: 7 years of age (mean) - Adult: 52 years of age (mean)
Malignancy
Tumor cells increase systemic inflammatory response, leading to increase of autoantigens associated with DM
↑ in DM-associated autoantibodies
(Ex. Anti-Mi-2/ Anti-Jo-1) Autoantibodies bind to DNA or RNA in muscles,
provoking a systemic inflammatory response
↑ chemokine and cytokine release in endothelial vasculature of muscles
Perivascular Capillary necrosis inflammation
Lack of blood supply to the myofibers causes endofascicular hypoperfusion and muscle ischemia
Muscle tissue damage:
Inflammatory infiltrates destroy cellular components of muscle (endoplasmic reticular, myofiber, and keratinocytes)
Environmental triggers
Infectious agents (ex. Picornavirus) or drugs (ex. statins) provoke immune response
Elevated Antinuclear Antibodies
Anti-Jo-1 Anti-OJ Anti-Mi2 Anti-SRP Anti-EJ Anti-PL12 Anti-PL7
These processes occur in the skin on the dorsum of the hands, forming hyperkeratotic flat red papules
These processes occur in the upper & lower eyelids, causing red-purple discoloration +/- swelling
Perifascicular atrophy
(Observed on histology)
Weaker GI tract musculature Weaker pulmonary musculature Weaker cardiac musculature
           Dermatomyositis only:
Gottron Papules Heliotrope Rash
                                  Damaged muscle cells release their internal cellular enzymes into the bloodstream
Elevation of muscle enzyme levels in serum:
Creatinine kinase (CK), lactate dehydrogenase (LD), aldolase, aspartate aminotransferase (AST), and alanine aminotransferase (ALT)
Muscle Biopsy Findings Muscle necrosis, fiber regeneration, diffuse CD8+ T lymphocytes infiltrates
Bilateral Muscle Weakness Subacute development, primarily deltoids and hip flexors affected
Dysphagia
Aspiration, respiratory compromise
Atrioventricular defects, tachyarrhythmias, dilated cardiomyopathy
     Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published April 18, 2019 on www.thecalgaryguide.com

acute-somatic-pain

Acute Somatic Pain:
Pathophysiology
Acute tissue damage, from three types of causes:
Mechanical (eg: sharp pin) Thermal (eg: hot stove) Chemical (eg: inflammation)
Nociceptors activated at site of injury (1st order sensory neurons)
Nociceptive fibres (A∂ and C) carry noxious sensory information to the ipsilateral dorsal horn of the spinal cord
Excitatory neurotransmitters are released and stimulate 2nd order sensory neurons
2nd order sensory neurons immediately cross the midline of the spinal cord, and ascend up the opposite side’s anterolateral (aka spinothalamic) tracts, terminating in various locations:
Authors: Lisa Murphy Yan Yu* Reviewers: Mackenzie Gault Melinda Davis* * MD at time of publication
Nociceptors: neurons that detect noxious or painful stimuli and carry this information to the spinal cord. There are two major types:
A∂ fibres: myelinated, initial “sharp, fast” feeling
C fibres: unmyelinated, delayed, “dull, burning” feeling
To hypothalamus
2nd order neuron synapses in the hypothalamus
Hypothalamic neurons coordinate the body’s visceral response to pain
                  2
nd
To thalamus
order neuron terminates in thalamus
To brainstem
2nd order neuron synapses in brainstem’s reticular formation
To midbrain
2nd order neuron synapses in periaqueductal gray area (PGA) in the midbrain
  In the thalamus, 2nd order sensory neurons synapse with 3rd order sensory neurons, which carry the signal to the cerebral cortex
Stimulates descending pathways to modulate the incoming pain signal
Decreased or increased perception of pain
         Pain localization and sensation
Emotional and behavioural response
↑ Heart Rate
Nausea
    Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published April 25, 2019 on www.thecalgaryguide.com

Crohn's Disease

Inflammatory Bowel Disease: Clinical findings in Crohn’s Disease
Authors: Yan Yu Amy Maghera Reviewers: Jennifer Au Danny Guo Jason Baserman Jessica Tjong Kerri Novak* * MD at time of publication
   Behavioural Factors:
Smoking, over-sanitation
Genetic Susceptibility
Environmental Factors
Diet, bacteria/viruses, drugs, vitamin D
    Systemic immune response primarily against the GI tract.
(Unclear mechanism, mediated by cytokine release and neutrophil inflammation)
  Inflammation of the GI tract lining
- Inflammation is “transmural”, spanning the entire thickness of the intestinal wall from luminal mucosa to the serosa.
- The inflammation occurs anywhere in the GI tract from the oral mucosa to the anal mucosa (from ‘gums to bum’) in skip lesion pattern.
       Atrophy, scarring of the intestinal villi
Inflammatory cytokines destroy the mucosa epithelial cells of the GI tract wall, causing cell apoptosis and ulceration
↑ permeability of the blood vessels supplying the GI tract wall
Chronic inflammation impairs healing responses
Dysregulated wound healingàexcess
extracellular matrix deposition
Fibrosis leads to scar tissue and thickening of all layers of the GI tract
Strictures
Inflammation is systemic, affecting:
Joints         Arthropathy Erythema
            Impaired absorption of nutrients
Weight loss
Prolonged GI bleeding
Anemia
Transporter proteins responsible for Na+ reabsorption gradually disappear from the epithelium
More sodium (and thus water) is
retained in the GI tract lumen
Microperforations can penetrate through the intestinal wall
Anal fistulae (“holes” connecting the anus to the skin, bladder, peritoneum, small bowel, etc.)
Continued inflammation and/or infection can lead to:
Leakage of fluid out of capillaries into the GI tract
Luminal edema and swelling
Narrowing of GI lumenàbowel obstruction
Skin
Mouth Eyes
Liver
nodosum, pyoderma gangreno- sum
>5 canker sores
Uveitis
Iritis, scleritis
Sclerosing cholangitis
                       ↓ fat absorption
Fatty acids (negatively charged) bind Ca2+, freeing oxalate from Ca2+
↑ oxalate absorbed into blood & filtered by kidney
Calcium oxalate kidney stones
Diarrhea
Abdominal cramping and pain
(see Bowel Obstruction page for full mechanism
                                         Anal abscesses Inflammatory masses
   Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Re-Published June 15, 2019 on www.thecalgaryguide.com

Ulcerative Colitis

Inflammatory Bowel Disease: Clinical Findings in Ulcerative Colitis
Authors: Yan Yu Amy Maghera Reviewers: Jennifer Au Danny Guo Jason Baserman Crystal Liu Danielle Chang Kerri Novak* * MD at time of initial publication
Inflammation is systemic, affecting:
   Environmental Factors
Diet, bacteria/viruses, drugs
Genetic Susceptibility
Behavioral Factors:
In UC, smoking and appendectomy are actually protective (unknown reason)
     Immune response against the GI tract. (Unclear mechanism, but thought to be mediated by cytokine release and neutrophil infiltration)
  Inflammation of the GI tract epithelial lining
- Starting at the rectum and moves up the colon and is continuous (does not invade the small intestine)
- Inflammation affects the mucosal and submucosal only
        Diarrhea, abdo pain and cramping causing avoidance of food
Weight loss
Apoptosis of GI tract mucosa
Transporter proteins responsible for Na+ reabsorption gradually disappear from the epithelium
Ulceration, into the anus, and more severe
Prolonged Bleeding - GI and anus
Anemia, often iron deficiency
Inflammation ↑ permeability of the blood vessels supplying the GI tract wall
Fluid leak out of capillaries into GI tract wall, causes edema and swelling
Swelling narrows the GI tract lumen, causing bowel obstruction
Inflammation, ulceration, or infection at the anus (all involve the RECTUM!)
Anal irritation stimulates autonomic and somatic nerves leading up to the brain, causing the pt to want to defecate
Tenesmus, urgency, frequency (feeling or urgency to defecate, but little stool is produced)
Joints Skin
Arthroplasty/ joint pain
Erythema nodosum, pyoderma gangrenosum
                     Mouth       >5 canker sores
          More sodium (and thus water) is retained in the GI tract lumen
Bloody Diarrhea, usually bloody due to anal bleeding and ulceration bleeding
Abdominal Cramping and pain (see Bowel Obstruction page for full mechanism)
Eyes (uvea, iris, sclera)
Liver Blood
Uveitis
Iritis, scleritis
Sclerosing Cholangitis
Autoimmune hemolytic anemia
                Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Re-Published May 5, 2019 on www.thecalgaryguide.com

perforated-viscous

Perforated “Viscous” (aka. GI tract; bowels):
Author:
Yan Yu
Reviewers:
Michael Blomfield, Tony Gu, Dean Percy, Danny Guo Maitreyi Ramran* * MD at initial time of publication
Chest X-Ray (CXR)
Pathogenesis and Clinical Findings
Diverticulitis
   Crohn’s disease Peptic ulcer (H. pylori
infection, NSAID use, ICU stress, etc)
Appendicitis
Malignant neoplasm
Irritates visceral peritoneum, stimulates autonomic nerves
            Severe inflammation causes destruction of GI tract mucosa
Over time, Perforation of the GI tract wall
Bowel contents (air, fluids) released into peritoneal cavity
Massive peritoneal inflammation
Diagnostic investigations if a GI perforation is suspected
Dull diffuse abdominal pain
          Severe, Sharp abdominal pain with peritoneal signs
Abdominal X-ray
  Irritation of parietal peritoneum, stimulates somatic nerves
      • Abdominal X-ray
• Intra-peritoneal air will coat the GI tract surfaces, giving them a faint white outline
under X-ray
• Chest X-ray of upright patient (Diagnostic)
• Intra-peritoneal air will rise above the peritoneal fluid when pt is upright, accumulating under the right hemi-diaphragm.
• Note: air under left hemi-diaphragm = normal gastric bubble
• CT? Most patients with suspected GI perforation will get a CT scan, but this is not the diagnostic gold standard (and access to CT can be limited, especially in rural settings)
 Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Re-Published June 30, 2019 on www.thecalgaryguide.com

acanthosis-nigricans-pathogenesis-and-clinical-findings

Acanthosis Nigricans: Pathogenesis and clinical findings
Authors: Laura Chin Reviewers: Taylor Woo Crystal Liu Laurie Parsons* * MD at time of publication
     Medications
E.g. systemic glucocorticoids, injected insulin, oral contraceptives
Hyperinsulinemia
Insulin inhibits IGFBP 1&2 secretion
Genetic Syndromes
E.g. Down syndrome, Rabson- Mendenhall syndrome, congenital generalized lipodystrophy
Defects in insulin receptor or anti-insulin receptor antibody production
Insulin Resistance
↑ IGFR1 binding
Type 2 Diabetes Mellitus
Polycystic Ovarian Syndrome
Obesity
Neoplasms
E.g. gastric carcinomas
Excess IGF-1, TGFα, and FGF production
TGFα is structurally similar to EGFα
TGFα can bind to EGFR
↑ EGFR binding
Inheritable Mutations
E.g. FGFR3 activating mutation
                           ↑ free IGF-1
↑ FGFR binding
     Moist environment and rubbing of intertriginous skin
Skin inflammation and thinning
Bacterial or yeast infection
Erosions Malodour
Dermal keratinocyte and fibroblast growth and differentiation of neck and intertriginous areas, occasionally mucosal surfaces
Hyperkeratosis
Abbreviations:
• IGF-1 – insulin-like growth factor • IGFR1 – insulin-like growth
factor receptor-1
• IGFBPs – insulin-like growth
factor binding proteins
• FGFR – fibroblast growth factor
receptor
• TGFα – transforming growth
factor-alpha
• EGFα – epidermal growth
factor-alpha
• EGFR – epidermal growth factor
receptor
         Hyperpigmentation
Crusting
Velvety plaques
       Pain
Pruritis
Pus
Acanthosis Nigricans
    Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published July 14, 2019 on www.thecalgaryguide.com

intraventricular-hemorrhage-in-preterm-infants-clinical-findings-and-complications

Intraventricular hemorrhage (IVH) in preterm infants:
Clinical findings and complications
Authors: Alexa Scarcello Reviewers: Nicola Adderley, Emily Ryznar, Yan Yu*, Jennifer Unrau* * MD at time of publication
Volpe Grading Grade I: germinal matrix
hemorrhage with no or minimal IVH (<10% of ventricular area)
Grade II: IVH (10-50% of ventricle) Grade III: IVH (>50% of ventricle;
usually distends lateral ventricle)
Grade IV/Intra-parenchymal echodensity (IPE): periventricular hemorrhagic infarction
Inflammation/dysfunction of arachnoid villi
↓ absorption of CSF 2° to obstruction of arachnoid villi
Communicating hydrocephalus (IVH grades II-IV)
Venous congestion
Venous infarction
Periventricular hemorrhagic necrosis
Destruction of periventricular motor tracts
Cerebral palsy
Rapid significant blood loss
↓ intravascular blood volume
Hypotension
↓ bloodflow to the brain to support brain function
Intraventricular Hemorrhage (IVH)
hemorrhage in periventricular subependymal germinal matrix
Ultrasound: blood in germinal matrix, ventricles, or cerebral parenchyma
Sudden ↓ hematocrit
Blood irritates contiguous structures
Variable neurologic findings; including altered level of consciousness, hypotonia, apnea, etc
                                      Neuro- developmental abnormalities (varying severity)
See slide - Hydrocephalus: Clinical Findings in Pediatrics
This mechanism leads to three different possible clinical manifestations:
1. Silent Presentation (most common)
2. Stuttering/Saltatory Course: non-specific findings - hypotonia, apnea, altered level of consciousness, bradycardia, and ↓ Spontaneous movements
3. Catastrophic Deterioration (least common) Stupor or coma, decerebrate posturing, seizures, bradycardia, metabolic acidosis, bulging fontanelles, abnormal pupillary reflexes, inappropriate ADH secretion
    Notes
 • Incidence & severity are inversely proportional to gestational age
• 50% occur within 1st day of life, 90% by 3rd day
• As explained in the flow chart, the postnatal clinical presentations of
IVH fall into three categories (1-3)
• Symptoms of catastrophic bleeds are uncommon and usually caused
by rapid significant blood loss with subsequent neurologic findings 2° to meningeal irritation, inflammation, and potential mass effect/acute hydrocephalus; severe bleeds may also occur in the absence of clinical findings attributable to IVH
  Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published July 27, 2019 on www.thecalgaryguide.com

Appendicitis

Appendicitis: Pathogenesis and Clinical Findings
Authors: Yan Yu Wayne Rosen* Reviewers: Wendy Yao Laura Craig Noriyah AlAwadhi* * MD at time of publication
Dull, crampy, diffuse peri- umbilical pain
Pt may develop fever, diarrhea, constipation, vomiting or anorexia as inflammation worsens
Focal, intense, persistent RLQ
pain, abdominal guarding and peritoneal signs (i.e. percussion and rebound tenderness
  Epidemiology
Dx of healthy adults:
• Men > women
• Commonly 10-30 years old,
can present at any age
• Most common cause of acute abdomen (5% prevalence in all ethnicities)
The appendix is anatomically located in the RLQ; appendicitis may be confused with disorders of surrounding structures: Gynecological Diseases
• RuleoutpregnancywithHCG pregnancy test
• Rupturedovariancyst
• Ectopicpregnancy
• Mittelschmerz(mid-cycle
pain)
Gastro-intestinal Diseases
• Meckel’sdiverticulum (presents identically to appendicitis; surgically located 2 feet from ileocecal valve; mostly seen in children)
• Diverticulitis(presentsasleft sided appendicitis)
Non-GI Abdominal Issues
• Mesentericadenitisinkids <15: swollen mesenteric lymph nodes
• Renalcolic
Obstruction of appendiceal lumen (by fecalith, fibrosis, neoplasia, foreign bodies or lymph nodes in kids)
Appendix distension and spasms
↑ lumen pressure, ↓ blood flow to appendix
Ischemia, tissue necrosis, loss of appendix structural integrity
Bacterial invasion of the appendix wall, causing transmural inflammationandnecrosis
Stretching of visceral peritoneum, stimulation of autonomic nerves T9-T10
Progression of inflammation over several days (variable length of time)
Irritation of parietal peritoneum, stimulation of somaticnerves
                              If appendix not surgically removed
Perforation of colon wall, causing peritonitis, abscesses or death
Note: Symptoms hugely variable. Only 30% present with classic history. Diagnosis is mostly clinical. Further investigations:
CBC: Leukocytosis (due to inflammatory response) CT: Gold standard test. Thickened visceral membrane with enhancing (white) rim due to ↑ blood flow
      Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Re-Published July 27, 2019 on www.thecalgaryguide.com

Acute GI Related Abdominal Pain

Acute GI-Related Abdominal Pain: Pathogenesis and Characteristics
Authors: Yan Yu Wayne Rosen* Reviewers: Laura Craig Danny Guo Julia Heighton Maitreyi Raman* * MD at time of publication
   Peritoneal cavity
Visceral peritoneum
(innervated by autonomic nerves)
Bowel stretching, pulling, contracting
Abdominal pain type:
Diffuse, non-localized Dull, crampy, periodic Not associated with movement
Patient may writhe around, trying to get rid of the pain
Mesentery Intestinal lumen
Parietal peritoneum
(innervated by somatic nerves)
          Cross-section of the GI tract
Cuts, structural damage, and inflammation in the bowel
       Important Notes
• Acute abdominal pain can also result from non-
gastrointestinal causes, such as kidney stones, female reproductive tract issues, and urinary tract issues. For simplicity’s sake, only the GI-related acute abdominal pain disorders are listed here.
• The DDx of visceral abdominal pain is broad. Please consult relevant sections of the Calgary Black Book for the DDx.
• Keep in mind that visceral abdominal pain can also be caused by the “acute abdomen” diseases (if the diseases are presenting in their initial phases).
• • •
• • •
Abdominal pain type:
Sharp, well-localized
Excruciatingly painful, persistent Associated with movement of bowels
Patient often lies still to avoid abdominal vibration
Peritoneal signs
Abdominal guarding, pain with abdominal vibration (coughing, shaking, percussion, palpation)
     Transition from diffuse to localized pain can indicate disease progression (e.g. from visceral to parietal peritoneal inflammation)
Note: bowel obstruction may or may not present as acute abdominal pain
Bowel Infarction
       Appendicitis Diverticulitis
Acute Cholecystitis
Acute Pancreatitis
Perforated Ulcer
 DDx of an “acute abdomen”:
 A sudden, non-traumatic disorder of the abdomen that needs urgent diagnosis and treatment. Each topic will be further explored in their respective slides.
 Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Re-Published July 27, 2019 on www.thecalgaryguide.com

Erythema Nodosum pathogenesis and clinical findings

Erythema Nodosum: Pathogenesis and clinical findings
Authors: Merna Adly Reviewers: Taylor Evart Woo Crystal Liu Yan Yu* Laurie Parsons* * MD at time of publication
Epidermal layer Dermal-Epidermal Junction
Dermal layer
Subcutaneous Fat Layer
Phase 1-5. Septal Fibrosis made of inflammatory cells, such as T lymphocytes, histocytes and eosinophils
     Genetic Dysregulation
Infections (Ex.
Streptococcal
Pharyngitis) ~28-48% of cases
Medications (Ex. Birth Control Pills, Sulfa drugs) ~3-10% of cases
Malignancy (ex. Lymphoma)
Autoimmune conditions (ex. Sarcoidosis and
Inflammatory Bowel Disease) ~11-25% of cases
Pregnancy ~1-3% of cases
    Antigenic Stimuli / Bacteria / Viruses / Chemical Agents all could trigger the following process: Phase 1. Neutrophils Infiltrate the fibrous septa between fat lobules in the subcutaneous fat
Phase 2. Neutrophils release reactive oxygen species, leading to oxidative tissue damage and inflammation
Phase 3. Opening of inter-endothelial junction and the migration of more inflammatory cells into the septal venules, including macrophages, histocytes, and eosinophils
Phase 4. Macrophages secrete inflammatory cytokines, which stimulates the proliferation of more helper T cells (Th1)
Phase 5. Th1 cells secrete more cytokines, leading to the further release of Th1 cytokines and mediating the immune complexes deposition in the septal venules of the subcutaneous fat (panniculitis). The Th1 immune reaction is called Type IV Delayed Hypersensitivity Reaction
Phase 6. Activated macrophages produce hydrolytic enzymes and transform into multi- nucleated giant cells, called Miescher’s Radial Granulomas. These consist of small, well defined aggregations of small histocytes arranged radially around a small cleft of variable shapes in the septal venules of the subcutaneous fat
Phase 1-4. Lesions are red tender nodules, poorly defined, vary in size from 2-6 cm, and usually on shins ( 1st week)
Fat Lobules T lymphocytes
Macrophages
                                                       Note: we’ve done extensive research and can’t figure out why erythema nodosum happens mostly on the shins. If you have an answer, please email us!
      Phase 5. Lesions become tense, hard, and painful; and they change in color into bluish or livid. (2nd week)
Phase 6. Lesions become fluctuant as in abscess, but do not ulcerate. Lesions fade to a yellowish color
Epidermal layer Dermal-Epidermal Junction
Dermal layer Subcutaneous Fat Layer
Phase 6. Miescher’s Radial Granulomas
                                                  Fat Lobules
T lymphocytes
Macrophages
  Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
  Complications
Published August 25, 2019 on www.thecalgaryguide.com

iga-vasculitis-henoch-scholein-purpura-pathogenesis-and-clinical-findings

IgA Vasculitis (Henoch-Schönlein purpura) : Pathogenesis and clinical findings
Authors: Mia Koegler Nela Cosic Reviewers: Crystal Liu Yan Yu* Martin Atkinson* * MD at time of publication
   Infectious Agents
50% have preceding upper respiratory tract infections, i.e., influenza virus or Group A Strep
Drugs
I.e., antibiotics (penicillin, erythromycin), NSAIDs and biologics (tumor necrosis factor α inhibitors)
Immunogenetic and cellular predisposition
Various genetic polymorphisms alter cell- mediated immune response, IgA levels elevated in 50% of people
    ↑ Circulating galactose-deficient IgA1 (GD-IgA1). Deficiency in galactosylation of IgAà↓ IgA serum clearanceàadhesion of IgA complexes, which then deposit into the endothelial lining of blood vesselsàattraction of various inflammatory cells to the area:
Formation of Secretion of Interleukin 8 (IL8) - cytokine that induces Neutrophils infiltrate Activation of complement immune complexes neutrophilic chemotaxis and macrophage phagocytosis the tissue site factors (C3, C4)
Leukocytoclastic vasculitis (histopathologic term for small vessels inflamed by neutrophilic autoimmune response)
              Inflamed cutaneous vessels become enlarged in clusters
Symmetrical palpable purpura (red/purple, non- blanchable papules) distributed on lower limbs and buttocks areas
Cutaneous small vessel vasculitis (100%)
Inflamed gastric vessels - hemorrhage and edema within bowel wall
Gastrointestinal (85%)
Colicky abdominal pain (commonly in the periumbilical region), nausea, vomiting
Gastrointestinal
GI bleeding (hematemesis, melena), Intussusception
Glomerular mesangial proliferation and inflammation
↑ mast cell deposition in joints
Joints (60-85%)
Arthralgia's (common), arthritis (especially knees and ankles)
Arthralgia often transient. No permanent sequelae
                Sympathetic nervous system activation
Glomerulosclerosis, tubulointerstitial and podocyte damage
Renal tissue ischemia
↑ Na sensitivity in renal tubules (↑ Na and water retention)
Renal (10-50%)
Increased renin secretion
          HTN, nephrotic/nephritic syndrome, renal insufficiency
  Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published September 1, 2019 on www.thecalgaryguide.com

Acute-diverticulitis

Acute Diverticulitis: Pathogenesis and clinical findings
Authors: Yan Yu Wayne Rosen* Reviewers: Laura Craig Noriyah AlAwadhi Danny Guo Erica Reed Maitreyi Raman* * MD at time of publication
These regions are perceived in the Left lower quadrant (LLQ) of the abdomen.
• PersistentLLQpain,abdominal guarding and peritoneal signs
• Constipation/obstipation
No bleeding (unlike diverticulosis)
Dehydration (low JVP, ↑ resting HR, orthostatic hypotension)
      Inherent weakness in the muscle layers of the colonic wall
Low Fiber diet
↑ Stool transit time (↓colonic motility)
Stool build-up, ↑ pressure in colonic lumen
Epidemiology
• Diagnosis of developed nations,
total prevalence = 15%
• More prevalent with ↑ age:
• 30-50% by age 60 • 70% by age 80
Diverticula most commonly form in the descending and sigmoid colon
Triggers cytokine release
Irritationofparietal peritoneum → stimulation ofsomaticnerves
Inflamed vessels are more permeable & leak fluid from the blood into the colon
      Mucosal and submucosal layers of the colon wall herniates through the circular muscle layer, creating colonic diverticula
Continued stress on diverticula causes micro-perforations → bacterial infection
Inflammation of diverticula reaches parietal peritoneum
Clotting of blood in the blood vessels feeding diverticula
Continued inflammation of diverticula causes complications over time
Fever
                               Complete bowel perforation (medical emergency)
Fistulae (through bladder, vagina, skin or gut)
Colonic fibrosis → GI strictures, colonic obstruction
Abscesses
Further Investigations:
• CBC: leukocytosis (due to inflammatory response) • CT = Gold Standard Diagnostic test: shows
inflamed diverticuli as well as complications (i.e. free air in peritoneum due to microperforations)
   Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Re-Published September 1, 2019 on thecalgaryguide.com

virchows-triad-and-deep-vein-thrombosis-dvt

Suspected Deep Vein Thrombosis (DVT):
Authors: Dean Percy Yan Yu Reviewers: Tristan Jones Ryan Brenneis Man-Chiu Poon* Maitreyi Raman* * MD at time of publication
Pregnancy, Oral Contraceptives (OCP)
Pathogenesis and Complications
Platelet Activation
Increased clot formation
Hypercoagulable State
↑ ability for the blood to coagulate upon stimulation
Inherited Disorders
Congenital defect in coagulation (ie. Factor V Leiden, Factor II
mutation, Protein S/C deficiency) ↑ blood clotting ability
Estrogen promotes
hypercoagulability, especially in presence of other risk factors
    Notes:
• Venous thrombus causes pulmonary embolism, arterial thrombus causes stroke
• Previous DVT is risk factor for current DVT
Trauma/Surgery
Malignancy
Abnormal release of coagulation-promoting cytokines
Systemic injuryà activation of coagulation cascade
                       Hypertension
Bacteria Artificial Valve
Physically damages blood vessel walls
Adhere/invade vessel wall
Abnormal surface
Vessel Injury
Exposes tissue factor on damaged cells and subendothelium for vWF binding
Virchow’s Triad
Venous Stasis
Low blood flow rate over site of vessel injury, concentrating blood clotting factors at that site
Fat contains more aromatase, converts more androgens to estrogen
Sedentary lifestyle, poor venous return
        Obesity
               Clot formation typically occurs in leg veins
Deep, large veins allow for blood pooling (stasis, hypercoagulability) Venous return from legs often against gravity (stasis)
Valves in leg veins prone to backflow (stasis)
↓ muscle motion = ↓ venous blood flow
Fracture, immobilization, bedrest, long vehicle/airplane ride
   Destruction of vein valve by clot
Venous Insufficiency
Clot prevents blood from returning to heart. Blood accumulating in the leg results in unilateral leg edema and venous inflammation (redness, warmth, tenderness)
1. 2. 3.
Clot embolizes to the lungs
Thromboembolus
-*Pulmonary embolism (acute life threatening complication)
-Chronic thromboembolic pulmonary hypertension
         Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Re-Published September 1, 2019 on thecalgaryguide.com

Acute-Pancreatitis

Acute Pancreatitis: Pathogenesis and Clinical Findings
Authors: Yan Yu Reviewers: Laura Craig Noriyah AlAwadhi Ryan Brenneis Maitreyi Raman* * MD at time of publication
Associated signs due to intra- abdominal hemorrhage from an unknown mechanism (classically associated with pancreatitis, but happens in <1% of cases):
   Note:
It is not enough to just diagnose “acute pancreatitis”. Full management requires determining underlying etiology with further work-up.
Alcohol
↑ Toxic metabolites within pancreas and Spincter of Oddi Spasms
Gallstones
Migration to common bile duct blocks Sphincter of Oddi
           Hypertriglyceridemia
Unknown
mechanism (rare)
Idiopathic
Further investigations:
CBC: Cell counts elevated, due to sever hypovolemia
Serum [Lipase]: Gold Standard Diagnostic Test; rupture of pancreatic cells releases lipase into circulation
Pancreatic secretions back up, ↑ pressure within pancreas
Hypercalcemia (Rare; Ca2+ depositions in bile ducts block outflow of pancreatic secretions)
Since pancreas is retroperitoneal, somatic
nerves in the parietal peritoneum are directly stimulated
Inflammation triggers cytokine release
Inflamed pancreas irritates adjacent intestines, causing ileus
Inflamed, more permeable blood vessels leak fluid into pancreas
• •
Cullen’s sign (bruising in peri-umbilical region) Grey-Turner’s sign (bruises along both flanks)
Sudden, severe epigastric pain (with peritoneal signs), radiates to the center of the back
Fever, nausea/vomiting
(general signs of inflammation)
Diminished bowel sounds Profound dehydration
(flat JVP, hypotension, tachycardia, oliguria) – may happen, not always
      1. Pressure compresses pancreatic blood vessels, causing tissue ischemia.
2. Activation of inactive proteases (zymogens) digesting pancreatic tissue
Necrosis (death) of pancreatic cells
               Inflammation self- perpetuates
    Massive systemic inflammatory response
         2 main complications, usually detected on CT;
may happen, but not always
1. Pancreatic pseudocyst (enlargement of the
pancreas due to fluid accumulation)
2. Pancreatic necrosis/abscesses (death of a part of the pancreas)
  Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Re-published September 1, 2019 on thecalgaryguide.com

Infantile Colic

Infantile Colic: Pathogenesis & Clinical Findings
Gastrointestinal (GI) factors
↑ bile acid wasting ↓ bile acid production in immature in immature gut enterohepatic circulation
↓ bile acid availability
  Psychosocial factors
Infant temperament Over/understimulation
Parental variables (e.g. parental stress)
Collectively, these factors influence the infant’s reactivity to adverse stimuli and the caregiver’s perception of whether crying is problematic.
Other biologic factors
Poor feeding techniques: under/ overfeeding, swallowing air, infrequent burping
Altered gut motility
GI discomfort
Infantile Colic
          Dietary intolerances: cow’s milk protein, lactose
↑ gas production and gut distension
↓ mucosal barrier function
Loss of bacteriostatic effects of bile acids
            Immature enteric nervous system
Intestinal microbial imbalance
↑ intestinal permeability
↑ systemic inflammation
Altered central and enteric neuronal function via microbiota-gut-brain axis
Altered perception of pain and other GI stimuli
            Crying for no apparent reason that lasts > 3 hours/day and occurs ≥ 3 times/week for > 3 weeks in an otherwise healthy infant < 3 months old. There must be normal growth, development, and physical exam. Colic itself is a benign, self-limiting condition that resolves with time.
      Facial flushing or grimacing Tense or distended abdomen
Drawing up of legs Clenching of fingers Stiffening of arms Arching of back
Distress expressed via behaviour
Loud, high- pitched, urgent cry
↑ risk of non- accidental trauma
GI factors directly affect infant’s behaviour
Hypothesized role of immature CNS regulation of circadian rhythm
↑ parental stress
Immature regulation of behaviour
Episodes cluster in evening and/or late afternoon
↑ risk of post- partum depression
Paroxysmal crying Inconsolable
Authors: Simonne Horwitz Nicola Adderley Reviewers: Crystal Liu Yan Yu* Danielle Nelson* * MD at time of publication
                  Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published September 28, 2019 on www.thecalgaryguide.com

Ischemic Colitis

Ischemic Colitis: Pathogenesis and clinical findings
    Superior and inferior mesenteric arteries (SMA and IMA) supply blood to colon
Surgical repair of aorta
Borders of SMA and IMA collaterals at the splenic flexure and rectosigmoid junction are vulnerable to ischemia (“watershed” areas)
Atherosclerosis and narrowing of mesenteric arteries
Low flow state
(e.g., CHF, hypotension, arrhythmia)
Underlying CAD/PVD
Atrial fibrillation, endocarditis
Embolic arterial occlusion of SMA and/or IMA
Trauma, infection, clotting abnormalities
Mesenteric vein thrombosis
Vascular risk factors (e.g., smoking, hypertension)
Thrombotic arterial occlusion of SMA and/or IMA
               Endograft coverage of IMA
Nonocclusive hypoperfusion
      Inadequate blood flow to meet the cellular metabolic needs in the colon
Ischemic Colitis
Tachypnea Tachycardia Hyperthermia Hypotension
         Ischemic period
Loss of oxygen and nutrients to bowel
Reperfusion period
Influx of O2àreacts to produce more oxygen free radicals
Lipid peroxidation
Systemic inflammatory response syndrome*
Nausea and vomiting
Abdominal pain (generally left sided)
Peritonitis
Leukocytosis
Systemic shock
(inadequate perfusion to tissue)
Author: Audrey Caron Michael Blomfield Reviewers: Tony Gu Yan Yu* Edwin Cheng* * MD at time of publication
                Systemic shock
(inadequate perfusion to body tissue)
Hematochezia (Bloody stool)
Gangrene (tissue death)
Hemorrhage
Tissue damage/cell death (starting from mucosa and submucosa going outwards to serosa)
          Mucosal ulceration
Colonic inflammation
    Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
  Complications
Published November 10, 2019 on www.thecalgaryguide.com

Endometritis

Endometritis: Pathogenesis and clinical findings
       Prolonged
rupture of membranes
> 24 hours between amnion
rupture and delivery
↑Time for vaginal flora to ascend into the uterus
Assisted vaginal delivery
Use of forceps or vacuum
Multiple digital vaginal exams
Manual examination of the vagina to assess cervical dilation
Internal monitoring
Intrauterine device to monitor the fetus or contractions
Group B Streptococcus colonization
Opportunistic bacteria present
in the normal vaginal flora of up to 30% of women
Bacterial Vaginosis
Overgrowth of anaerobic bacteria with associated decrease in protective Lactobacillus species
Foreign bacterial ascension into the uterus
Sepsis
Cesarean delivery
                       ↑ Exposure of vaginal flora to the uterus
Introduction of bacteria directly into the uterus
Spread of infection to myometrial and parametrial layers of uterus
Authors: Gabrielle Wagner Reviewers: Danielle Chang Crystal Liu Aysah Amath* * MD at time of publication
  ↑ Susceptibility to bacterial invasion of the uterine lining
Endometritis
Postpartum infection of the uterine endometrial lining
Activation of innate Fever,
         immune response Inflammation of uterus
Leukocytosis
   Accumulation of WBCs within vaginal discharge
Purulent or foul-smelling lochia (vaginal discharge)
Uterine tenderness
Pelvic and/or abdominal pain
          Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published November 26, 2019 on www.thecalgaryguide.com

Viral-Hepatitis

Viral Hepatitis: Pathogenesis and clinical findings Infection with a virus that targets the
Authors: Sean Spence Tyler Anker Yan Yu Reviewers: Dean Percy Crystal Liu Sam Lee* * MD at time of publication
  liver, e.g. HAV, HBV, HCV, HDV, HEV
Hepatocytes are invaded & damaged
Foreign particles and tissue damage activate immune systemàliver inflammation
Lysis (bursting) of hepatocytes
Infection with chronic viruses (HBV and HCV) persist over time and additional symptoms may develop
RUQ pain/tenderness
If infection is prolonged or severe, inflammation becomes systemic
Release of hepatocyte’s cellular contents into the bloodstream
Infection with acute viruses (HAV and HEV) resolve over time, and the symptoms above normalize
Notes:
• HDV can only infect people with concomitant HBV infection
• HAV and HBV vaccines are the only ones that currently exist
• Not all patients with viral hepatitis will develop each of these symptoms. The presentations vary.
Fever, nausea, vomiting ↑ serum ALT, AST
                         ↓ Hepatic metabolic activity (e.g. reduction of gluconeogenesis)           ↓Serum Glucose
↓ Synthesis of plasma proteins (albumin, clotting factors, etc)         ↓ Albumin, ↑ INR
Abbreviations:
• HAV - Hepatitis A Virus
• HBV - Hepatitis B Virus
• HCV - Hepatitis C Virus
• HEV - Hepatitis E Virus
• RUQ - Right Upper Quadrant
• ALT - Alanine Aminotransferase
• AST - Aspartate Aminotransferase
• INR - International Normalized Ratio
 ↓ Bilirubin clearance from blood, bilirubin ends up under the skin         Jaundice Portal Hypertension
Encephalopathy, Splenomegaly, Esophageal Varices, Ascites, Caput Medusae, Edema
Encephalopathy, Muscle Wasting, Metabolic Bone Disease, Terry’s Nails, Ascites, Bruising, Clubbing, Edema
Spider Nevi, Altered Hair Patterns, Testicular Atrophy, Gynecomastia, Palmar Erythema
      Progressive deterioration in liver function, possibly ending up in cirrhosis. (See slide on “Cirrhosis: pathogenesis and complications” for more details on mechanisms and full explanations.)
Hepatic Insufficiency Hyperestrogenism
        Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Re-Published January 12, 2020 on www.thecalgaryguide.com

Innate-Immune-Response

Innate immune response: Pathogenesis and clinical findings
Authors: Erin Stephenson Reviewers: Jessica Tjong Crystal Liu Nicola Wright* * MD at time of publication
  Pathogens overcome chemical barriers (e.g., lysozyme, low pH)
Pathogens overcome physical barriers (e.g., epithelium, cilia)
Trauma
Damage-associated molecular patterns
       Pathogen-associated molecular patterns
Examples of tissue-resident macrophages: • Alveolar macrophages – Lung
• Histiocytes – Connective tissue
• Kupffer cells – Liver
  Recognition by pattern recognition receptors (e.g., toll-like receptors)
• Mesangial cells – Kidney • Microglial cells – Brain
• Osteoclasts – Bone
Microbe engulfed and exposed to oxidative burst
Microbes destroyed
Pus
      Pro-inflammatory chemokines
Recruitment of circulating
granulocytes and monocytes
Pro-inflammatory cytokines (e.g., IL-1β, TNFα, IL-6)
Tissue-resident macrophage activation
Antimicrobial proteins
Unresolved infection/ inflammation
Antigen presented to T cells
Recruitment of adaptive immune response
Enhanced immune responses
                        Acute phase protein production by liver (i.e., C- reactive protein)
Prostaglandin production in the hypothalamus
Fever
Endothelial tight junctions on vasculature disrupted
Intravascular fluid leak into extravascular space
Edema
              Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published January 19, 2020 on www.thecalgaryguide.com

vomiting-pathogenesis

Vomiting: Pathogenesis
Authors: Julena Foglia Reviewers: Varun Suresh Matthew Harding Haotian Wang *Yan Yu *Eldon Shaffer *MD at time of publication
Intracranial:
Trauma, Infection, Tumor, Stroke
↑ Intracranial pressure
Mechanism Unknown
     Irritation of GI mucosa: Inflammation, Distention, Chemotherapy, Radiation
Activates receptors in gut mucosa
GI Disease: Upper: GERD, PUD, Cancer Lower: Ischemia, obstruction, IBD
Mechanical pharyngeal stimulation
Signal travels via vagal and sympathetic afferent nerves
Metabolic:
Pregnancy, Diabetes, Uremia,
Thyroid disease, Hypercalcemia
Pain, Smells, Foul Sights, Memories
Sensory inputs to cortical region
Cerebral Cortex
Vomiting (Emetic) Center
Toxins circulating in bloodstream: Chemotherapy, Opioids
Offending substance travels through circulation and binds to receptors in the CTZ, outside the blood brain barrier
Abbreviations:
GERD: Gastroesophageal Reflux Disease PUD: Peptic Ulcer Disease
IBD: Inflammatory Bowel Disease
CTZ: Chemoreceptor Trigger Zone
CNX: Cranial Nerve Ten
H1: Histamine Receptor
M1: Muscarinic Receptor
Disrupted inner ear balance: Motion Sickness
Activation of H1 & M1 receptors in vestibular center traveling via Cerebellum
                        Stimulates Solitary Tract Nucleus (Medulla)
  (Medulla)
Vagus Nerve (CNX) and enteric nervous system activation, resulting in:
        Gastric relaxation, ↓ pylorus tone, retrograde duodenal peristalsis
Downward diaphragm contraction, abdominal & chest wall muscles contract: ↑ intra-gastric pressure
Vomiting
(Forceful expulsion of material from stomach and intestines)
Upper and lower esophageal sphincter relaxation and glottis closure
    Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
  Complications
 Re-published February 16, 2020 on www.thecalgaryguide.com

C5-C9-deficiency

C5-C9 deficiency: pathophysiology and clinical findings
Authors: Heather Yong Reviewers: Jessica Tjong, Crystal Liu, Yan Yu*, Nicola Wright* * MD at time of publication
   Normal complement response
The complement pathway is a non- specific response to bacterial pathogens
Bacterial infection
Classical, alternative, or lectin pathway activation
Complement cascade
MAC formation on bacterial surface C5b, C6, C7, C8, C9
Complement proteins create trans-
membrane channels within bacterial cell walls/cell membranes
Critical bacterial proteins leak out of the cell, breakdown of entire cell
Primary (hereditary) causes Secondary (acquired) causes
All are autosomal recessive Biologic therapy ex. eculizumab Absence or suboptimal functioning of
    Abbreviations:
• MAC: Membrane Attack
Complex
• CH50: Classic Hemolytic
Complement Test
• AH50: Alternative Hemolytic
Complement Test
• CNS: Central Nervous System • CSF: Cerebrospinal Fluid
≥1 terminal complement proteins
C5-C9 deficiency
Inability to form MAC
↑ susceptibility to systemic Neisseria infection
Commonly N. meningitidis Rarely N. monorrhoeae
Nasopharyngeal colonization of N. meningitidis, ↑ susceptibility to bacteremia
CH50 ± AH50 assay No lysis
Note:
Total complement activity assay <10% activity C5, C6, C7, C9 <50% activity C8
              Varied bactericidal action via other complement proteins
• Risk of invasive meningococcal disease is 1000-10000X higher in C5-C9 deficiency than in the general population
• Reason is unknown
• C5-C9 deficient patients are not at greater
risk for contracting other gram (-) infections • Clinical meningitis in C5-C9 deficiency is less
       severe and fatality is rare
                   Bacterial lysis
Especially gram (-)ve bacteria like Neisseria
Bacteria cross the blood-brain barrier, causing swelling and damaging brain tissue
Fatigue, fever, headache, altered mental status, etc.
Inflammation of CSF and meninges
Activation of dura and pia mater fibres
Headache, neck stiffness
Bacteria release toxins
Damage to surface blood vessels
Maculopapular rash
Exact mechanism unknown
Recurrent meningitis
         CNS damage due Sepsis to recurrent
meningitis
  Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
  Complications
Published February 16, 2019 on www.thecalgaryguide.com

Fecal-Incontinence

Fecal Incontinence: Pathogenesis and complications
Note: The majority of fecal incontinence is multifactorial in cause
Authors: Timothy Fu Reviewers:
    Chronic bowel straining
Difficult vaginal delivery
Direct internal anal sphincter impairment (controls ~70% of anal resting tone)
↓ anal resting tone
Aging:
Degene- ration of muscle fibers
Movement disorders (e.g. arthritis, Parkinson’s); aging is a risk factorà ↓ mobility
↓ timely access to bathrooms
Inflammation
of colon (e.g., Ulcerative colitis, Radiation proctitis)
↓ capacity of
rectal smooth muscle to stretch
↓ capacity to store stool
↑ urgency of defecation
↑ reflex relaxation of internal anal sphincter
Chronic diarrhea, diarrhea- predominant irritable bowel syndrome, laxatives
Yan Yu* Erika Dempsey* * MD at time of publication
         Stretch injury of     Pelvic surgery
Chronic constipation
Build up of solid, immobile mass of stool in the rectum
Loose stool is able to flow around impacted stool, exiting anal canal (overflow diarrhea)
Sensory neuro- pathy (e.g. Diabetes)
Altered
mental conditions (e.g. stroke, dementia)
 pudendal nerve (innervating the pelvic muscles and external anal sphincter)
Local neuronal damage
Impaired pelvic muscle and external anal sphincter motor control
Pelvic trauma
Rectal prolapse
Direct external anal sphincter impairment
↑ Stool volume
↑ Loose stools
Rectal hyposensitivity (↓ perception of rectal distension)
Patient fails to sense rectal fullness and voluntarily releases their external anal sphincter
                                     Voluntary external anal sphincter contraction is no longer sufficient in closing the anus
Loose stool is more prone to escape through anal canal compared to solid stool
        Continence mechanisms are impaired
Fecal Incontinence: The unintentional loss of solid or liquid stool
Skin Skin
Continence mechanisms are intact, but overwhelmed or ignored
       infection Skin erythema
erosion
   Inability to control what is widely considered a basic, fundamental bodily process
↑ caretaker burden Social stigma
↑ skin contact with acidic irritant (stool)
        ↑ rate of institutionalization, (e.g., admission into long-term care)
↓ confidence, sense of agency
↑ stress, anxiety
Skin inflammation
↓ social activity, work ↓ help-seeking ↓ treatment
       Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published May 2, 2020 on www.thecalgaryguide.com

Diabetic-Nephropathy

Diabetic Nephropathy: Pathogenesis Type I or Type II Diabetes
      ↑ Glucose uptake & glycolysis in glomerular & tubular cells
Poor glycemic control = ↑ Glucose load
↑ Proximal tubule reabsorption of Na via Na/glucose co-transporter
↓ NaCl to distal tubule
Activation of tubulo- glomerular feedback at macula densa
   to kidney
↑ Activation of renin- angiotensin system (RAS)
↑ Intrarenal angiotensin II
      ↑ Advanced glycation end products (AGE)
Shunting of glucose through non- glycolytic pathways (e.g. polyol)
Activation of Protein Kinase C (PKC) pathway
Excessive production and accumulation of glycolytic intermediates (e.g. sorbitol, hexosamine, succinate)
hyperglycemia
Succinate via GPR91
         ↑ Free radical production (oxidative stress)
Activation of cellular signalling, transcription factors and cytokines (e.g. TGF-β-Smad-MAPK, IGF-1, NF-κB)
↑NADPH oxidase activity
       ↑Blood volume ↑ Blood pressure ↑ Renal perfusion
Relative afferent arteriole dilatation, efferent arteriole constriction
Initial ↑ in glomerular filtration rate (GFR)
Podocyte loss/injury
         Authors: Steven Chen Shannon Gui Yan Yu* Reviewers: Julia Heighton Ryan Brenneis Sophia Chou* * MD at time of publication
Initial glomerular hyperfiltration at time of diagnosis
↓ Production of matrix metallo- proteinases
Aberrant extracellular matrix (ECM) protein expression and accumulation
Sheer stress to glomeruli à pressure-induced damage
↑ Glomerular basement membrane permeability to proteins like albumin
               ↓ Extracellular matrix regulation
“Metabolic Pathway”
Mesangial matrix expansion
Kimmelstiel-
Wilson lesions (pink
hyaline nodules due to accumulation of damaged proteins)
Tubular fibrosis
Scarred glomeruli are less able to effectively filter blood
↓ in glomerular filtration rate (GFR)
Albuminuria
Usually occurs after ~5 years from time of diagnosis in T1DM; can occur at time of diagnosis in T2DM
            Abbreviations
IGF: Insulin-like growth factor
MAPK: Mitogen-activated protein kinases NADPH: Nicotinamide adenine dinucleotide phosphate
NF-κB: Nuclear factor kappa-light-chain- enhancer of activated B cells
TGF-β: Transforming growth factor-β
Protein endocytosis into tubular cells causing inflammation
“Hemodynamic Pathway”
     Diabetic Nephropathy
Overt diabetic nephropathy may take upwards of 15-25 years to develop
Note: The mechanisms presented here have been simplified. The cross- talk and signaling between the metabolic and hemodynamic factors do not manifest in a step-wise fashion, but rather occur in parallel.
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
  Complications
Published May 3, 2020 on www.thecalgaryguide.com

Infarctus du myocarde: Antécédents médicaux

Infarctus du myocarde:
Antécédents médicaux
Infarctus du myocarde (nécrose 8ssulaire)
Auteur: Yan Yu Traductrice: Olivia Genereux Réviseurs: Sean Spence Tristan Jones Nanette Alvarez* Marie Giroux* *MD au moment de publication
Sang du ventricule gauche reflue à l’oreille[e gauche et finalement s'accumule dans le système vasculaire pulmonaire
Haute pression du système vasculaire pulmonaire force fluide hors des capillaires et dans l'interstitium pulmonaire et les alvéoles
     Inflammation myocardique locale
Médiateurs inflammatoires irritent les nerfs cardiaques (plexus cardiaque)
 ̄ Fonc8on systolique (myocarde nécro6que ne peut pas se contracter suffisamment)
        Invasion généralisée des cytokines inflammatoires
Cytokines agissent les régulateurs de température hypothalamique
Fièvre légère
Irritation des nerfs sympathiques afférents (T1-T4)
Signal entre moelle épinière, a/n dermatomes T1-T4
Cerveau perçoit irritation des nerfs comme douleur des dermatomes T1-T4
Douleur écrasante, pression, oppression de la poitrine: Souvent rétrosternale, avec radiation à épaule, cou et l'intérieur des deux bras (D>G) (Apparition: au repos, crescendo)
IrritaNon des branches cardiaques du nerf vague
AcNvaNon réponse vasovagale
Fatigue, étourdissements, nausée, vomissement
↓ volume systolique (VS) ↓ debit cardiaque (DC)
­ Activité sympathique (pour essayer de maintenir DC)
                ­ Sueurs (diaphorèse)
Peau moite
VasoconstricNon généralisé
Vasoconstriction des artérioles de peaux
Peau froide
Interstitium
pulmonaire
mou ↓
compliance
pulmonaire
d
Fluide compresse les voies respiratoires, ↑ résistance au flux d’air
                           Abrévia8ons:
• a/n – au niveau
À Noter: Douleur myocardique ischémique peut présenter différemment entre les patients, mais les symptômes récurrents habituellement présentent les mêmes pour un patient donné.
Muscles respiratoires travaillent plus fort pour venNler les poumons
Essoufflement
(difficulté à respirer)
 Légende:
 Physiopathologie
Mécanisme
Signe/Symptôme/Résultats de Laboratoire
  Complications
 Publié Janvier 20, 2013 sur www.thecalgaryguide.com

preterm-labour-pathogenesis-maternal-complications

Preterm Labour: Pathogenesis & Maternal Complications
        Cervical Procedures
Removing part of the cervix
Ex. Cone biopsy, cervical LEEP
Genitourinary Infections
E.g. Urinary tract infection, bacterial vaginosis, chorioamnionitis, abnormal vaginal flora
↑ bacterial colonization of fetal membrane
↑ bacterial enzymes and immune reactions
Antepartum Hemorrhage
↑ risk placental abruption
↑ decidual tissue factor release
Activates coagulation cascade
↑ thrombin ↑ proteases
Substance Use
Smoking or cocaine use
Vasoconstriction in uterine circulation, endothelial dysfunction
Placental hypoperfusion and ischemia
↑ fetal ACTH
↑ placental prostaglandins
Maternal Stress
Malnutrition, depression, trauma- related disorders, work-related stress
↑ cortisol
↑ placental corticotropin releasing hormone
Uterine Abnormalities
Mullerian duct anomalies: congenital abnormalities in uterine shape
Septate uterus: ridge of tissue dividing uterus into two horns
Intracavitary leiomyoma: benign mass inside uterus
↑ Uterine volume
Maternal Genome
Family hx or personal hx of preterm birth, previous preterm premature rupture of membrane
                        ↓cervical stroma & ↑cervical scarring
↓ cervical glandsà ↓ mucous production
↑risk of infection ↑ inflammation
↑prostaglandins
Cervical collagen degradation
                   Rapid growth exceeds blood supply
Ischemia & necrosis of fetal tissue
↑prostaglandins and cytokines
↓ functional volume of uterine cavity
Multifetal pregnancy, poly- hydramnios
               ↓tensile strength and plasticity
Uterine stretch
Upregulation of oxytocin receptors
Genes for ↑ risk
Mechanism unknown
                      Cervical Insufficiency
Pathologic cervical dilation and/or effacement (thinning)
Authors: Skye Russell Reviewers: Danielle Chang, Crystal Liu, Yan Yu*, Nicholas Papalia*
*MD at time of publication
Digest and weaken amniotic membrane
Preterm Premature Rupture of Membranes
↑ myometrial sensitivity to oxytocin
Uterine contractions Preterm Labour
           Uterine contractions and cervical change occurring <37wk gestational age ↑ Risk of future preterm labour and preterm birth
Abbreviations:
• LEEP – Loop electrosurgical
excision procedure
• ACTH – adrenocorticotropic
hormone
   Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
  Complications
 Published July 12, 2020 on www.thecalgaryguide.com

acute-pancreatitis-complications

Acute pancreatitis:
Complications
Inflammation causes vasodilation and vasculature leakage
Mild, (85%):
Interstitial edematous pancreatitis
Local accumulation of fluid in the pancreas
<2 weeks after onset
Acute peripancreatic fluid collection (not encapsulated)
Walled off by fibrous & granulation tissue
>2 weeks after onset
Pancreatic pseudocyst
(completely encapsulated)
Peritoneal irritation à pain
Large cyst can (very rarely) compress surrounding bowel
Acute Pancreatitis
Inflammatory cytokines are released from damaged pancreas
If recurrentàchronic pancreatitis (see relevant slide)
Inflammation damages pancreatic exocrine
cellsàInappropriate release of pancreatic enzymes into surrounding tissue & vasculature àdigesting pancreatic parenchyma
Authors: Nissi Wei, *Yan Yu Reviewers: Dean Percy, Miles Mannas, Varun Suresh, Brandon Hisey, *Kerri Novak, *Sylvain Coderre * MD at time of publication
                     complete resolution (most cases)
Necrotic tissue is vulnerable to
infection (esp. Gram neg GI bacteria)
inflammation & necrosis activate cytokine cascade
Severe, necrosis (15%): Necrotizing pancreatitis
Local infection
Severe pancreatic inflammation shifts body fluid into retroperitoneal spaceàintravascular volume depletion
                 Systemic Inflammatory Response Syndrome (SIRS) (see relevant slide)
Organ failure (may be sole feature on presentation)
Stagnant fluid can more easily become infected
Infection spreads to bloodstream
Cardiac failure Hypovolemic shock Renal failure
Local accumulation of fluid & necrosis in the pancreas
< 4 weeks after onset:
Acute necrotic collection (not encapsulated)
Walled off by fibrous & granulation tissue
> 4 weeks after onset
walled-off necrosis
(completely encapsulated)
When treated with excess fluid resuscitation:
Intra- abdominal hypertension
                Respiratory failure (ARDS)
Disseminated intravascular coagulation (DIC)
          Bowel obstruction Gastric outlet (see relevant slide) obstruction
Infected pancreatic necrosis
  Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
  Complications
Published September 20, 2016, updated September 7, 2020 on www.thecalgaryguide.com

Cellulitis

Cellulitis: Pathogenesis, clinical findings and complications
Authors: Tegan Evans, Spencer Yakaback Reviewers: Brian Rankin, Timothy Fu, Laurie Parsons*, Yan Yu* * MD at time of publication
Cracked skin Surgery
  Normal Skin
Epidermal layer
Dermal-Epidermal Junction
Dermal layer
Subcutaneous fat
Resident skin flora:
Coagulase-negative Staphylococci*
Transient skin flora:
Staphylococcus aureus* Streptococcus pyogenes Gram negative bacteria Fungi
                      Pathogen in deep dermis and subcutaneous fat
*most common pathogens
Break in skin barrier (may not be obvious) and entry of pathogen
    Risk Factors: Immunocompromised Host: -Diabetes mellitus+ -Lymphedema -Malnourishment
-Older patient+
-Obesity+
-Peripheral vascular disease General Infection Risk: -History of cellulitis+ +highest risk factors
Risk Factors for MRSA Cellulitis: Increased exposure to MRSA: -Contact sports
-Crowded living conditions -Health care workers -Indigenous descent
-Sharing towels, equipment
Increased susceptibility:
-Immunodeficiency -Young age
Direct inoculation (e.g. trauma) Organism virulence overwhelms host defense mechanisms (related to risk factors)
  Cellulitis: A bacterial infection in which pathogens penetrate deep dermis and/or subcutaneous fat
Cytokines activate immune response
Accumulation of pus (bacteria, white blood cells, dead skin)
Abscess formation
  Infection spreads to nearby lymph nodes
Lymphadenitis
Infection spreads through lymph vessels
Ascending lymphangitis
Local inflammatory response in skin
Pain Warmth Edema Erythema (redness)
with indistinct margins
Vesicles and bullae
Organisms penetrate blood vessels
Bacteremia (presence of bacteria in blood)
                         Systemic inflammation
Distant spread to bone
Osteomyelitis
Distant spread to endocardium (inner lining of heart chambers and valves)
Endocarditis
           Fever Malaise
Chills
Sepsis
     (rarely)
 Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published September 27, 2020 on www.thecalgaryguide.com

Tumour-Lysis-Syndrome

Tumour Lysis Syndrome
High sensitivity to treatment
Highly proliferative malignancy with ↑cell-turnover
Large tumour mass
Author: Joshua Yu Reviewers: Hannah Yaphe Davis Maclean Tejeswin Sharma Yan Yu* *Peter Duggan *ManChiu Poon *Lynn Savoie *Juliya Hemmett * MD at time of publication
        High serum phosphate
High serum Advanced uric acid Age
Volume depletion
Pre-existing renal disease
       Patient risk factors
Initiation of cancer treatment
Massive tumour cell lysis and release of cellular contents into circulation overwhelms homeostatic mechanisms
Tumour properties (hematologic malignancies most common)
Though rare, aggressive tumours can spontaneously lyse without treatment
     Intracellular potassium released into bloodstream
           Intracellular phosphate released
Hyperphosphatemia
↑ serum phosphate
Intracellular lactate dehydrogenase (LDH) released
↑ serum LDH
Intracellular nucleic acids released
Nucleic acids metabolized to uric acid
Hyperuricemia
↑ serum uric acid
            Hyperkalemia
↑ serum potassium
(see Hyperkalemia: clinical findings)
Uric acid (a crystallizing substance) ↑ precipitation of calcium phosphate
↑ filtration of poorly soluble uric acid into acidic environment of renal tubules
        Serum phosphate binds serum calcium, forming solid calcium phosphate precipitate crystals
High levels of calcium phosphate ↑ uric acid precipitation
Uric acid precipitates as crystals and deposits in kidney tubules and collecting ducts
Tubular injury and/or intraluminal obstruction
Endothelial dysfunction in renal vasculature
Renal inflammation, vasoconstriction, and
impaired renal vascular autoregulation
Decreased renal filtration
Acute Kidney Injury
(see Acute Kidney Injury Overview)
          Crystal deposition in the heart
Depletion of soluble calcium
Hypocalcemia
Calcium phosphate crystals deposit in kidneys
Main mechanism of Acute Kidney Injury
         Cardiac Arrythmias
↓ serum calcium
(see Hypocalcemia: clinical findings)
  Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
  Complications
Published October 25, 2020 on www.thecalgaryguide.com

Multiple-Sclerosis-on-Brain-MRI

Classic Findings of Multiple Sclerosis (MS) on Brain MRI
Authors: Evan Allarie Davis Maclean Viesha Ciura* Reviewers: Yan Yu* * MD at time of publication
infiltrates in the infratentorial region
   Note: variation in findings exist. The findings shown here are not exhaustive but are some of the most common areas implicated on brain MRI in MS. Most common sites that lesions are observed are: juxtacortical regions, periventricular, infratentorial, spinal cord, and the optic nerve. However lesions can occur anywhere there is myelin in the CNS.
Active inflammation and destruction allows for gadolinium contrast to cross the blood- brain barrier, which can be visualized as a marker of active inflammation in MS
Classic optic nerve (CN II) lesion seen in optic neuritis. Gadolinium enhancement (↑ signal
intensity of lesion after gadolinium injection) in this T1 image demonstrates active inflammation
Image Credits: Dr. Viesha Ciura
For further pathogenesis, see the Calgary Guide slide: Multiple Sclerosis (MS): Pathogenesis and Clinical Findings
Inflammation within the central nervous system (CNS) T-cell, B-cell, and macrophages infiltrate CNS
     Infiltration occurs through veinsàlocal inflammation Perivenular infiltrates around the medullary veins
     (perpendicular to ventricles)
Inflammation and blood-brain barrier destruction ↑ extravascular inflammatory fluid around lesions, which appears hyperintense/bright
Loss of neuronal/axonal density in affected region, replaced by gliosis over time
          Lesions extend out around veins, creating characteristic ovoid lesions
Classic hyperintense T2/FLAIR perpendicular periventricular plaques following the medullary veins – ‘Dawson’s Fingers’
Middle cerebellar peduncle T2/FLAIR hyperintense lesion in classic location for MS
             Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
  Complications
Published October 25, 2020 on www.thecalgaryguide.com

Bronchiolitis-updated

Bronchiolitis: Pathogenesis and clinical findings
Viral pathogen, most commonly respiratory syncytial virus (RSV) - but can be others such as rhinovirus,
adenovirus, parainfluenza, influenza, and coronaviruses - initially colonizes the nasopharyngeal mucosa Virus travels via the epithelium to the lower airways to the terminal bronchioles (small airways)
Authors: Nick Baldwin Rebecca Lindsay Reviewers: Kayla Nelson, Yan Yu Timothy Fu, Danielle Nelson* *MD at time of publication
         Upper airway mucosal inflammation
Bronchiolitis
(bronchiole inflammation)
Apnea
(cessation of breathing; via unknown mechanism, potentially apnea reflex)
RSV-fusion protein facilitates fusion of the virus to the host cell and directs viral penetration as well as facilitates fusion of the infected cell with its healthy neighbors
Forms syncytia (multinucleated cells)
          Cytokines are released into circulation
↑ thermo- regulatory set- point at the hypothalamus
Mild Fever
Copious coryza
(nasal discharge)
Protein and fluid leak into nasopharyngeal interstitium
↑ Capillary permeability
Protein and fluid leak into the bronchiole interstitium, accumulating around airway walls
Airway wall becomes thickened, more readily apparent on x-ray
Peribronchial cuffing
(X-ray finding: bronchi appear like thickened ‘cuffs’ when viewed head-on)
Inflammation stimulates the upregulation of mucous secreting goblet cells
↑ mucous production
             Mucous within alveoli ↑ intra- alveolar surface tensionà collapsing alveolar walls
During inspiration, air enters the collapsed alveoli if airway is not yet occluded
↑ intra-alveolar pressure causes the alveoli to suddenly pop open
Inspiratory crackles on auscultation
Syncytia slough off the bronchial epithelium into airways
Airways become narrower and occlude
Disruption of the ciliated epithelial cells (which transport mucous out of the airways and into the pharynx, to be swallowed or evacuated)
↓ mucous clearance from airways
Excess airway mucous triggers cough reflex
Cough
               Interstitial edema
Nasal Congestion
Air is absorbed distal to occlusion (gas trapping)
When all air is absorbed, alveoli collapse (resorptive atelectasis)
↓ gas exchange between blood and air in remaining alveoli
↓ O2 saturation & ↑ CO2 content of blood
Narrower airways, especially during expiration, causes audible turbulent airflow
Wheeze on auscultation
                      Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
  Complications
Re-Published November 29, 2020 on www.thecalgaryguide.com

mrna-vaccines-against-coronavirus-disease-2019-covid-19-production-and-mechanism-of-action

mRNA Vaccines against Coronavirus Disease 2019 (COVID-19):
Production and Mechanism of Action
Vaccine Production
The “spike protein” is known to be a major viral surface
Authors: Ryan Brenneis, Yan Yu* Reviewers: Davis MacLean, Hannah Yaphe, Timothy Fu, Stephen Vaughan* * MD at time of publication
References
1. ACS Nano 2020, 14, 10, 12522–12537, Publication Date: October 9, 2020, https://doi.org/10.1021/acsnano.0c07197
2. NEJM 2020, Publication Date: December 10, 2020, DOI: 10.1056/NEJMoa2034577
3. Expert Review of Vaccines 2017, 16, 9, 871-881, Publication Date: 2017, DOI: 10.1080/14760584.2017.1355245
4. NEJM 2020, 383, 2439-2450, Publication Date: December 17, 2020, DOI: 10.1056/NEJMoa2027906
5. NEJM 2020, 383, 2427-2438, Publication Date: December 17, 2020, DOI: 10.1056/NEJMoa2028436
6. BMJ 2000, 321, 7271, 1237-1238, Publication Date: November 18, 2000, DOI: 10.1136.bmj.321.7271.1237
Notes:
  SARS-CoV-2 (RNA virus causing COVID-19) collected from an infected patient (ie. with a nasopharyngeal swab)
Various reagents are added to the sample containing both human cells and viral constituents
Reagents cause cell/viral membrane lysisàspilling cell contents, viral particles, and viral RNA
Fats, proteins, and carbohydrates removed through various washing reagents, leaving nucleic acids (like RNA)
Reverse transcriptase polymerase chain reaction produces complementary DNA (cDNA) from viral RNA
cDNA library allows for SARS-CoV-2 genome to be mapped through whole-genome sequencing technology
SARS-CoV-2’s spike protein DNA sequence is identified, and is used as a template to create synthetic viral spike protein mRNA
Extra RNA bases are added to this mRNA strand to promote its stabilityàresulting RNA strand is now called “nucleoside-modified RNA” (“modRNA”)
          antigen (substance that elicits an immune response) from studies of other coronaviruses (e.g. SARS- CoV-1 and MERS-CoV)
             Pfizer/BNT162b2 vaccine contents:
Moderna mRNA-1273 vaccine:
  Note: Lipid nano- particles are spherical hollow “balls” made of an outer lipid membrane plus other emulsifiers and membrane stabilizers.
Lipid nanoparticles are capable of engulfing smaller molecules (like RNA) and merging with normal cell membranes
Spike protein modRNA is then isolated (using a series of precipitation, extraction, and chromatography methods)
Final modRNA lipid nanoparticle vaccine is now created and ready for intramuscular injection
The modRNA vaccine is injected intramuscularly into a healthy person 2nd dose after 3-4 weeks needed to strengthen the immune response
(to a level exceeding the immune response in patients recovered from Covid-19), boosting vaccine efficacy especially in older individuals4,5
Lipid nanoparticle fuses with human cells’ phospholipid membranes via endocytosis, releasing modRNA into the cell’s cytosol
modRNA is translated by human ribosomes naturally found in the cell’s cytosol, producing viral spike protein components
•
•
Foreign substance can cause local tissue inflammation
The spike proteins encoded by the modRNA of each of the two vaccines are similar
It is the proprietary lipid nanoparticle formulation (unknown to the public) that is unique to each vaccine
Pain, redness, swelling at injection site (Transient)
    Proprietary Pfizer/ BioNTech lipid nanoparticle
The modRNA encodes a
full-length spike protein modified with two proline amino acids (for stability and immunogenicity)2
The modRNA encodes a full-length spike protein modified with two proline amino acids (for stability)1
Proprietary Moderna lipid nanoparticle
         Encapsulating this modRNA within Pfizer/ BioNTech’s lipid nanoparticle creates the 162b2 vaccine
This specific formulation requires colder storage temperatures (-700C)
Encapsulating this modRNA within Moderna’s lipid nanoparticle creates the mRNA-1273 vaccine
This specific formulation can be stored at slightly warmer temperatures (-200C)
            Muscles are preferred injection sites as they have greater blood supply than other body tissues
Vaccine Action
Able to bring in immune cells faster to process foreign antigens6
Able to drain away foreign vaccine material fasterà minimizing local reactions6
Cell-mediated Immunity
               Spike protein degraded by intracellular enzymes into fragments
Humoral Immunity
Natural cellular processes release spike protein components from the cell into the bloodstream
Spike protein components are engulfed by antigen presenting cells (dendritic cells, B cells, macrophages), fragmented, & bound to unique MHC Class II proteins
MHC Class II proteins bring spike protein fragments to the antigen presenting cell’s surface, to present them to circulating naïve CD4+ (helper) T cells
Some naïve helper T cells are able to successfully bind to the spike protein-MHC Class II protein complexes
Binding activates these spike-protein specific helper T cells
       Spike protein fragments bound by MHC Class I proteins
MHC Class I proteins bring spike protein fragments to the human cell surface
MHC Class I proteins present spike protein fragments to naïve CD8+ T cell
Naïve CD8+ T cells that able to bind to the spike protein-MHC Class I protein complex become activated, and travel to the lymphatic system to mature
MHC = Major Histocompatability Complex; cell surface proteins key to immune function
CD = Cluster of Differentiation; glycoproteins on T cell surfaces that are co-receptors and facilitate T cell binding to antigens/MHC complexes. They also distinguish the types of T cells.
               Some of these T-Cells mature into cytotoxic T cells that now recognize the viral spike protein
Cytotoxic T-cells bind to human cells infected with SARS-CoV-2 expressing spike protein or spike protein fragments
Cytotoxic T cell releases enzymes perforating infected cell, causing cell death to occur
Immune system identifies and destroys human cells infected with SARS-CoV-2, slowing viral spread
Other T cell’s can mature into memory T cells (stimulated by cytokines released by helper T cells)
Memory T cells travel to lymphatic tissue, awaiting activation from future exposure to spike protein
More rapid cell-mediated immune response to future SARS-CoV-2 infection (immunity)
Activated helper T cells specific to the viral spike protein secrete cytokines to stimulate immune activity
Systemic cytokine releaseàsystemic reactions like fever, chills, fatigue, myalgias (Transient)
Some B cells mature into plasma cells that produce IgG antibodies against the viral spike protein
Antibodies to spike protein mark SARS-CoV-2, allowing immune system to destroy virus
Eradication of SARS-CoV-2 in extracellular compartments
Activated helper T cell interacts with
naïve B cells in lymphatic tissue
Some B cells mature into memory B cells specific to SARS-CoV- 2 spike protein
                       Future exposure to spike protein re-activates memory B cell in lymphatic tissue & creates plasma cells, producing antibodies more rapidly
Rapid humoral immune response to future SARS-CoV-2 infection (immunity)
         Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Clinical Finding
  End Result
 Published December 19, 2020 on www.thecalgaryguide.com

Adenovirus-Vector-Vaccines-Against-COVID19-Production-and-Mechanism-of-Action

Adenovirus Vector Vaccines Against COVID-19: Production and Mechanism of Action
 Johnson & Johnson
Adenovirus type 26 (Ad26), a mild human adenovirus, is isolated
Previous exposure to Ad5 or Ad26 may have sensitized immune system to the adenovirus vector1
Potential for human adenovirus vaccine to fail due to previous exposureàimmunity not built against spike protein
CanSino
Adenovirus type 5 (Ad5), a mild human adenovirus, is isolated
Oxford/AstraZeneca
Chimpanzee adenovirus AZD1222 (ChAdOx1), previously shown to be safe & to elicit an immune response in humans2, is isolated
Vaccine Production
SARS-CoV-2 (virus causing COVID-19) synthetic DNA library sequenced from viral RNA using reverse transcriptase polymerase chain reaction and whole genome sequencing technology
Spike protein DNA sequence isolated from SARS-CoV-2 genome
A promoter sequence is added to the spike protein DNA sequence, allowing human RNA polymerase to recognize and transcribe the spike protein DNA when introduced into human cells
Recombinant genetic technology inserts the modified spike protein DNA into a plasmid: a circular piece of DNA that acts as a shuttle allowing for the insertion of new genes (such as the spike protein gene) into host genomes (like the adenovirus vector DNA genome)
          Adenoviral DNA isolated using various lytic & washing reagents (chemicals that break open cell membranes and remove non- nucleic acid cellular materials)
Adenoviral DNA sequenced using whole genome sequencing, then modified as follows:
Chimpanzee virus negates possibility of previous immunity to the viral vector1
Chimpanzee viral vector more likely to successfully
generate immune response to the spike protein
               E1 region of adenoviral genome E3 region of adenoviral genome deleted to create deleted to block viral replication3 room for insertion of SARS-CoV-2 spike protein DNA3
       Adenovirus used in the final vaccine cannot replicate
within human cells and cannot cause human disease
References
1. ACS Nano 2020, 14, 10, 12522–12537, Publication Date: October 9, 2020, https://doi.org/10.1021/acsnano.0c07197
Modified adenoviral DNA genome is reinserted into   The viral vector & the spike protein plasmid are mixed together, and DNA recombination
the adenovirus particle, creating the “viral vector”
technology inserts the spike protein gene from the plasmid into the adenovirus DNA2
  Adenovirus containing transcribable SARS-CoV-2 spike protein DNA is introduced into a special cellular culture, allowing the virus to replicate despite its modified DNA2, 3
Authors: Ryan Brenneis, Yan Yu* Reviewers: Davis MacLean, Hannah Yaphe Stephen Vaughan* * MD at time of publication
     2. Nature 2020, 586, 578–582, Publication Date: October 20, 2020, https://doi.org/10.1038/s41586- 020-2608-y
3. Frontiers in Immunology 2018, 9, 1963, Publication Date: September 19, 2018, doi: 10.3389/fimmu.2018.01963
4. NPJ Vaccines 2020, 5, 69, Publication Date: July 27, 2020, doi: 10.1038/s41541-020-00221-3
5. The Lancet 2020, Publication Date: Dec. 8, 2020, https://doi.org/10.1016/S0140-6736(20)32623-4
6. BMJ 2000, 321, 7271, 1237-1238, Publication Date: November 18, 2000,
DOI: 10.1136.bmj.321.7271.1237
7. NEJM 2021, Publication Date: Jan. 13, 2021, DOI: 10.1056/NEJMoa2034201
Adenovirus containing transcribable SARS-CoV-2 spike protein DNA is isolated and concentrated to a high enough level for administration as a vaccine
Adenoviruses have an outer protein layer (called a capsid) to protect its DNA
DNA is more stable than mRNA due to deoxyribose sugar backbone and intermolecular bonds between strands
Enhanced stability compared to mRNA lipid nanoparticle vaccines
Can be stored at 2-8°C for up to 3-6 months
              Muscles are preferred injection sites as they have greater blood supply than other body tissues
Immune cells arrive faster to     The viral vector vaccine is injected intramuscularly into a healthy person process foreign antigens6
Foreign substance can cause local tissue inflammation
Pain, redness, swelling at injection site (Transient)
Note: The Johnson and Johnson vaccine may be 90% effective after a single dose7
        Foreign vaccine material drains away fasterà minimizing local reactions6
2nd dose after 28 days recommended to strengthen the immune response (to a level exceeding the immune response in patients recovered from Covid-19), boosting vaccine efficacy especially in older individuals5
  Vaccine Action
Cell-mediated Immunity
Spike protein degraded by intracellular enzymes into fragments
 Adenovirus surface antigens interact with human cellular receptors, allowing viral entry into human cell via endocytosis3 Adenovirus vector travels to cell nucleus, merges with nuclear envelope and injects its DNA (including the spike protein DNA) into the nucleus
RNA polymerases in the nucleus transcribe the viral DNA, making messenger RNA (mRNA) for SARS-CoV-2 spike protein
      mRNA is transported back into the cytosol & translated by ribosomes naturally found there, producing full length SARS-CoV-2 spike protein
Humoral Immunity
Natural cellular processes release spike protein components from the cell into the bloodstream
Spike protein components are engulfed by antigen presenting cells (dendritic cells, B cells, macrophages), fragmented, & bound to unique MHC Class II proteins
MHC Class II proteins bring spike protein fragments to the antigen presenting cell’s surface, to present them to circulating naïve CD4+ (helper) T cells
Some naïve helper T cells are able to successfully bind to the spike protein-MHC Class II protein complexes
Binding activates these spike-protein specific helper T cells
        Spike protein fragments are bound by MHC Class I proteins
MHC Class I proteins bring spike protein fragments to the human cell surface MHC Class I proteins present spike protein fragments to naïve CD8+ T cell
Naïve CD8+ T cells that able to bind to the spike protein-MHC Class I protein complex become activated, and travel to the lymphatic system to mature3
MHC = Major Histocompatability Complex; cell surface proteins key to immune function
CD = Cluster of Differentiation; glycoproteins on T cell surfaces that are co-receptors and facilitate T cell binding to antigens/MHC complexes. They also distinguish the types of T cells.
                 Some of these T cells mature into cytotoxic T cells that now recognize the SARS-CoV-2 spike spike protein
Cytotoxic T cells bind to human cells expressing spike protein or spike protein fragments (e.g. future COVID-19 infection)
Cytotoxic T cells release enzymes perforating infected human cells, causing cell death to occur
Immune system can now more quickly identify & destroy human cells showing signs of COVID-19 infection
Some T cell’s can mature into memory T cells (stimulated by cytokines released by helper T cells)
Memory T cells travel to lymphatic tissue, awaiting activation from exposure to spike protein in the future
More rapid cell-mediated immune response to
future SARS-CoV-2 infection (immunity)
Activated helper T cells specific to the viral spike protein secrete cytokines to stimulate immune activity
Systemic cytokine releaseàsystemic reactions like fever, chills, fatigue, myalgias (Transient)
Note: Duration of cellular/ humoral immunity is unknown
Some B cells mature into plasma cells that produce IgG antibodies against the viral spike protein
Antibodies to spike protein mark SARS-CoV-2, allowing immune system to destroy virus
Eradication of SARS-CoV-2 in extracellular compartments
Activated helper T cell interacts with naïve B cells in lymphatic tissue
Some B cells mature into memory B cells specific to SARS-CoV- 2 spike protein
                      Future exposure to spike protein re-activates memory B cell in lymphatic tissue & creates plasma cells, producing antibodies more rapidly
Rapid humoral immune response to future SARS-CoV-2 infection (immunity)3
          Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Clinical Finding
  End result
 Published February 11, 2021 on www.thecalgaryguide.com

Potassium-Sparing-Diuretics-Mechanism-of-Action-and-Side-Effects

Potassium-Sparing Diuretics: Mechanism of Action and Side Effects Potassium-Sparing Diuretics
Authors: Samin Dolatabadi, Yan Yu* Reviewers: Jessica Krahn, Timothy Fu, Juliya Hemmett* * MD at time of publication
    Epithelium Sodium Channel Blockers (Amiloride and Triamterene) Inhibit Na+ channels (involved in Na+ reabsorption) in the luminal membrane of the principal cells in the cortical collecting duct
Aldosterone Antagonists (Spironolactone and Eplerenone) Competitively blocks mineralocorticoid receptors and ↓ aldosterone effect in the renal tubules and throughout the body
    ↓ aldosterone effectà↓ expression of basolateral Na+/K+ pumps & luminal epithelium Na+ channels on the principal cells of cortical collecting duct
Spironolactone’s molecular structure is similar to that of steroid
hormonesàspironolactone can also block androgen receptors
Anti-androgenic effects
(due to ↓ androgen function in reproductive organs, skin, and on brain centers)
        Triamterene can form triamterene crystals
and granular casts (unclear mechanism)
↓ Na+ reabsorption by principal cells
↓ in serum Na+ concentration: Hyponatremia
↓ K+ pumped out into the tubule by principal cells
               Crystals & casts obstruct tubular lumen → inflammation (unclear mechanism)
Triamterene crystals are
directly cytotoxic to tubular cells
Only ~2-5% of Na+ filtered by the glomerulus is normally reabsorbed in the
cortical collecting ductàthe ↑ in Na+ retained in cortical collecting duct is mild
Water follows Na+ to maintain a balanced osmotic pressureà↑ in water in cortical collecting duct available for excretion
↑ positive charges in lumen relative to surroundings generates an electropositive tubular lumen
Unfavorable electrical gradient ↓ secretion of positive charged ions into electropositive lumen
↓ libido
Menstrual abnormalities (in women)
↓ acne Gynecomastia
(in men)
              ↓ secretion of K+
↑ in serum K+ concentration:
Hyperkalemia
Cardiac Arrythmias (See relevant slide on Hyperkalemia: Clinical Findings)
  Interstitial inflammation and tubular injury
Drug-induced Nephrotoxicity
Mild Diuresis
↓ blood volume
↓ Blood pressure/ Hypotension
↓ secretion of H+
↑ in serum H+ concentration:
Metabolic Acidosis
             Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published February 15, 2021 on www.thecalgaryguide.com

AAA-Pathogenesis

Abdominal Aortic Aneurysm (AAA): Pathogenesis
        Different parts of the aorta have different embryologic origins
Atherosclerosis
Hypertension
Age > 65
Progressive deterioration of aorta structural integrity over life span
Connective Tissue Disease
Structurally abnormal protein or protein organization in aorta
Autoimmunity
Infection (e.g Chlamydia, Mycoplasma pneumoniae, Helicobacter pylori, human cytomegalovirus, herpes simplex virus)
Antigens (substance that causes immune response) on virus or bacteria resemble local proteins in abdominal aorta
Antibodies produced in response to infection inappropriately target host cells in the aorta
Antibodies tag cells in the abdominal aorta for destruction by T-lymphocytes
Immune-mediated destruction of aorta
Smoking
Genetics
Unclear mechanisms
           Subacute (not clinically detectable) inflammation of aortic tissue
Inflammatory cytokines are released and immune cells are recruited
↑ pressure on aorta and other vessel walls
            Infiltration of vessel wall by lymphocytes and macrophages
Production of enzymes that break down elastin & collagen proteins (which provide most tensile strength to aorta)
Aorta susceptible to damage
       Degradation of aortic connective tissue
Biomechanical stress on vessels
Authors: Olivia Genereux Davis Maclean Reviewers: Jason Waechter* Amy Bromley* Yan Yu* *MD at time of publication
 The exact mechanisms are complex, debatable, and an area of intensive research – the 3 mechanisms and associated pathophysiology presented here are generally thought to be the main causes of abdominal aortic aneurysms
   Infrarenal aorta has poorly developed vaso vasorum (dedicated blood supply to vessel wall)
Infrarenal aorta relies solely on nutrient diffusion from aortic blood that crosses abdominal aorta
Infrarenal aortic wall has fewer “lamellar” units (fibromuscular units) than other regions of the aorta
Infrarenal aorta is less elastic & less able to distribute stress
Loss of smooth muscle cells & thinning of tunica media
Destruction of elastin in tunica media
Normal layers of the aortic wall
   ↓ aortic tensile strength (ability to withstand stretching) Aorta expands and dilates due to internal pressure
Tunica Intima (inner-most tissue layer of aorta)
Tunica Media (layers of elastic
tissue (elastin) and muscle fibers)
Adventitia (thin outermost collagenous layer)
(longitudinal section)
             Aortic aneurysms are usually infrarenal (85%)
Abdominal Aortic Aneurysm
   Infrarenal aorta more prone to ischemia and has impaired repair potential
Abnormal, irreversible dilation of a focal area of abdominal aorta (area of aorta between diaphragm & aortic bifurcation) to twice the diameter of adjacent normal artery segment
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Re-Published February 27, 2021 on www.thecalgaryguide.com

Cubital-Tunnel-Syndrome-Ulnar-Neuropathy

Cubital Tunnel Syndrome (Ulnar Neuropathy): Pathogenesis and clinical findings
      Osteoarthritis
Rheumatoid arthritis
Diabetes mellitus (see Calgary Guide slide on Diabetic Neuropathy)
Degenerative bone & joint changes
Autoimmune damage to joints
Abnormal bone structure/lesions
Idiopathic
Hyperglycemia causes nerve damage (complex mechanisms)
Elbow trauma
Repetitive elbow flexion
Authors: Chris Oleynick Alexandros Mouratidis Yan Yu* Reviewers: Annalise Abbott Sean Crooks Davis Maclean Hannah Koury Jeremy LaMothe* Ian Auld* * MD at time of publication
      Inflammation or edema in cubital tunnel (space between medial epicondyle of humerus and olecranon of ulna) where ulnar nerve is found
↓ size of the cubital tunnelà↑ pressure on internal contents (e.g. ulnar nerve)
             Axonal conduction is interrupted
Myelin sheath is damaged
↓ blood supply to nerve
Weakness of adductor pollicus longus (works to adduct thumb)
↑ compensatory activity of flexor
pollicis longus with pinching
↓ activity of hypothenar muscles (which move the 5th digit)
↓ activity of 5th digit palmar interosseus muscles (which adduct the 5th digit)
   Cubital Tunnel syndrome: compression neuropathy
      Paresthesia
Abnormal sensations of skin (“pins & needles”, tingling, burning, and/or numbness) in ulnar nerve sensory distribution
ulnar nerve
↓ activity of muscles innervated by ulnar nerve (medial forearm flexors, hypothenar muscles, interossei muscles, adductor pollicis longus)
+ Tinel sign
over cubital tunnel (tapping at medial elbow causes discomfort and/or paresthesia)
Weak pinch & ↓ grip strength
+ Froment’s sign
(thumb hyperflexion compensates for weak pinch)
Hypothenar atrophy
+ Wartenburg’s sign
(involuntary, excess abduction of 5th digit)
of the
Altered sensation in areas innervated by ulnar nerve (medial forearm and wrist, 5th digit, and medial half of 4th digit)
+ elbow flexion test
(flexing elbow & extending wrist further ↓ volume of the cubital tunnelà paresthesia in ulnar nerve sensory distribution
               Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
  Complications
First published January 12, 2017, re-published February 28, 2021 on www.thecalgaryguide.com

Achilles-Tendon-Rupture

Achilles Tendon Rupture: Pathogenesis and Clinical Findings
Achilles tendon ruptures are multifactorial, and may arise from any one or combination of the factors below
Authors: Alyssa Federico, Joseph Kendal Reviewers: Davis Maclean, Hannah Koury , Amanda Eslinger, Maninder Longowal, Dave Nicholl, Mehul Gupta, Gerhard Kiefer*, Yan Yu*, Jeremy LaMothe* * MD at time of publication
        Aging
Iatrogenic factors
E.g. use of fluoroquinolone antibiotics,or local or systemic corticosteroids
Exact mechanisms remain unclear
Exercise related factors
Vascular compromise
Any process that impairs blood flow to the Achilles tendon (e.g. peripheral artery disease)
Impaired ability to heal from microtrauma
Mechanical factors
Abnormal biomechanics:
bowleg, flatfoot, excessive supination, obesity, change in terrain
           Participation in vigorous exercise
Excessive heat generation
(hyperthermia) in Achilles tendon induces changes in the extracellular matrix
Intra-tendinous collagen degeneration, disorientation, and thinningàcompromised Achilles tendon integrity
Sudden shear stress and high tension in an already weakened Achilles tendon, stretching it beyond its capacity
Haglund’s deformity: enlargement of the calcaneal postero- superior tuberosity
Bony enlargement rubs on soft tissue and causes inflammation of Achilles tendon
        ↓ tendon strength from
changes in extracellular matrix
Episodic participation or sudden ↑ in exercise
Irregular use renders Achilles tendon weaker
       Microtrauma to the
and less flexible     Achilles tendon   on Achilles Tendon
Abnormal loading
   Sudden, forced dorsiflexion of foot
Eccentric contraction of the gastrocnemius-soleus complex
      The gastrocnemius muscle normally attaches to Achilles tendon to control plantar flexion of the foot
With the Achilles tendon ruptured, contraction of the gastrocnemius muscle cannot induce plantar flexion
Achilles tendon rupture
Achilles tendon innervated by the sural nerve and tibial nerve
Pain over rupture site (e.g. “like being kicked in the back of the leg”)
Antalgic gate: abnormal gait developed to avoid pain while walking
High tension at the origin and insertion points of the tendon causes the ends of the tear to separate
The Achilles tendon contains blood vessels. Rupturing the tendon also ruptures the blood vessels withinàblood leaks out into tendon rupture site
Bruising and swelling over rupture site
              ↓ resting ankle plantar flexion (Observed in prone position with knees flexed at 90 degrees)
Passive contraction (e.g. squeezing) of the gastrocnemius does not cause plantar flexion
+ Thompson (calf squeeze) test
Weak ankle plantar flexion
Lack of push-off in gait
Audible “pop” at time of injury
Palpable gap between rupture ends
              Difficulty ambulating
↓ muscle use over time
Calf atrophy
     Legend:
 Pathophysiology
Mechanism
 Sign/Symptom/Lab Finding
  Complications
 Re-Published March 21, 2021 on www.thecalgaryguide.com

Thyroïdite

Thyroïdite : Pathogénie et résultats cliniques
Authors: Jaye Platnich Reviewers: Matthew Harding Mark Elliott Hanan Bassyouni* * MD at time of publication
Hypertrophie ferme et diffuse de la glande thyroïde (douloureuse dans le cadre d'étiologies subaiguë, radique et infectieuse)
Symptômes généraux de l’hyperthyroïdie
   Post-natal
Médicaments/ Radiation
Thyroïdite bactérienne
Subaigüe (post-virale)
Abbréviations:
• TSH- Hormone stimulant de la
thyroïde
Inflammation de la glande thyroïde
Plusieurs méchanismes
Brianna Ghali Sylvain Coderre* Phillippe Couillard*
Translator:
         Infiltration de la glande thyroïde avec des globules blancs
Destruction immunitaire de la glande thyroïde
↑ libération de T4 et T3 stockés par glande thyroïde endommagée
durant 2-6 semaines (phase hyperthyroïde)
↓ sécrétion de T4 et T3 de la glande thyroïde endommagée
durant des semaines-mois (phase hypothyroïde)
Souvent une résolution à un état euthyroïde (normal), mais peut aussi rester dans un état hypothyroïde
↑ T4/T3 inhibe la
libération de la TSH par la     ↓ sécrétion TSH
               Réaction inflammatoire altère la fonction folliculaire normale (production T4/T3 et importation d’iode)
glande pituitaire
↓ Absorption de l'iode par la glande thyroïde
↓ Capture de l'iode radioactif à la scintigraphie de médecine nucléaire
       Plusieurs méchanismes     Symptômes généraux de l’hypothyroïdie
↓ T4/T3 stimule la
liberation de la TSH par la     ↑ sécrétion TSH
      Dommage altère la fonction folliculaire normale, (production T4/T3 et importation d’iode)
glande pituitaire
↓ Absorption de l'iode par la glande thyroïde
↓ Capture de l'iode radioactif à la scintigraphie de médecine nucléaire
      Légende:
 Pathophysiologie
 Méchanisme
 Signes/Symptômes/Résultats labo
 Complications
 Publié June 19, 2013 on www.thecalgaryguide.com

Fat-Embolism-Syndrome

Fat Embolism Syndrome: Pathogenesis and clinical findings
Panniculitis (conditions causing
inflammation of subcutaneous fat)
Non-trauma related (rare)
        Long bone fracture
Pelvic fracture
Orthopedic Trauma
Intraosseous access
Soft tissue injuries
Chest compressions
Bone marrow transplant
Pancreatitis
Diabetes mellitus
                   Fat from bone marrow is disrupted and leaks into bloodstream via damaged blood vessels
Fat globules obstruct dermal capillaries
Capillaries rupture
Blood leaks into the skin
Petechial rash
Non-Orthopedic Trauma (less common)
Fat from injured adipose tissue is released from adipocytes into bloodstream
Metabolic disturbance mobilizes stored fat and moves it into circulation
     Fat Embolism Syndrome
the presence of fat globules in circulation
Fat globules damage blood vessel walls
Platelets stick to damaged areas Platelet aggregation
↑ circulating free fatty acids
↑ inflammatory cytokines (TNF, IL1, IL6)
↑ serum C Reactive Protein (an acute phase reactant)
C reactive protein binds to lipid vesicles in circulation
↑ formation of lipid complexes in the blood
                      Obstruction of cerebral vasculature
↓ blood flow and oxygen delivery to brain tissue
Neurological findings: ranging from ↓ level of consciousness to seizures
Notes:
Large quantities of fat globules can obstruct pulmonary vasculature
           Blood clots form throughout the body
Disseminated intravascular coagulopathy
Back up of blood into right heart àRight ventricular dysfunction
   ↓ pulmonary arterial blood flow à↓ gas exchange in the lungs
Higher CO2 & lower O2 levels in blood àdetected by chemoreceptors
Chemoreceptors stimulate respiratory centre in the brain to ↑ rate of respiration
Dyspnea / Tachypnea
Authors: Tabitha Hawes Reviewers: Hannah Koury, Alyssa Federico, Davis MacLean, Mehul Gupta, Yan Yu*, Jeremy Lamothe* * MD at time of publication
              • Underlined findings indicate classic triad of symptoms (petechial rash, neurologic findings, dyspnea/tachypnea)
• Clinical presentation of fat embolism syndrome is variable and may present with any or all of these findings
↓ pumping of blood into systemic circulation
Hypotension Obstructive shock
      Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
  Complications
Published July 19, 2021 on www.thecalgaryguide.com

Hypertriglycéridémie primaire

Hypertriglycéridémie primaire : Pathogénie et résultats cliniques
Traduction:
Brianna Ghali Philippe Couillard* Autheur: Gillian Goobie Rédacteurs: Peter Vetere Yan Yu Hanan Bassyouni* * MD au moment de la publication
    Acronymes:
LPL = lipoprotéine lipase TG = triglycérides
CM = chylomicrons
LDL = lipoprotéine de basse densité
VLDL = lipoprotéine de très basse densité
FFA = acides gras libres CV = cardiovasculaire MC=maladie coronarienne
Déficit en lipoprotéine lipase (LPL)
La LPL présente dans le cœur,
les muscles squelettiques, les tissus adipeux et d'autres tissus est incapable d'éliminer les TG des CM et des VLDL.
Hyperlipidémie combinée familiale
Surproduction de TG-riches VLDL, LDL et/ou ApoB100
Dysbétalipoprotéinémie familiale
Mutation de ApoE (l'apoprotéine responsable de l'élimination des CMs et VLDLs de la circulation)
         VLDLs & CMs riches en TG s’accumulent dans le sang
Hypertriglycéridémie
Primaire versus Secondaire:
- L'hypertriglycéridémie primaire est due à une cause génétique. -L'hypertriglycéridémie secondaire est due à une cause médicale ou à un mode de vie acquis.
Mécanismes inconnus du risque ↑ CV, distincts du risque d'athérosclérose associé à l'hypercholestérolémie.
MC Prématurée
(âge d'apparition avant 50 ans chez les hommes et 55 ans chez les femmes)
Remarque : les signes les plus pathognomoniques de l'hypertriglycéridémie sont les xanthomes éruptifs et la lipémie rétinienne.
         La diffusion de la lumière par les TG sanguins fait paraître les vaisseaux de la rétine blanc au fond d’oeil
Lipemia Retinalis
La lipase pancréatique libère les FFA des CM et des VLDL.
La concentration plasmatique des FFA dépasse la capacité de fixation de l'albumine.
Les AGF s'accumulent dans le pancréas, provoquant une inflammationàla lyse des cellules pancréatiques et la libération d'enzymes digestives pancréatiques.
Pancréatite
(le risque de pancréatite augmente considérablement avec un taux de TG >10mmol/L)
Des niveaux excessifs de CM et de TG traversent les vaisseaux sanguins pour atteindre le derme ou la cornée.
TGs engloutis par les macrophages dermiques or cornéens (histiocytes)
Xanthomes éruptifs
(papules jaunes de 1 à 3 mm généralement observées sur les tendons et surfaces d'extension comme les coudes, les genoux, les fesses, ainsi que sur le dos, la poitrine et les extrémités proximales).
                  Légende:
 Pathophysiologie
Méchanisme
 Signe/Symptôme/Résultat Laboratoire
 Complications
 Publié 19 juin 2013 à www.thecalgaryguide.com

Dry-Eye-Syndrome-Pathogenesis

Dry Eye Syndrome (Keratoconjunctivitis sicca): Pathogenesis
The Pathophysiology of Dry eye disease is complex and an area of active investigation – The mechanism and causes presented here represented the highest yield causes and mechanism for students
   Post laser eye surgery
Disruption of corneal nerves
↓ corneal sensitivity
Damage to trigeminal nerve, the sensory innervation of the eye (due to: Herpes Zoster, tumor, trauma, etc)
Blepharitis (eyelid inflammation)
Many medications can cause dry eye via
multiple mechanisms presented here (e.g. ↓ corneal sensitivity, Meibomian gland dysfunction, lacrimal gland atrophy)
Sex Hormones (e.g. androgens & estrogens) play a complex and poorly understood role in mediating dry eye disease (net effect is that women are more often affected by dry eye)
Obstructed meibomian glands
Eyelid damage Gland atrophy
These items represent general causes of meibomian gland dysfunction – exact causes are numerous, their pathophysiology is beyond the scope of this slide
         Contact lens (long term use)
Corneal nerve adaptation to chronic mechanical stimulation
Autoimmune disease (e.g. Sjögren's syndrome)
Chronic inflammatory infiltration of the lacrimal gland (and salivary gland)
Autoimmune Lymphocytic infiltration
Inflammatory cytokine release
Autoantibody production
Cell death and apoptosis
Lacrimal gland degeneration
Meibomian gland dysfunction (Located along the eyelid margins, these glands produce meibum, an oily substance that prevents evaporation of the tear film)
↓ meibum secretion Loss of lipid layer
covering the eye, ↓ the barrier that blocks evaporation of tear film
Tear Film instability
Lifestyle
Extended reading or
TV or electronic device uses
Exposure Keratopathy (any condition causing dryness due to incomplete or inadequate eyelid closure, e.g. Bell’s Palsy)
                         ↓ activity of the afferent portion of
corneal reflex arc (responsible for reflex tearing: tearing in response to irritation of the eye)
Mechanical damage to goblet cells
secrete mucins – a substance that lubricates the eye and preserves tear film
↓ blink rate
↑ time and area
                  ↓ normal reflex tearing
for evaporation
Dry climate Wind exposure
   Infiltrative diseases
(e.g. sarcoidosis)
Lacrimal gland infiltration
↓ Lacrimal gland secretion of the the watery aqueous layer of the tear film (Aqueous deficient dry eye)
Deficient or unstable tear film (Evaporative dry eye)
↑ tear evaporation
               Direct damage to lacrimal gland (e.g. infection or trauma of the eye)
Authors: Davis Maclean, Yan Yu*, Michael Penny, O.D.
Reviewers: Natalie Arnold, Saleel Jivraj, O.D., Adam Muzychuk*, Victor Penner* *MD at time of publication
Hyperosmolar Tear Film (hyperosmolarity = ↑ solutes and ↓ solvent)
     (Further) Tear Film instability
Corneal and conjunctival epithelial
cells dry out, including goblet cells (which secrete mucins – a substance that lubricates the eye)
Inflammatory immune response àRecruitment and activation of CD4+ (Helper) T-Cells, further produce cytokines
       Further irritation and damage to ocular surface structures (cornea, conjunctiva and Meibomian glands) and lacrimal glands
See Calgary Guide: “Dry Eye Syndrome
(Kerato- conjunctivitis sicca):Clinical Findings” for signs and symptoms
 Dry Eye Syndrome (Keratoconjunctivitis sicca): A multifactorial disease of the ocular surface and tears characterized by loss of tear film homeostasis, tear film hyperosmolality and inflammation
  Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
  Complications
Published August 7, 2021 on www.thecalgaryguide.com

Dry-Eye-Syndrome-Clinical-Findings

Dry Eye Syndrome (Keratoconjunctivitis
  sicca):
Clinical Findings
If dry eye is chronic or severe (e.g Sjögren’s syndrome) corneal ulcerations or scarring may occur
Impaired visual acuity/ Blindness (does not improve with lubrication)
See Calgary Guide: “Dry Eye Syndrome (Keratoconjunctivitis sicca): Pathogenesis” for explanation of etiology and pathogenesis
Dry Eye Syndrome (Keratoconjunctivitis sicca): Disease of the ocular surface and tears characterized by loss of tear film homeostasis, tear film hyperosmolality and inflammation
     Tear film instability & hyperosmolarity
Ocular surface inflammation: Immune cell (specifically T-Cell) destruction of corneal and conjunctival epithelium
↓ quantity and/or quality of tear film
                 Inflammatory factors vasodilate conjunctival vessels, making the eye look redder in appearance
Conjunctival injection
Red eye
Corneal surface irregularities
Light rays pass through
disrupted tear film and ocular surface structures as they enter the eye
Degraded image quality as light passes through the eye
Impaired visual acuity
(improves with lubrication)
↓ lubrication over the eye surface
↑ friction on the eye during blinking or when opening eyes after sleep
May cause corneal abrasion (See Calgary Guide: Corneal Abrasion)
↑ stimulation of corneal and conjunctival nerve endings
    Irritation and damage to corneal and conjunctival cells
↑ stimulation of nerve endings
                  Chronic inflammation surrounding corneal nerves in conjunctiva and epithelium can lead to decreased neuronal firing thresholds (Neurosensory dysfunction / hyperalgesia)
↑ activity of the afferent portion of corneal reflex arc (responsible for reflex tearing: tearing in response to irritation of the eye)
Foreign body sensation
(scratching / feeling as if a foreign body, like a grain of sand, is in the eye)
      Minor (typically non-painful stimuli) may cause pain due to hypersensitive corneal nerves
Corneal neuropathic pain (pain not fully explained by ocular surface changes, often resistant to standard treatments, sometimes still present following ocular surface anaesthesia if central pathways affected)
Pain with temperature change and wind exposure
Exact mechanism unknown
Photophobia
Burning sensation Paradoxical reflex tearing (↑ tearing, which does
 not improve dry eye due to altered tear composition, poor surface coverage or overflow of temporary tearing out of the eye
    Authors: Michael Penny, O.D. Davis Maclean Yan Yu*
Reviewers: Natalie Arnold Saleel Jivraj, O.D. Adam Muzychuk* Victor Penner* *MD at time of publication
   Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
  Complications
Published August 6. 2021 on www.thecalgaryguide.com

epithelial-ovarian-cancer-pathogenesis-and-clinical-findings

Epithelial Ovarian Cancer: Pathogenesis and clinical findings
Authors: Brian Yu Chieh Cheng, Yan Yu* Reviewers: Mehul Gupta, Hannah Yaphe, Sarah Glaze* * MD at time of publication
 See Epithelial Ovarian Cancer Risk Factors slide
BRCA1 or BRCA2 mutationà faulty double strand DNA
repairà↑ mutations & loss of controlled cell division
Asymptomatic serous tubal intraepithelial carcinoma (STIC)
lesions of the fallopian tube fimbria develop overtime
STIC cells may break off from the fallopian tube & become trapped during inclusion cyst formation
As part of the ovarian cycle, the coelomic epithelium (CE) of the ovary is ruptured & repaired after ovulation
Incomplete repair of the CE results in invagination of the
rupture site, forming a benign cortical inclusion cyst
CE cells trapped during cyst formation undergoes metaplasia to tubal or other types of epithelium
Endometriosis
Endometriosis causes endometrial tissue to start growing on ovarian CE
Endometrial cells trapped during cyst formation progresses to endometrioma or “chocolate cysts”
Immune cell infiltration & cytokine release inside the
ovary results in dysregulated inflammation
Cancer cells proliferate in distant organs, invading & destroying native cells
Organ failure
          Metastatic spread to liver, lung, brain & lymph nodes
Lymphadenopathy
Systemic immune activation
↑ metabolic consumption
           ↑ capillary surface area & permeability
Fatigue & weight loss ↑ fluid entry into Ascites
peritoneal cavity
         Further somatic mutation accumulation leads to malignant transformation of epithelium
Tumor growth stimulates new blood vessel formation to supply itself with nutrients & O2
Omental / peritoneal seeding of cancer cells
     Early-stage disease
Asymptomatic
Epithelial Ovarian Cancer
In late-stage disease, tumor may secrete a glycoprotein called mucin 16, also known as CA-125 (sensitive but non-specific biomarker for ovarian cancer)
↑ serum CA-125 levels
Cancer cells proliferate inward & eventually ruptures the ovary
Cancer cells are released into the peritoneal cavity
Tumor growth in local organs (bladder/uterus) progresses to symptomatic cancer
     ↑ tumor volume & local tumor spread directly disrupts neighboring structures
Pelvic/abdominal pain
 Palpable pelvic/abdominal mass Altered urinary frequency
  Compression of
colon/bladder Change in bowel habits
   Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
Published August 15, 2021 on www.thecalgaryguide.com

COPD Acute Exacerbations

COPD Acute Exacerbations: Triggers and Signs/Symptoms
Authors: Brianne McDonald, Yan Yu* Reviewers: Nilani Sritharan, Sean Doherty, Zihong Xie (谢梓泓), Zesheng Ye (叶泽生), Yonglin Mai (麦泳琳)*, Kerri Johannson* * MD at time of publication
Notes:
• The triggers for acute exacerbations of COPD are unknown in approximately 1/3 of cases
• Changes in pulmonary function (e.g. FEV1) are poorly sensitive in the individual diagnosis of acute exacerbations of COPD due to individual variability
   Acute Bacterial Infection
Activation of proinflammatory cytokines and recruitment of neutrophils
Acute Viral Infection
Epithelial cell secretion of cytokines and ↑ airway lymphocythemia
Acute ↑ in airway inflammation
(accumulation of inflammatory cells and release of harmful mediators, such as reactive oxygen species and proteases, into the lung tissue/airways)
Air pollution
(including cigarette smoke)
↑ reactive oxygen species in lungs
            Airway inflammation ↑ secretions that accumulate in the airway lumen
Airway wall edema
Limits outflow of air from lungs on expiration
Airway bronchoconstriction in response to inflammation
Constricted airways create audible turbulent airflow on expiration
Systemic spread of inflammatory markers via the bloodstream cause inflammation throughout the body
↑ Cardiac Morbidity
↑ CRP
Worsening of respiratory symptoms
           Irritation of cough reflexes in airways
Cough
↑ Sputum production and sputum purulence
↑ Wheeze Unexpired air becomes trapped in
the lungsàDynamic Hyperinflation
  Bloodflow to lungs continue to perfuse under- ventilated regions of lungs àV/Q Mismatch
Lungs are larger àmore blood
remains in lungs à↓ preload
              Acute Respiratory Failure
Tachypnea
↑ Dyspnea
Accessory Muscle Use
  Attempts to restore normal arterial CO2 and O2 levels
 Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published April 21, 2019, updated October 6, 2021 on www.thecalgaryguide.com

Hypercortisolemia

 Hypercortisolemia (Cushing’s Syndrome): Clinical Findings
Cortisol is a net catabolic hormone affecting many body systems, serving to release energy into the blood in response to stress. Excess cortisol also impacts circulation and impairs immune function. Excess Serum Cortisol affects:
Authors: Samin Dolatabadi, Yan Yu* Reviewers: Meena Assad, Amanda Henderson, Brooke Fallis, David Campbell* * MD at time of publication
Bone and Calcium Metabolism
          Kidney and vasculature
Excess cortisol in the renal tubule saturates the enzyme 11β- HSD2, which converts cortisol to cortisone
Capacity of body to convert cortisol to cortisone is exceeded
Excess cortisol can mimic aldosterone
and bind to mineralocorticoid receptors (cortisone can’t bind to these receptors)
↑ Aldosterone effect → ↑ Na+ reabsorption from the cortical collecting duct into blood vessels
Liver and Peripheral Tissue
Cortisol ↑ gluco- neogenesis in liver, and ↑ insulin resistance by body tissue (unclear mechanisms)
Hyper- glycemia
Reproductive System
Cortisol exerts negative feedback on hypothalamus
à↓ gonadotropin releasing hormone (GnRH) secretion
↓ GnRH → ↓ LH/FSH → ↓ estrogen and testosterone production (especially important in females)
Infertility, ↓ Libido, Irregular Menses
Adipose Tissue
Cortisol ↑ fat breakdown (lipolysis)
Selective expression of cortisol receptor on different adipose tissuesàcentral, facial, dorsal fat is less broken down than in other areas (mechanism unclear)
Combined with cortisol ↑ appetite:
Skin & Connective Tissue
↑ Serum cortisol à↓ Fibroblast proliferation → ↓ Collagen synthesis
Skin atrophy with loss of connective tissue
Muscle
↑ Proteolysis & ↓ Protein synthesisà↓ muscle growth and function
Immune System
Normal serum cortisol protects against damaging effects of uncontrolled inflammatory and immune responses
↑ Serum cortisolà over-suppression of inflammation and impaired cell- mediated immunity
↑ Serum cortisol leads to ↓ Intestinal Ca2+ absorption and ↓ renal Ca2+ reabsorption
↓ Serum Ca2+ ↑ PTH secretion
↑ Serum cortisolà↑ RANKL:OPG ratio
↓ Osteoblast activity & ↑ Osteoclast activity
            Cardiac muscle
Cardio- myopathy, Heart Failure
Skeletal
muscle, especially upper arms & thighs (for unclear reasons)
Proximal Muscle Weakness
             Easy Bruising
↑ abdominal size stretches the fragile skin to become thinneràvenous blood of the underlying dermis becomes visible
Purple Striae
If hypertension is chronic
Ca2+ resorption from bone into the blood
Osteoporosis
         Supraclavicular & Dorsal Fat Pads
Central Obesity
Poor Wound Healing
Susceptibility to infection
   Round Face (Moon Face)
Abbreviations:
• RANKL – Receptor activator of nuclear factor kappa-Β ligand • OPG – Osteoprotegerin
• 11β-HSD2 – 11β-hydroxysteroid dehydrogenase type 2
   Water follows Na+ into blood vessels to balance the osmotic pressure between the blood and renal tubules
Water reabsorption → Expansion of blood volume
Hypertension
Both primary Cushing’s (e.g. adenomas that extend into zona reticularis of the adrenal cortex) and central/secondary Cushing’s (e.g. ↑ ACTH stimulation of zona reticularis) are associated with ↑ adrenal androgen secretion
        Removal of positively charged Na+ from tubular lumen creates a negative luminal environment
K+ follows the electrical gradient and is secreted into tubular lumen
↓ Serum K+ concentration
Hypokalemia
Arrhythmia, Paralysis, Cramps
(see Hypokalemia: Clinical Findings slide)
          Legend:
Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
Complications
Published October 17, 2021 on www.thecalgaryguide.com
Hirsutism, Acne

Acne Vulgaris Complications

Acne vulgaris: Complications Acne vulgaris
Cutaneous inflammation caused by acne lesions
True Scars
Authors: Stephen Williams Reviewers: Mehul Gupta, Lauren Lee, Yan Yu*, Laurie Parsons* * MD at time of publication
Elevated Acne Scars
↑ collagen exceeds collagen degradation
         Prostaglandins (PGE2), leukotrienes (LTC4, LTD4), and thromboxane A2 are released in response to inflammation
Atrophic Acne Scars
Collagen degradation and disordered deposition during healing
Formation of round-
rectangular depressions with defined edges
Box Car Scar
True scars with textural change, very slow autoresolution over time
The pigmentary changes and true scar formation resulting from acne may be more psychologically distressing than the original acne lesions
Healing processes favoring deposition of Type 1 & Type 3 collagen
Proliferation beyond the borders of original acne lesion
Keloid Formation
Healing processes favoring deposition of Type 4 collagen
Growth remains within the margins of acne lesion
Hypertrophic Scar Formation
            Dermal inflammation: Due to disruption of basal cell layer, melanin released and trapped by macrophages in the dermis.
Epidermal inflammation: ↑ melanin production and transfer of melanin to keratinocytes
Microvascular dilatation and ↑presence of erythrocytes
Erythematous macules appear where acne was present
Post- inflammatory Erythema (PIE). More common in Fitzpatrick Skin Types I-III
Formation of V- shaped tracts with sharp margins
Ice Pick Scar
Formation of a
shallow edged depression layer anchored to dermal layer and subcutis
Rolling Scar
                Pigmented
macules or patches appear where acne was present
Post-inflammatory Hyperpigmentation (PIH). More common in
Fitzpatrick Skin Types III-VI
No textural change, slow
autoresolution over time
True scars with textural change, do not resolve over time
Psychosocial Concerns (Depression, Anxiety, Social Isolation)
            Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published October 19, 2021 on www.thecalgaryguide.com

Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS)

Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS):
Authors: Lauren D. Lee, Harry C. Liu* Reviewers: Mehul Gupta, Brian Rankin, Julia Chai, Stephen Williams Yan Yu* Laurie Parsons* *MD at time of publication
Pathogenesis and clinical findings
Genetic susceptibility
Certain HLAs (e.g., HLA-B*58:01, HLA-B*57:01, and HLA-A*31:01)
HLA alleles encode for MHC structure and may influence how specific drugs/drug metabolites interact with T cell receptors and MHC proteins on antigen-presenting cells
Exposure to offending drug
E.g., Aromatic anticonvulsants (lamotrigine, carbamazepine, and phenytoin), allopurinol, and sulfonamides
Drug-specific CD4+ and CD8+ T cells produce tumor necrosis factor-alpha and interferon gamma
↑ activated T-cells 2-6 weeks after drug exposure
Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS)
Latent Viral infection
Latent viruses (HHV-6, HHV- 7, CMV, and EBV) concealed in regulatory T-cells
Reactivation of latent viruses may be contributory or secondary to T cell activation by drugs
               Eosinophilia +/- atypical lymphocytes
T helper type 2 cells recruit and activate eosinophils by releasing cytokines
Activated leukocytes create a humoral immune and allergic response
Dysfunction of regulatory T cells, resulting in failure to control unwanted immune responses against “self”
Autoimmune processes with single- or multi-organ involvement
CMV- Cytomegalovirus EBV- Epstein-Barr Virus HHV- Human Herpesvirus HLA- Human Leukocyte Antigens
MHC – Major Histo- compatibility Complex
Endocrine system
mechanism of dysfunction unclear but may be linked to autoreactive T cells
Autoimmune Type 1 thyroiditis diabetes
             Lymphadenopathy
Fever
Facial edema
Liver
lobular inflammation, dispersed foci of necrotic hepatocytes, granulomatous infiltrates consisting of eosinophils
Hepatitis
Kidney
Interstitial edema and infiltrates of lymphocytes, histiocytes, eosinophils, and plasma cells
Acute interstitial nephritis
Lung
increased pulmonary infiltrate and edema
 Morbilliform Skin Eruption
Spongiosis
Epidermal layer
Dermal- Epidermal Junction Dermal layer
Interface dermatitis
Skin eruption
(morbilliform to diffuse erythema with follicular accentuation)
Interstitial pneumonitis
Pleural effusion
              Eosinophilic infiltration
 Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published October 19, 2021 on www.thecalgaryguide.com

Vestibular Neuritis

Vestibular Neuritis: Pathogenesis and Clinical Findings
Authors: Ryan Chan Jonathan Wong Reviewers: Mehul Gupta Davis Maclean Saud Sunba Yan Yu* Euna Hwang* * MD at time of publication
   Recent viral illness or upper respiratory tract infection
Virus spreads along upper respiratory mucosa and into the inner ear structures
Vestibular Neuritis
Presumed idiopathic viral-induced inflammation of the vestibular nerve; typically unilateral
Reactivation of Herpes Simplex Virus-1 in vestibular (Scarpa’s) ganglion
 Inflammatory cell infiltration leads to degeneration and atrophy of the vestibular nerve
    Superior Vestibular Neuritis
(40-48%) Inflammatory cells traverse through only one long bony canal, easier for inflammatory cell infiltration
Loss of afferent innervation from the superior and horizontal semicircular canal (SCC), utricle, and parts of the saccule
Combined Superior Vestibular Neuritis and Inferior Vestibular Neuritis (34-56%)
Inferior Vestibular Neuritis (1.3-18%) Inflammatory cells must pass through two separate bony canals, making inflammatory cell infiltration more difficult
Loss of afferent innervation from the posterior semicircular canal (SCC) and saccule
              Utricular degeneration
Displacement of otoliths/ otoconia (commonly into the posterior SCC)
BPPV
(can occur several months after onset of neuritis)
Loss of horizontal SCC afferent neuron signaling to the brain
Unilateral Loss or ↓ of normal nystagmus response to Caloric Testing (insertion of cold and warm water/air into the ear canal while supine)
Loss of utricular afferent neuron signaling to the brain
↓ or absent Ocular Vestibular- Evoked Myogenic Potentials (VEMPs)
and Normal Cervical VEMPs
↓ unilateral vestibular input to the brain leads to acute phase symptoms (over time, brain can compensate which allows for some symptom improvement)
Loss of posterior SCC and saccular afferent neuron signaling to the brain
↓ or absent
Cervical
VEMPs, but Ocular VEMPs are normal
     ↓ or absent input to the ipsilateral vestibular nuclei elicits a vestibular nucleus response similar to contralateral SCC excitation
Acute-Phase Spontaneous Nystagmus Fast phase beats away from the affected side (3- 10 days)
And
Loss of ocular fixation on Head Impulse Test
↓ or absent saccular input to the lateral vestibular nuclei (e.g., lateral vestibulospinal tract) results in the loss of lower limb postural adjustability
Postural Instability
(e.g., abnormal Romberg/sharpened Romberg, Fukuda step test)
Bilateral mismatch of vestibular
information to the brain
Peripheral Vertigo (lasts several hours to days; rapid onset, severe, constant)
Nausea and Vomiting
Only the vestibular
part of the vestibulo- cochlear nerve is affected, not the cochlear nerve
No Hearing
Loss or Tinnitus
            Legend:
 Pathophysiology
Mechanism
 Sign/Symptom/Lab Finding
 Complications
Published October 19, 2021 on www.thecalgaryguide.com

COPD-发病机制

COPD: 发病机制
作者: Yan Yu 审稿人:Jason Baserman, Jennifer Au, Naushad Hirani*, Juri Janovcik* 译者: Zihong Xie (谢梓泓) 翻译审稿人:Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生
  /012
(如a1-抗胰蛋白酶缺乏) 阻止肺组织损伤的能力↓
+,-.
(如长期吸烟、环境污染、感染)
    肺内产生自由基
34*5
肺抗蛋白酶的失活
  ↑氧化应激,炎性细胞因子,蛋白酶功能
   支气管的持续、反复损伤
炎性细胞浸润, 杯状细胞增殖, 气道上皮纤毛 尤其中性粒细 黏液产生↑ 细胞死亡
气道弹性↓ (弹性回缩
肺实质的蛋白水解破坏↑ 维持气道开放 肺泡永久性异常
的结构支持↓ 扩张
      胞 力)
          肺气体潴留 气道狭窄与 肺过度 肺大泡
   气道黏液潴留,成为感染 狭窄 病灶
塌陷 充气
肺气肿
(容易肺泡 破裂)
气道纤维化和
    %&'()*
  慢性阻塞性肺疾病(COPD)
    临床表现 并发症 (参阅相关幻灯片) (参阅相关幻灯片)
  图注:
 病理生理
机制
 体征/临床表现/实验室检查
 并发症
 2013年1月7日发布于 www.thecalgaryguide.com
  
COPD: Clinical Findings Lung tissue
Chronic Obstructive Pulmonary Disease (COPD)
        damage
↓ elastic recoil to push air out of lungs on expiration
Lungs don’t fully empty, air is trapped in alveoli (lung hyperinflation)
↑ lung volume means diaphragm is tonically contracted (flatter)
If occurring around airways
Airflow obstruction
↑ mucus production
↓ number of epithelial ciliated cells to clear away the mucus (the cells have been killed by airway inflammation)
Chronic cough with sputum
Author: Yan Yu Reviewers: Jason Baserman Jennifer Au Naushad Hirani* Juri Janovcik* * MD at time of publication
      During expiration, positive pleural pressure squeezes on airwaysà↑ obstruction
↓ ventilation of alveoli
↓ oxygenation of blood (hypoxemia)
↓ perfusion of body tissues (i.e. brain, muscle)
Fatigue; ↓ exercise tolerance
      Total expiration time takes longer than normal
Prolonged expiration
More effort needed to ventilate larger lungs
Respiratory muscles must work harder to breathe
Turbulent airflow in narrower airways is heard on auscultation
Expiratory Wheeze
                 Diaphragm can’t flatten much further to generate deep breaths
To breathe, chest wall must expand out more
Dyspnea
Shortness of breath, especially on exertion
     Breathes are rapid & shallow
If end-stage:
Chronic fatigue causes deconditioning
Muscle weakness & wasting
  Barrel chest
If end-stage: diaphragm will be “flat”. Continued
Patient tries to expire against higher mouth air pressure, forcing airways to open wider
Pursed-lip breathing
Patient breathes with accessory muscles as well as diaphragm to try to improve airflow
    inspiratory effort further contracts diaphragmà pull the lower chest wall inwards
Hoover’s sign
(paradoxical shrinking of lower chest during inspiration)
Tripod sitting position (activates pectoral muscles)
Neck (SCM, scalene) muscles contracted
             Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published January 7, 2013 on www.thecalgaryguide.com
  
COPD: ! 45 (on ABGs) Ventilation- perfusion mismatch High A-a gradient (calculated from ABGs) Low, flat diaphragm, >10 posterior ribs (on frontal CXR) High TLC and VC (on spirometry) • • PaO2: partial pressure of O2 in arterial blood PaCO2: partial pressure of CO2 in arterial blood • In the setting of fever and productive cough, especially if lung field opacifications are seen on CXR: consider sputum gram stain and culture to rule out pneumonia. Air does not block X-ray beams, will appear black on X-ray film Chronic hypercapnia makes breathing centers less sensitive to the high PaCO2 stimulus for breathing, & more reliant on the low PaO2 stimulus (“CO2 retention”) Give O2 carefully to these patients (high PaO2 may suppress patients’ hypoxic respiratory drive, ↓ their breathing, & ↑↑↑ PaCO2) ↑ retrosternal air space (on lateral CXR) Hyper-lucent (darker) lung fields, ↓ lung markings (on frontal CXR) • Arterial Blood Gasses (ABGs) • Chest X-Ray (CXR): frontal and lateral Legend: Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications Published January 7, 2013 on www.thecalgaryguide.com COPD: !"#$ 气流阻塞 肺泡通气↓ 呼气时,胸膜腔正压挤压气 道à 阻塞↑ 作者: Yan Yu 审稿人: Jason Baserman, Jennifer Au, Naushad Hirani*, Juri Janovcik* 译者:Zihong Xie (谢梓泓) 翻译审稿人: Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生 慢性阻塞性肺疾病 (COPD) 肺组织损伤 没有弹性回缩力将 气体排出肺 肺实质与血管分布减少导 致气体交换面积↓ 弥散功能↓ (肺功能检查) 更多的CO2残留 并扩散到血液中 高碳酸血症: PaCO2 > 45 (动脉血气) 血流灌注通气不良的肺泡 时无法获得足够的氧气 总呼气时长较正常长 FEV1/FEV < 0.7 (肺功能检查) 肺无法完全排空 更多空气潴留在肺部 (肺过度充气) 低氧血症: PaO2 < 70mmHg (动脉血气) 通气-灌注不匹配 肺泡-动脉氧分压差↑ (可通过动脉血气分析计算得出) 横膈低平, 下移至第10肋后端 及以下部位 (胸部正位片) TLC与VC增大 (肺功能检查) 缩写: • • FEV1: 1秒用 • VC:肺活量 PaO2: 动脉血 力呼气量 氧分压 空气不会阻挡X射线, 在X光片上呈现为黑色 慢性高碳酸血症使呼吸中枢对PaCO2 刺激呼吸的敏感性下降 & 更依赖于低PaO2的刺激 (“二氧化碳潴留”) 给患者吸氧时需注意(高PaO2 可能会抑制患者低氧时对呼吸的 刺激,使呼吸驱动↓ & PaCO2↑↑↑ ) • FVC: 用力肺 • 活量 • TLC:肺总量 慢阻肺相关检查 : PaCO2: 动脉 血二氧化碳 分压 胸骨后间隙↑ (胸部侧位片) 肺纹理↓ • 肺功能检查 • 动脉血气分析(Arterial Blood Gasses, ABGs) • 胸部正侧位片 • 当患者发热和湿咳,特别是胸片上见肺野不清晰时: 肺透亮度↑, (胸部正位片) 考虑进行痰革兰氏染色及痰培养以排除肺炎可能 图注: 病理生理 机制 体征/临床表现/实验室检查 并发症 2013年1月7日发布于 www.thecalgaryguide.com COPD: Complications Lung inflammation Chronic Obstructive Pulmonary Disease (COPD) Airway obstruction ↓ inhaled air in alveoli and terminal bronchioles Rupture of emphasematous bullae on surface of lung Inhaled air leaks into pleural cavity and is trapped there Pneumothorax Feeling a loss of control over one’s life, and hopelessness for the future Goblet cell proliferation, ↑ mucus production Death of airway epithelium ciliated cells ↓ oxygenation of the blood passing through the lungs Chronic hypoxemia Kidneys compensate by ↑ erythropoietin (EPO) production ↑ Hemoglobin and red blood cell synthesis Polycythemia (secondary) Hypoxic alveoli cause the pulmonary arterioles perfusing them to reflexively vasoconstrict Since most alveoli in the lungs are hypoxic, hypoxic vasoconstriction occurs across entire lung Vasoconstriction ↑ blood pressure within lung vasculature Pulmonary hypertension ↑ workload of the right ventricle (to pump against higher pressures) To compensate, the right ventricle progressively hypertrophies and dilates, but over time its output ↓ Cor pulmonale (Right heart failure in isolation, not due to Left heart failure) Mucus trapped in airways, serve as nidus for infection Acute exacerbation of COPD (AECOPD) Pneumonia The chronic, systemic inflammation in COPD is a hyper-metabolic state that consumes calories Macro-nutrient deficiency Trouble with respiration lead to inactivity and deconditioning Wasting, muscle atrophy More inactivity and deconditioning perpetuates the cycle Depression Author: Yan Yu Reviewers: Jason Baserman Naushad Hirani* Juri Janovcik* * MD at time of publication Legend: Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications Published January 7, 2013 on www.thecalgaryguide.com COPD: !"# 肺部炎症 杯状细胞增殖, 气道上皮纤毛 粘液产生↑ 细胞死亡 黏液潴留呼吸道,成为感 染的病灶 慢性阻塞性肺疾病 (COPD) 气道阻塞à 吸入肺泡和终末细 肺大疱破裂 吸入的空气渗入 并潴留于胸腔 气胸 感觉生活失控,对未 来感到绝望 抑郁 作者: Yan Yu 审稿人: Jason Baserman, Naushad Hirani*, Juri Janovcik* 译者: Zihong Xie (谢梓泓) 翻译审稿人: Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生 支气管的空气 ↓ 流经肺的血液进行气 缺氧的肺泡à灌注肺泡的肺小动 慢性阻塞性肺疾 病急性加重期 (AECOPD) 肺炎 体交换↓ 慢性低氧血症 肾脏合成促红细胞 生成素进行代偿↑ 血红蛋白与红 细胞合成↑ 红细胞增多症 (继发性) 脉发生反射性血管收缩 肺大部分肺泡缺氧à整个肺 都出现缺氧性血管收缩 肺血管收缩 à 肺血管压力↑ 肺动脉高压 ↑ 右心室负荷(泵血时对抗高压) 为了代偿,右心室逐渐肥大和扩张, 但随着病程进展,右心室输出量 ↓ 肺心病 (单独出现右心衰竭,非左心衰) COPD所致的慢性全身 呼吸困难导致活 性炎症会使机体处于高 动量减少和活动 代谢状态,消耗能量 耐量降低 宏量营养 素缺乏症 消瘦,肌肉萎缩 运动量下降和活动耐量 的降低造成恶性循环 图注: 病理生理 机制 体征/临床表现/实验室检查 并发症 2013年1月7日发布于 www.thecalgaryguide.com " title="COPD: 发病机制 作者: Yan Yu 审稿人:Jason Baserman, Jennifer Au, Naushad Hirani*, Juri Janovcik* 译者: Zihong Xie (谢梓泓) 翻译审稿人:Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生 /012 (如a1-抗胰蛋白酶缺乏) 阻止肺组织损伤的能力↓ +,-. (如长期吸烟、环境污染、感染) 肺内产生自由基 34*5 肺抗蛋白酶的失活 ↑氧化应激,炎性细胞因子,蛋白酶功能 支气管的持续、反复损伤 炎性细胞浸润, 杯状细胞增殖, 气道上皮纤毛 尤其中性粒细 黏液产生↑ 细胞死亡 气道弹性↓ (弹性回缩 肺实质的蛋白水解破坏↑ 维持气道开放 肺泡永久性异常 的结构支持↓ 扩张 胞 力) 肺气体潴留 气道狭窄与 肺过度 肺大泡 气道黏液潴留,成为感染 狭窄 病灶 塌陷 充气 肺气肿 (容易肺泡 破裂) 气道纤维化和 %&'()* 慢性阻塞性肺疾病(COPD) 临床表现 并发症 (参阅相关幻灯片) (参阅相关幻灯片) 图注: 病理生理 机制 体征/临床表现/实验室检查 并发症 2013年1月7日发布于 www.thecalgaryguide.com COPD: Clinical Findings Lung tissue Chronic Obstructive Pulmonary Disease (COPD) damage ↓ elastic recoil to push air out of lungs on expiration Lungs don’t fully empty, air is trapped in alveoli (lung hyperinflation) ↑ lung volume means diaphragm is tonically contracted (flatter) If occurring around airways Airflow obstruction ↑ mucus production ↓ number of epithelial ciliated cells to clear away the mucus (the cells have been killed by airway inflammation) Chronic cough with sputum Author: Yan Yu Reviewers: Jason Baserman Jennifer Au Naushad Hirani* Juri Janovcik* * MD at time of publication During expiration, positive pleural pressure squeezes on airwaysà↑ obstruction ↓ ventilation of alveoli ↓ oxygenation of blood (hypoxemia) ↓ perfusion of body tissues (i.e. brain, muscle) Fatigue; ↓ exercise tolerance Total expiration time takes longer than normal Prolonged expiration More effort needed to ventilate larger lungs Respiratory muscles must work harder to breathe Turbulent airflow in narrower airways is heard on auscultation Expiratory Wheeze Diaphragm can’t flatten much further to generate deep breaths To breathe, chest wall must expand out more Dyspnea Shortness of breath, especially on exertion Breathes are rapid & shallow If end-stage: Chronic fatigue causes deconditioning Muscle weakness & wasting Barrel chest If end-stage: diaphragm will be “flat”. Continued Patient tries to expire against higher mouth air pressure, forcing airways to open wider Pursed-lip breathing Patient breathes with accessory muscles as well as diaphragm to try to improve airflow inspiratory effort further contracts diaphragmà pull the lower chest wall inwards Hoover’s sign (paradoxical shrinking of lower chest during inspiration) Tripod sitting position (activates pectoral muscles) Neck (SCM, scalene) muscles contracted Legend: Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications Published January 7, 2013 on www.thecalgaryguide.com COPD: !"#$ 慢性阻塞性肺疾病 (COPD) 如果出现在气道周围 气流阻塞 肺不能完全排空 气体,气体潴留 于肺泡(肺过度 充气) 总呼气时长大于 正常时长 呼气相延长 肺组织损伤 呼气时,将空气排出肺外 的弹性回缩力↓ 肺不能完全排空气体, 气体潴留于肺泡内 (肺过度充气) 肺容积↑,膈肌紧张 性收缩(膈肌平坦) 呼气时,胸膜腔正压挤压气道 à 气道阻塞↑ 肺泡通气↓ 血液氧合↓ (低 氧血症) 身体组织灌注 量↓ (比如脑、 肌肉) 疲劳; 运动耐量↓ 黏液生成↑ 清除黏液的上皮纤 毛细胞数量↓ (受 气道炎症损伤) 慢性咳嗽伴咳 痰 作者: Yan Yu 审稿人: Jason Baserman, Jennifer Au, Naushad Hirani*, Juri Janovcik* 译者: Zihong Xie (谢梓泓) 翻译审稿人: Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生 容积较大 的肺需要 更加努力 才能通气 呼吸肌必须 更用力才能 呼吸 听诊闻及狭窄气 道中的湍流气流 呼气喘鸣音 呼吸困难 气促,尤其是劳累 膈肌无法进一步收缩以 产生深呼吸 呼吸浅快 为了呼吸, 胸壁必须延 展得更大 桶状胸 晚期病人: 患者试图在较高的口 慢性疲劳导致 患者动用辅助呼吸肌和膈肌呼吸, 腔内气压下进行呼气, 活动耐量下降 从而使气道更开放 以改善气流 晚期病人:膈肌 “平坦” ,持续吸气进一步压 缩膈肌à 向内拉季肋部胸壁 胡佛征 (吸气时,胸廓下侧季肋部内收) 缩唇呼吸 肌肉无力 & 消瘦 端坐呼吸 (调动胸肌) 颈部肌肉收 缩(胸锁乳 突肌、斜角 肌) 图注: 病理生理 机制 体征/临床表现/实验室检查 并发症 2013年1月7日发布于 www.thecalgaryguide.com COPD: Findings on Investigations Chronic Obstructive Pulmonary Disease (COPD) Author: Yan Yu Reviewers: Jason Baserman Jennifer Au Naushad Hirani* Juri Janovcik* * MD at time of publication Airflow obstruction Lung tissue damage ↓ ventilation of alveoli Blood perfusing ill- ventilated alveoli does not receive normal amounts of oxygen During expiration, positive pleural pressure squeezes on airwaysà↑ obstruction) No elastic recoil to push air out of lungs Loss of lung parenchyma and vasculature ↓ surface area for gas exchange ↓ diffusion capacity (on spirometry) Hypoxemia: PaO2 < 70mmHg (on ABGs) Abbreviations: • FEV1: Forced expiratory volume in 1 second • FVC: Forced vital capacity • TLC: Total lung capacity • VC: Vital Capacity Investigations for COPD : • Spirometry (Pulmonary function test) Total expiration time takes longer than normal FEV1/FEV < 0.7 (on spirometry) Lungs don’t fully empty More air trapped within lungs (hyperinflation) More CO2 remains and diffuses into the blood Hypercapnia: PaCO2 > 45 (on ABGs) Ventilation- perfusion mismatch High A-a gradient (calculated from ABGs) Low, flat diaphragm, >10 posterior ribs (on frontal CXR) High TLC and VC (on spirometry) • • PaO2: partial pressure of O2 in arterial blood PaCO2: partial pressure of CO2 in arterial blood • In the setting of fever and productive cough, especially if lung field opacifications are seen on CXR: consider sputum gram stain and culture to rule out pneumonia. Air does not block X-ray beams, will appear black on X-ray film Chronic hypercapnia makes breathing centers less sensitive to the high PaCO2 stimulus for breathing, & more reliant on the low PaO2 stimulus (“CO2 retention”) Give O2 carefully to these patients (high PaO2 may suppress patients’ hypoxic respiratory drive, ↓ their breathing, & ↑↑↑ PaCO2) ↑ retrosternal air space (on lateral CXR) Hyper-lucent (darker) lung fields, ↓ lung markings (on frontal CXR) • Arterial Blood Gasses (ABGs) • Chest X-Ray (CXR): frontal and lateral Legend: Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications Published January 7, 2013 on www.thecalgaryguide.com COPD: !"#$ 气流阻塞 肺泡通气↓ 呼气时,胸膜腔正压挤压气 道à 阻塞↑ 作者: Yan Yu 审稿人: Jason Baserman, Jennifer Au, Naushad Hirani*, Juri Janovcik* 译者:Zihong Xie (谢梓泓) 翻译审稿人: Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生 慢性阻塞性肺疾病 (COPD) 肺组织损伤 没有弹性回缩力将 气体排出肺 肺实质与血管分布减少导 致气体交换面积↓ 弥散功能↓ (肺功能检查) 更多的CO2残留 并扩散到血液中 高碳酸血症: PaCO2 > 45 (动脉血气) 血流灌注通气不良的肺泡 时无法获得足够的氧气 总呼气时长较正常长 FEV1/FEV < 0.7 (肺功能检查) 肺无法完全排空 更多空气潴留在肺部 (肺过度充气) 低氧血症: PaO2 < 70mmHg (动脉血气) 通气-灌注不匹配 肺泡-动脉氧分压差↑ (可通过动脉血气分析计算得出) 横膈低平, 下移至第10肋后端 及以下部位 (胸部正位片) TLC与VC增大 (肺功能检查) 缩写: • • FEV1: 1秒用 • VC:肺活量 PaO2: 动脉血 力呼气量 氧分压 空气不会阻挡X射线, 在X光片上呈现为黑色 慢性高碳酸血症使呼吸中枢对PaCO2 刺激呼吸的敏感性下降 & 更依赖于低PaO2的刺激 (“二氧化碳潴留”) 给患者吸氧时需注意(高PaO2 可能会抑制患者低氧时对呼吸的 刺激,使呼吸驱动↓ & PaCO2↑↑↑ ) • FVC: 用力肺 • 活量 • TLC:肺总量 慢阻肺相关检查 : PaCO2: 动脉 血二氧化碳 分压 胸骨后间隙↑ (胸部侧位片) 肺纹理↓ • 肺功能检查 • 动脉血气分析(Arterial Blood Gasses, ABGs) • 胸部正侧位片 • 当患者发热和湿咳,特别是胸片上见肺野不清晰时: 肺透亮度↑, (胸部正位片) 考虑进行痰革兰氏染色及痰培养以排除肺炎可能 图注: 病理生理 机制 体征/临床表现/实验室检查 并发症 2013年1月7日发布于 www.thecalgaryguide.com COPD: Complications Lung inflammation Chronic Obstructive Pulmonary Disease (COPD) Airway obstruction ↓ inhaled air in alveoli and terminal bronchioles Rupture of emphasematous bullae on surface of lung Inhaled air leaks into pleural cavity and is trapped there Pneumothorax Feeling a loss of control over one’s life, and hopelessness for the future Goblet cell proliferation, ↑ mucus production Death of airway epithelium ciliated cells ↓ oxygenation of the blood passing through the lungs Chronic hypoxemia Kidneys compensate by ↑ erythropoietin (EPO) production ↑ Hemoglobin and red blood cell synthesis Polycythemia (secondary) Hypoxic alveoli cause the pulmonary arterioles perfusing them to reflexively vasoconstrict Since most alveoli in the lungs are hypoxic, hypoxic vasoconstriction occurs across entire lung Vasoconstriction ↑ blood pressure within lung vasculature Pulmonary hypertension ↑ workload of the right ventricle (to pump against higher pressures) To compensate, the right ventricle progressively hypertrophies and dilates, but over time its output ↓ Cor pulmonale (Right heart failure in isolation, not due to Left heart failure) Mucus trapped in airways, serve as nidus for infection Acute exacerbation of COPD (AECOPD) Pneumonia The chronic, systemic inflammation in COPD is a hyper-metabolic state that consumes calories Macro-nutrient deficiency Trouble with respiration lead to inactivity and deconditioning Wasting, muscle atrophy More inactivity and deconditioning perpetuates the cycle Depression Author: Yan Yu Reviewers: Jason Baserman Naushad Hirani* Juri Janovcik* * MD at time of publication Legend: Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications Published January 7, 2013 on www.thecalgaryguide.com COPD: !"# 肺部炎症 杯状细胞增殖, 气道上皮纤毛 粘液产生↑ 细胞死亡 黏液潴留呼吸道,成为感 染的病灶 慢性阻塞性肺疾病 (COPD) 气道阻塞à 吸入肺泡和终末细 肺大疱破裂 吸入的空气渗入 并潴留于胸腔 气胸 感觉生活失控,对未 来感到绝望 抑郁 作者: Yan Yu 审稿人: Jason Baserman, Naushad Hirani*, Juri Janovcik* 译者: Zihong Xie (谢梓泓) 翻译审稿人: Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生 支气管的空气 ↓ 流经肺的血液进行气 缺氧的肺泡à灌注肺泡的肺小动 慢性阻塞性肺疾 病急性加重期 (AECOPD) 肺炎 体交换↓ 慢性低氧血症 肾脏合成促红细胞 生成素进行代偿↑ 血红蛋白与红 细胞合成↑ 红细胞增多症 (继发性) 脉发生反射性血管收缩 肺大部分肺泡缺氧à整个肺 都出现缺氧性血管收缩 肺血管收缩 à 肺血管压力↑ 肺动脉高压 ↑ 右心室负荷(泵血时对抗高压) 为了代偿,右心室逐渐肥大和扩张, 但随着病程进展,右心室输出量 ↓ 肺心病 (单独出现右心衰竭,非左心衰) COPD所致的慢性全身 呼吸困难导致活 性炎症会使机体处于高 动量减少和活动 代谢状态,消耗能量 耐量降低 宏量营养 素缺乏症 消瘦,肌肉萎缩 运动量下降和活动耐量 的降低造成恶性循环 图注: 病理生理 机制 体征/临床表现/实验室检查 并发症 2013年1月7日发布于 www.thecalgaryguide.com " />

COPD-临床表现

COPD: 临床表现
作者: Yan Yu 审稿人:Jason Baserman, Jennifer Au, Naushad Hirani*, Juri Janovcik* 译者: Zihong Xie (谢梓泓) 翻译审稿人:Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生
  /012
(如a1-抗胰蛋白酶缺乏) 阻止肺组织损伤的能力↓
+,-.
(如长期吸烟、环境污染、感染)
    肺内产生自由基
34*5
肺抗蛋白酶的失活
  ↑氧化应激,炎性细胞因子,蛋白酶功能
   支气管的持续、反复损伤
炎性细胞浸润, 杯状细胞增殖, 气道上皮纤毛 尤其中性粒细 黏液产生↑ 细胞死亡
气道弹性↓ (弹性回缩
肺实质的蛋白水解破坏↑ 维持气道开放 肺泡永久性异常
的结构支持↓ 扩张
      胞 力)
          肺气体潴留 气道狭窄与 肺过度 肺大泡
   气道黏液潴留,成为感染 狭窄 病灶
塌陷 充气
肺气肿
(容易肺泡 破裂)
气道纤维化和
    %&'()*
  慢性阻塞性肺疾病(COPD)
    临床表现 并发症 (参阅相关幻灯片) (参阅相关幻灯片)
  图注:
 病理生理
机制
 体征/临床表现/实验室检查
 并发症
 2013年1月7日发布于 www.thecalgaryguide.com
  
COPD: Clinical Findings Lung tissue
Chronic Obstructive Pulmonary Disease (COPD)
        damage
↓ elastic recoil to push air out of lungs on expiration
Lungs don’t fully empty, air is trapped in alveoli (lung hyperinflation)
↑ lung volume means diaphragm is tonically contracted (flatter)
If occurring around airways
Airflow obstruction
↑ mucus production
↓ number of epithelial ciliated cells to clear away the mucus (the cells have been killed by airway inflammation)
Chronic cough with sputum
Author: Yan Yu Reviewers: Jason Baserman Jennifer Au Naushad Hirani* Juri Janovcik* * MD at time of publication
      During expiration, positive pleural pressure squeezes on airwaysà↑ obstruction
↓ ventilation of alveoli
↓ oxygenation of blood (hypoxemia)
↓ perfusion of body tissues (i.e. brain, muscle)
Fatigue; ↓ exercise tolerance
      Total expiration time takes longer than normal
Prolonged expiration
More effort needed to ventilate larger lungs
Respiratory muscles must work harder to breathe
Turbulent airflow in narrower airways is heard on auscultation
Expiratory Wheeze
                 Diaphragm can’t flatten much further to generate deep breaths
To breathe, chest wall must expand out more
Dyspnea
Shortness of breath, especially on exertion
     Breathes are rapid & shallow
If end-stage:
Chronic fatigue causes deconditioning
Muscle weakness & wasting
  Barrel chest
If end-stage: diaphragm will be “flat”. Continued
Patient tries to expire against higher mouth air pressure, forcing airways to open wider
Pursed-lip breathing
Patient breathes with accessory muscles as well as diaphragm to try to improve airflow
    inspiratory effort further contracts diaphragmà pull the lower chest wall inwards
Hoover’s sign
(paradoxical shrinking of lower chest during inspiration)
Tripod sitting position (activates pectoral muscles)
Neck (SCM, scalene) muscles contracted
             Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published January 7, 2013 on www.thecalgaryguide.com
  
COPD: ! 45 (on ABGs) Ventilation- perfusion mismatch High A-a gradient (calculated from ABGs) Low, flat diaphragm, >10 posterior ribs (on frontal CXR) High TLC and VC (on spirometry) • • PaO2: partial pressure of O2 in arterial blood PaCO2: partial pressure of CO2 in arterial blood • In the setting of fever and productive cough, especially if lung field opacifications are seen on CXR: consider sputum gram stain and culture to rule out pneumonia. Air does not block X-ray beams, will appear black on X-ray film Chronic hypercapnia makes breathing centers less sensitive to the high PaCO2 stimulus for breathing, & more reliant on the low PaO2 stimulus (“CO2 retention”) Give O2 carefully to these patients (high PaO2 may suppress patients’ hypoxic respiratory drive, ↓ their breathing, & ↑↑↑ PaCO2) ↑ retrosternal air space (on lateral CXR) Hyper-lucent (darker) lung fields, ↓ lung markings (on frontal CXR) • Arterial Blood Gasses (ABGs) • Chest X-Ray (CXR): frontal and lateral Legend: Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications Published January 7, 2013 on www.thecalgaryguide.com COPD: !"#$ 气流阻塞 肺泡通气↓ 呼气时,胸膜腔正压挤压气 道à 阻塞↑ 作者: Yan Yu 审稿人: Jason Baserman, Jennifer Au, Naushad Hirani*, Juri Janovcik* 译者:Zihong Xie (谢梓泓) 翻译审稿人: Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生 慢性阻塞性肺疾病 (COPD) 肺组织损伤 没有弹性回缩力将 气体排出肺 肺实质与血管分布减少导 致气体交换面积↓ 弥散功能↓ (肺功能检查) 更多的CO2残留 并扩散到血液中 高碳酸血症: PaCO2 > 45 (动脉血气) 血流灌注通气不良的肺泡 时无法获得足够的氧气 总呼气时长较正常长 FEV1/FEV < 0.7 (肺功能检查) 肺无法完全排空 更多空气潴留在肺部 (肺过度充气) 低氧血症: PaO2 < 70mmHg (动脉血气) 通气-灌注不匹配 肺泡-动脉氧分压差↑ (可通过动脉血气分析计算得出) 横膈低平, 下移至第10肋后端 及以下部位 (胸部正位片) TLC与VC增大 (肺功能检查) 缩写: • • FEV1: 1秒用 • VC:肺活量 PaO2: 动脉血 力呼气量 氧分压 空气不会阻挡X射线, 在X光片上呈现为黑色 慢性高碳酸血症使呼吸中枢对PaCO2 刺激呼吸的敏感性下降 & 更依赖于低PaO2的刺激 (“二氧化碳潴留”) 给患者吸氧时需注意(高PaO2 可能会抑制患者低氧时对呼吸的 刺激,使呼吸驱动↓ & PaCO2↑↑↑ ) • FVC: 用力肺 • 活量 • TLC:肺总量 慢阻肺相关检查 : PaCO2: 动脉 血二氧化碳 分压 胸骨后间隙↑ (胸部侧位片) 肺纹理↓ • 肺功能检查 • 动脉血气分析(Arterial Blood Gasses, ABGs) • 胸部正侧位片 • 当患者发热和湿咳,特别是胸片上见肺野不清晰时: 肺透亮度↑, (胸部正位片) 考虑进行痰革兰氏染色及痰培养以排除肺炎可能 图注: 病理生理 机制 体征/临床表现/实验室检查 并发症 2013年1月7日发布于 www.thecalgaryguide.com COPD: Complications Lung inflammation Chronic Obstructive Pulmonary Disease (COPD) Airway obstruction ↓ inhaled air in alveoli and terminal bronchioles Rupture of emphasematous bullae on surface of lung Inhaled air leaks into pleural cavity and is trapped there Pneumothorax Feeling a loss of control over one’s life, and hopelessness for the future Goblet cell proliferation, ↑ mucus production Death of airway epithelium ciliated cells ↓ oxygenation of the blood passing through the lungs Chronic hypoxemia Kidneys compensate by ↑ erythropoietin (EPO) production ↑ Hemoglobin and red blood cell synthesis Polycythemia (secondary) Hypoxic alveoli cause the pulmonary arterioles perfusing them to reflexively vasoconstrict Since most alveoli in the lungs are hypoxic, hypoxic vasoconstriction occurs across entire lung Vasoconstriction ↑ blood pressure within lung vasculature Pulmonary hypertension ↑ workload of the right ventricle (to pump against higher pressures) To compensate, the right ventricle progressively hypertrophies and dilates, but over time its output ↓ Cor pulmonale (Right heart failure in isolation, not due to Left heart failure) Mucus trapped in airways, serve as nidus for infection Acute exacerbation of COPD (AECOPD) Pneumonia The chronic, systemic inflammation in COPD is a hyper-metabolic state that consumes calories Macro-nutrient deficiency Trouble with respiration lead to inactivity and deconditioning Wasting, muscle atrophy More inactivity and deconditioning perpetuates the cycle Depression Author: Yan Yu Reviewers: Jason Baserman Naushad Hirani* Juri Janovcik* * MD at time of publication Legend: Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications Published January 7, 2013 on www.thecalgaryguide.com COPD: !"# 肺部炎症 杯状细胞增殖, 气道上皮纤毛 粘液产生↑ 细胞死亡 黏液潴留呼吸道,成为感 染的病灶 慢性阻塞性肺疾病 (COPD) 气道阻塞à 吸入肺泡和终末细 肺大疱破裂 吸入的空气渗入 并潴留于胸腔 气胸 感觉生活失控,对未 来感到绝望 抑郁 作者: Yan Yu 审稿人: Jason Baserman, Naushad Hirani*, Juri Janovcik* 译者: Zihong Xie (谢梓泓) 翻译审稿人: Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生 支气管的空气 ↓ 流经肺的血液进行气 缺氧的肺泡à灌注肺泡的肺小动 慢性阻塞性肺疾 病急性加重期 (AECOPD) 肺炎 体交换↓ 慢性低氧血症 肾脏合成促红细胞 生成素进行代偿↑ 血红蛋白与红 细胞合成↑ 红细胞增多症 (继发性) 脉发生反射性血管收缩 肺大部分肺泡缺氧à整个肺 都出现缺氧性血管收缩 肺血管收缩 à 肺血管压力↑ 肺动脉高压 ↑ 右心室负荷(泵血时对抗高压) 为了代偿,右心室逐渐肥大和扩张, 但随着病程进展,右心室输出量 ↓ 肺心病 (单独出现右心衰竭,非左心衰) COPD所致的慢性全身 呼吸困难导致活 性炎症会使机体处于高 动量减少和活动 代谢状态,消耗能量 耐量降低 宏量营养 素缺乏症 消瘦,肌肉萎缩 运动量下降和活动耐量 的降低造成恶性循环 图注: 病理生理 机制 体征/临床表现/实验室检查 并发症 2013年1月7日发布于 www.thecalgaryguide.com " title="COPD: 临床表现 作者: Yan Yu 审稿人:Jason Baserman, Jennifer Au, Naushad Hirani*, Juri Janovcik* 译者: Zihong Xie (谢梓泓) 翻译审稿人:Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生 /012 (如a1-抗胰蛋白酶缺乏) 阻止肺组织损伤的能力↓ +,-. (如长期吸烟、环境污染、感染) 肺内产生自由基 34*5 肺抗蛋白酶的失活 ↑氧化应激,炎性细胞因子,蛋白酶功能 支气管的持续、反复损伤 炎性细胞浸润, 杯状细胞增殖, 气道上皮纤毛 尤其中性粒细 黏液产生↑ 细胞死亡 气道弹性↓ (弹性回缩 肺实质的蛋白水解破坏↑ 维持气道开放 肺泡永久性异常 的结构支持↓ 扩张 胞 力) 肺气体潴留 气道狭窄与 肺过度 肺大泡 气道黏液潴留,成为感染 狭窄 病灶 塌陷 充气 肺气肿 (容易肺泡 破裂) 气道纤维化和 %&'()* 慢性阻塞性肺疾病(COPD) 临床表现 并发症 (参阅相关幻灯片) (参阅相关幻灯片) 图注: 病理生理 机制 体征/临床表现/实验室检查 并发症 2013年1月7日发布于 www.thecalgaryguide.com COPD: Clinical Findings Lung tissue Chronic Obstructive Pulmonary Disease (COPD) damage ↓ elastic recoil to push air out of lungs on expiration Lungs don’t fully empty, air is trapped in alveoli (lung hyperinflation) ↑ lung volume means diaphragm is tonically contracted (flatter) If occurring around airways Airflow obstruction ↑ mucus production ↓ number of epithelial ciliated cells to clear away the mucus (the cells have been killed by airway inflammation) Chronic cough with sputum Author: Yan Yu Reviewers: Jason Baserman Jennifer Au Naushad Hirani* Juri Janovcik* * MD at time of publication During expiration, positive pleural pressure squeezes on airwaysà↑ obstruction ↓ ventilation of alveoli ↓ oxygenation of blood (hypoxemia) ↓ perfusion of body tissues (i.e. brain, muscle) Fatigue; ↓ exercise tolerance Total expiration time takes longer than normal Prolonged expiration More effort needed to ventilate larger lungs Respiratory muscles must work harder to breathe Turbulent airflow in narrower airways is heard on auscultation Expiratory Wheeze Diaphragm can’t flatten much further to generate deep breaths To breathe, chest wall must expand out more Dyspnea Shortness of breath, especially on exertion Breathes are rapid & shallow If end-stage: Chronic fatigue causes deconditioning Muscle weakness & wasting Barrel chest If end-stage: diaphragm will be “flat”. Continued Patient tries to expire against higher mouth air pressure, forcing airways to open wider Pursed-lip breathing Patient breathes with accessory muscles as well as diaphragm to try to improve airflow inspiratory effort further contracts diaphragmà pull the lower chest wall inwards Hoover’s sign (paradoxical shrinking of lower chest during inspiration) Tripod sitting position (activates pectoral muscles) Neck (SCM, scalene) muscles contracted Legend: Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications Published January 7, 2013 on www.thecalgaryguide.com COPD: !"#$ 慢性阻塞性肺疾病 (COPD) 如果出现在气道周围 气流阻塞 肺不能完全排空 气体,气体潴留 于肺泡(肺过度 充气) 总呼气时长大于 正常时长 呼气相延长 肺组织损伤 呼气时,将空气排出肺外 的弹性回缩力↓ 肺不能完全排空气体, 气体潴留于肺泡内 (肺过度充气) 肺容积↑,膈肌紧张 性收缩(膈肌平坦) 呼气时,胸膜腔正压挤压气道 à 气道阻塞↑ 肺泡通气↓ 血液氧合↓ (低 氧血症) 身体组织灌注 量↓ (比如脑、 肌肉) 疲劳; 运动耐量↓ 黏液生成↑ 清除黏液的上皮纤 毛细胞数量↓ (受 气道炎症损伤) 慢性咳嗽伴咳 痰 作者: Yan Yu 审稿人: Jason Baserman, Jennifer Au, Naushad Hirani*, Juri Janovcik* 译者: Zihong Xie (谢梓泓) 翻译审稿人: Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生 容积较大 的肺需要 更加努力 才能通气 呼吸肌必须 更用力才能 呼吸 听诊闻及狭窄气 道中的湍流气流 呼气喘鸣音 呼吸困难 气促,尤其是劳累 膈肌无法进一步收缩以 产生深呼吸 呼吸浅快 为了呼吸, 胸壁必须延 展得更大 桶状胸 晚期病人: 患者试图在较高的口 慢性疲劳导致 患者动用辅助呼吸肌和膈肌呼吸, 腔内气压下进行呼气, 活动耐量下降 从而使气道更开放 以改善气流 晚期病人:膈肌 “平坦” ,持续吸气进一步压 缩膈肌à 向内拉季肋部胸壁 胡佛征 (吸气时,胸廓下侧季肋部内收) 缩唇呼吸 肌肉无力 & 消瘦 端坐呼吸 (调动胸肌) 颈部肌肉收 缩(胸锁乳 突肌、斜角 肌) 图注: 病理生理 机制 体征/临床表现/实验室检查 并发症 2013年1月7日发布于 www.thecalgaryguide.com COPD: Findings on Investigations Chronic Obstructive Pulmonary Disease (COPD) Author: Yan Yu Reviewers: Jason Baserman Jennifer Au Naushad Hirani* Juri Janovcik* * MD at time of publication Airflow obstruction Lung tissue damage ↓ ventilation of alveoli Blood perfusing ill- ventilated alveoli does not receive normal amounts of oxygen During expiration, positive pleural pressure squeezes on airwaysà↑ obstruction) No elastic recoil to push air out of lungs Loss of lung parenchyma and vasculature ↓ surface area for gas exchange ↓ diffusion capacity (on spirometry) Hypoxemia: PaO2 < 70mmHg (on ABGs) Abbreviations: • FEV1: Forced expiratory volume in 1 second • FVC: Forced vital capacity • TLC: Total lung capacity • VC: Vital Capacity Investigations for COPD : • Spirometry (Pulmonary function test) Total expiration time takes longer than normal FEV1/FEV < 0.7 (on spirometry) Lungs don’t fully empty More air trapped within lungs (hyperinflation) More CO2 remains and diffuses into the blood Hypercapnia: PaCO2 > 45 (on ABGs) Ventilation- perfusion mismatch High A-a gradient (calculated from ABGs) Low, flat diaphragm, >10 posterior ribs (on frontal CXR) High TLC and VC (on spirometry) • • PaO2: partial pressure of O2 in arterial blood PaCO2: partial pressure of CO2 in arterial blood • In the setting of fever and productive cough, especially if lung field opacifications are seen on CXR: consider sputum gram stain and culture to rule out pneumonia. Air does not block X-ray beams, will appear black on X-ray film Chronic hypercapnia makes breathing centers less sensitive to the high PaCO2 stimulus for breathing, & more reliant on the low PaO2 stimulus (“CO2 retention”) Give O2 carefully to these patients (high PaO2 may suppress patients’ hypoxic respiratory drive, ↓ their breathing, & ↑↑↑ PaCO2) ↑ retrosternal air space (on lateral CXR) Hyper-lucent (darker) lung fields, ↓ lung markings (on frontal CXR) • Arterial Blood Gasses (ABGs) • Chest X-Ray (CXR): frontal and lateral Legend: Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications Published January 7, 2013 on www.thecalgaryguide.com COPD: !"#$ 气流阻塞 肺泡通气↓ 呼气时,胸膜腔正压挤压气 道à 阻塞↑ 作者: Yan Yu 审稿人: Jason Baserman, Jennifer Au, Naushad Hirani*, Juri Janovcik* 译者:Zihong Xie (谢梓泓) 翻译审稿人: Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生 慢性阻塞性肺疾病 (COPD) 肺组织损伤 没有弹性回缩力将 气体排出肺 肺实质与血管分布减少导 致气体交换面积↓ 弥散功能↓ (肺功能检查) 更多的CO2残留 并扩散到血液中 高碳酸血症: PaCO2 > 45 (动脉血气) 血流灌注通气不良的肺泡 时无法获得足够的氧气 总呼气时长较正常长 FEV1/FEV < 0.7 (肺功能检查) 肺无法完全排空 更多空气潴留在肺部 (肺过度充气) 低氧血症: PaO2 < 70mmHg (动脉血气) 通气-灌注不匹配 肺泡-动脉氧分压差↑ (可通过动脉血气分析计算得出) 横膈低平, 下移至第10肋后端 及以下部位 (胸部正位片) TLC与VC增大 (肺功能检查) 缩写: • • FEV1: 1秒用 • VC:肺活量 PaO2: 动脉血 力呼气量 氧分压 空气不会阻挡X射线, 在X光片上呈现为黑色 慢性高碳酸血症使呼吸中枢对PaCO2 刺激呼吸的敏感性下降 & 更依赖于低PaO2的刺激 (“二氧化碳潴留”) 给患者吸氧时需注意(高PaO2 可能会抑制患者低氧时对呼吸的 刺激,使呼吸驱动↓ & PaCO2↑↑↑ ) • FVC: 用力肺 • 活量 • TLC:肺总量 慢阻肺相关检查 : PaCO2: 动脉 血二氧化碳 分压 胸骨后间隙↑ (胸部侧位片) 肺纹理↓ • 肺功能检查 • 动脉血气分析(Arterial Blood Gasses, ABGs) • 胸部正侧位片 • 当患者发热和湿咳,特别是胸片上见肺野不清晰时: 肺透亮度↑, (胸部正位片) 考虑进行痰革兰氏染色及痰培养以排除肺炎可能 图注: 病理生理 机制 体征/临床表现/实验室检查 并发症 2013年1月7日发布于 www.thecalgaryguide.com COPD: Complications Lung inflammation Chronic Obstructive Pulmonary Disease (COPD) Airway obstruction ↓ inhaled air in alveoli and terminal bronchioles Rupture of emphasematous bullae on surface of lung Inhaled air leaks into pleural cavity and is trapped there Pneumothorax Feeling a loss of control over one’s life, and hopelessness for the future Goblet cell proliferation, ↑ mucus production Death of airway epithelium ciliated cells ↓ oxygenation of the blood passing through the lungs Chronic hypoxemia Kidneys compensate by ↑ erythropoietin (EPO) production ↑ Hemoglobin and red blood cell synthesis Polycythemia (secondary) Hypoxic alveoli cause the pulmonary arterioles perfusing them to reflexively vasoconstrict Since most alveoli in the lungs are hypoxic, hypoxic vasoconstriction occurs across entire lung Vasoconstriction ↑ blood pressure within lung vasculature Pulmonary hypertension ↑ workload of the right ventricle (to pump against higher pressures) To compensate, the right ventricle progressively hypertrophies and dilates, but over time its output ↓ Cor pulmonale (Right heart failure in isolation, not due to Left heart failure) Mucus trapped in airways, serve as nidus for infection Acute exacerbation of COPD (AECOPD) Pneumonia The chronic, systemic inflammation in COPD is a hyper-metabolic state that consumes calories Macro-nutrient deficiency Trouble with respiration lead to inactivity and deconditioning Wasting, muscle atrophy More inactivity and deconditioning perpetuates the cycle Depression Author: Yan Yu Reviewers: Jason Baserman Naushad Hirani* Juri Janovcik* * MD at time of publication Legend: Pathophysiology Mechanism Sign/Symptom/Lab Finding Complications Published January 7, 2013 on www.thecalgaryguide.com COPD: !"# 肺部炎症 杯状细胞增殖, 气道上皮纤毛 粘液产生↑ 细胞死亡 黏液潴留呼吸道,成为感 染的病灶 慢性阻塞性肺疾病 (COPD) 气道阻塞à 吸入肺泡和终末细 肺大疱破裂 吸入的空气渗入 并潴留于胸腔 气胸 感觉生活失控,对未 来感到绝望 抑郁 作者: Yan Yu 审稿人: Jason Baserman, Naushad Hirani*, Juri Janovcik* 译者: Zihong Xie (谢梓泓) 翻译审稿人: Yonglin Mai (麦泳琳), Zesheng Ye (叶泽生) * 发表时担任临床医生 支气管的空气 ↓ 流经肺的血液进行气 缺氧的肺泡à灌注肺泡的肺小动 慢性阻塞性肺疾 病急性加重期 (AECOPD) 肺炎 体交换↓ 慢性低氧血症 肾脏合成促红细胞 生成素进行代偿↑ 血红蛋白与红 细胞合成↑ 红细胞增多症 (继发性) 脉发生反射性血管收缩 肺大部分肺泡缺氧à整个肺 都出现缺氧性血管收缩 肺血管收缩 à 肺血管压力↑ 肺动脉高压 ↑ 右心室负荷(泵血时对抗高压) 为了代偿,右心室逐渐肥大和扩张, 但随着病程进展,右心室输出量 ↓ 肺心病 (单独出现右心衰竭,非左心衰) COPD所致的慢性全身 呼吸困难导致活 性炎症会使机体处于高 动量减少和活动 代谢状态,消耗能量 耐量降低 宏量营养 素缺乏症 消瘦,肌肉萎缩 运动量下降和活动耐量 的降低造成恶性循环 图注: 病理生理 机制 体征/临床表现/实验室检查 并发症 2013年1月7日发布于 www.thecalgaryguide.com " />

Chronic Cough Pathogenesis_2021

Chronic Cough: Pathogenesis
       ACE Inhibitors
Protussive mediator accumulation (bradykinin, substance P)
Infection
IP; likely chronic ↑ cough receptors commonly pertussis- related or post-viral
Asthma
↑ EDN
↑ MBP levels in RT
Neoplasm
Bronchiectasis
COPD
GERD
Allergic Rhinitis
        ↑ sputum production
and accumulation
Damage from chronic inflammation causes poor mucus clearance
Inhalants trigger ↑ cytokines ↑ mucus
Aspiration of refluxed microdroplets
Irritation by post- nasal drip
            ↑ inflammatory mediators in RT
Mechanical obstructions (i.e. mediastinal masses, neoplasms)
Foreign body Presence irritation of irritants
  Stimulation of sensory nerve receptors expressed on nerve endings penetrating the epithelia of the upper RT
         Abbreviations:
• RT: respiratory tract
• IP: indefinite
pathophysiology
• GERD: Gastro-esophageal
reflux disease
• EDN: Eosinophil-derived
neurotoxin
• MBP: Major basic protein
• COPD: Chronic
Obstructive Pulmonary Disease
Complications: Syncope, insomnia, hemoptysis, rib fractures
Slowly adapting receptors
Respond to mechanical forces during breathing
Stimulation of the vagus nerve
Activation of nucleus tractus solitarius in central respiratory generator (brainstem)
Generation of efferent cough signal
Chronic cough: Cough of over 8 weeks duration with no dyspnea or fever
C-fibre receptors
Respond to chemical stimuli (bradykinin, capsaicin, acid/alkaline solutions, mannitol, hypertonic saline, and other inhaled irritants)
Authors: Arsalan Ahmad Lance Bartel Reviewers: Midas (Kening) Kang Usama Malik Ciara Hanly Yonglin Mai (麦泳琳) Yan Yu*, Eric Leung* * MD at time of publication
Rapidly adapting receptors
Responds to mechanical stimuli, such as physical obstruction of the airways
       Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Feb 06, 2018, updated Dec 4, 2021 on www.thecalgaryguide.com

complications-of-chronic-kidney-disease-ckd

Complications of Chronic Kidney Disease Chronic Kidney Disease (CKD)
Authors: Samin Dolatabadi, Brooke Fallis Reviewers: Jessica Krahn, Meena Assad, Yan Yu* Juliya Hemmett* * MD at time of publication
Abnormalities of kidney structure or function that is present for 3 or more months
          Kidney tubules atrophy & kidney interstitial tissue undergo fibrosis
Kidney damage ↓ erythropoietin production (normally a kidney function)
↓stimulation of bone marrow → ↓ red blood cell production
Build up of toxic substances (e.g. urea, guanidine, and indoxyl sulfate)
↓ Conversion of calcidiol to calcitriol in by kidney
↓ Calcitriol levels in blood
↓ Ca+2 absorption from small intestine
Hypocalcemia
↓ Glomerular Filtration Rate
↑ Vascular calcification and endothelial dysfunction (e.g. changes in permeability, clotting response to inflammation, amongst other mechanisms)
Atherosclerotic disease (see “Complications of Atherosclerosis” slide)
↓ Lipoprotein lipase, apoA-1, and Lecithin-cholesterol acyltransferase
activity acting on serum lipoproteins (complex mechanisms)
↓ Lipoprotein clearance from blood
Dyslipidemia (↑ LDL, ↑ triglycerides, ↓ HDL)
       Toxic damage ↓ red blood cell survival
Anemia
Uremic Syndrome
              ↓ Renal excretion of phosphate
↑ phosphate remaining in bloodstream
Hyperphosphatemia
↑ serum phosphate binds with ionized calcium
↓ Renal excretion of K+
↑ K+ remaining in bloodstream
Hyperkalemia Edema
↓ Renal excretion of ammonium
Reduced filtration capability ↑ organic anions remaining in blood→ ↑ anion gap
↓ Renal excretion of salt and water
↑ in extracellular fluid → systemic volume overload
      ↓ calcium in blood ↓ inhibition of Parathyroid Hormone (PTH) release
↑ PTH in blood
Secondary hyperparathyroidism
Metabolic Acidosis
       Hypertension
Pitting
   Chronic kidney disease – mineral and bone disorder (CKD-MBD)
 Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
Published Jan 4, 2022 on www.thecalgaryguide.com

NSAIDs and the Kidney Nephrotoxicity

NSAIDs and the Kidney: NSAID induced Nephrotoxicity Non-steroidal anti-inflammatory drugs (NSAID)
Authors: Kyle Moxham Mehul Gupta Reviewers: Emily Wildman Yan Yu* Adam Bass* * MD at time of publication
  Inhibition of Cyclooxygenase COX-1 (expressed in kidney) and COX-2 (expressed in kidney and sites of inflammation)
NSAID induced nephrotoxicity: associated with chronic NSAID usage independent of dosage
COX inhibition ↑ conversion of arachidonic acid (AA) to leukotrienes, causing systemic T-cell dysfunction (unknown mechanism)
Type IV systemic hypersensitivity (delayed
T helper cell mediated) reaction to drug exposure
T cells release inflammatory cytokines into the bloodstream
(see Calgary Guide slide on NSAIDs and the Kidney: Mechanism of Action and Side Effects)
       T cells infiltrate the renal interstitium, sparing the glomeruli and blood vessels
Overproduction of cytokines by T cells causing inflammation,
tissue damage, and cell death, of the renal intersitium
Drug Induced Acute interstitial nephritis (AIN): a type of immune-mediated tubulointerstitial injury
Activated T-cells infiltrate the glomerulus and cause podocyte injury (epithelial cells attached to the glomerular basement membrane)
    Membranous nephropathy:
nephrotic syndrome involving autoimmune glomerular basement membrane thickening & complete podocyte effacement (seen on kidney biopsy)
Minimal change disease: nephrotic syndrome caused by autoimmune podocyte effacement (seen on kidney biopsy)
      Cytokine mediated activation and
proliferation of immune cells like macrophages and eosinophils
Cytokines travel to hypothalamus,
causing change in the body’s thermal set point
Fever
Podocyte effacement allows for serum proteins to across the glomerulus into the tubular lumen (see Calgary Guide slide on Nephrotic Syndrome for full mechanisms)
      Repeated NSAID exposure causing
recurrent unrecognized AIN and damage of the kidney
Chronic interstitial nephritis /analgesic nephropathy
Infiltrating immune cells (predominantly neutrophils) are filtered into/enter the renal tubules, and form clumps (“casts”) within the tubulesà casts are then released into urine
WBC cast on urinalysis
↑ Blood eosinophils
Immune cells infiltrate the
dermis and epidermis of the skin
Rash
Abnormal quantities of protein present in urine
Protein present in the blood is improperly filtered into the filtrate at the glomerulus
           Protein- Creatinine
Ratio >3.5mg/mg
Proteinuria >3.5g/d
Hypo- albuminemia
↓ plasma oncotic pressure resulting in fluid extravasation into the interstitium (see Calgary guide slide on Edema for full mechanisms)
   Pitting Edema
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published January 13, 2022 on www.thecalgaryguide.com

Langerhans Cell Histiocytosis

Langerhans Cell Histiocytosis: Pathogenesis and clinical findings
Precursor cells differentiateàClonal expansion of abnormal (constitutive MAPK activation) CD1a+/CD207+ (Langerhans cell phenotype surface receptors) dendritic cells in tissue(s)
     Somatic BRAFV600E mutation: BRAF is a kinase in the MAPK pathway
Other somatic (non- reproductive cell) mutations
Idiopathic (unknown cause)
Mutation(s) can occur in one of these precursor cell types*
Hematopoietic stem cell (earliest cell of blood cell differentiation) in bone marrow
Committed dendritic cell (type of myeloid antigen-presenting cell) precursor in bone marrow or blood
Committed dendritic cell precursor in tissue
Constitutive activation of the MAPK pathway (signalling pathway that regulates variety of cellular processes) in one of these precursor cell types
          *Note: Based on the “misguided myeloid differentiation” modelàthe earlier the mutation(s) occur in the myeloid cell differentiation pathway, the more severe the disease.
Langerhans Cell Histiocytosis
Accumulation of abnormal CD1a+/CD207+ dendritic cells (Langerhans Cell Histiocytosis cells or LCH cells) with an inflammatory background in one or more organs
  Authors:
Ran (Marissa) Zhang Reviewers:
Mehul Gupta
Kiera Pajunen
Yan Yu*
Lynn Savoie*
* MD at time of publication
↑ recruitment & activation of T cells, macrophages, eosinophils in tissue(s) around the body
↓ CCR7 & CXCR4 (chemokine receptors) expression on LCH cellsàinhibits migration of LCH cells to lymph nodes
↑ BCLXL (an apoptosis regulator protein) expression on LCH cellsà inhibits apoptosis of LCH cells
       Immune cell infiltration & ↑ pro-inflammatory chemokine/cytokine release à dysregulated local & systemic inflammation
Accumulation of LCH cells in tissue(s) around the body
  Inflammatory lesion (an area of abnormal tissue) formation in one or more organs:
       In pituitary stalk
Mass effectà
obstruction of antidiuretic hormone (complex mechanisms)
In liver
Invasion & accumulation of cells foreign to liverà expands liver
Chronic local inflammation
Scarring of bile ducts
↓ bilirubin clearance from liveràbuildup into serum
↑ serum bilirubin
Jaundice
In cortical bone Cytokine production
à↑ osteoclast
(cells that break down bone) activity
↑ rate of bone loss
Osteolytic bone lesions
In bone marrow
Unclear mechanism but likely due to macrophage activation
↑ phagocytosis (ingestion & destruction) of blood cells
In spleen
Invasion & accumulation of cells foreign to spleen
Forms aggregates that expand the red pulp (functions as the blood filter in spleen)
Splenomegaly
In skin
Unclear mechanisms
Variable presentations: most commonly pinpoint erythematous (red) papules or erythematous plaques with crusting & scaling
            Hepatomegaly • BRAF- B-Raf proto-oncogene, serine/threonine kinase
• CCR7- C-C motif chemokine receptor type 7
• CD1a- Cluster of differentiation 1A
• CD207- C-type lectin domain family 4 member k • CXCR4- C-X-C chemokine receptor type 4
• LCH- Langerhans Cell Histiocytosis
• MAPK- Mitogen-activated protein kinase
Diabetes Insipidus
     Abbreviations:
• BCLXL- B-cell lymphoma-extra large
   Anemia, thrombocytopenia &/or neutropenia
   Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published January 13, 2022 on www.thecalgaryguide.com

NSAIDs and the Kidney mechanism of action and side effects

NSAIDs and the Kidney: Mechanism of Action and Side Effects
Authors: Kyle Moxham Mehul Gupta Reviewers: Emily Wildman Yan Yu* Adam Bass* *MD at time of publication
  Concurrent use of angiotensin- converting enzyme inhibitors (ACEi) or angiotensin II receptor blockers (ARB)
AECi and ARBs act to ↓ renin- angiotensin-aldosterone system (RAAS) activation
Reduced angiotensin (AT) 2 activity at its receptors on efferent arteriole of the nephron
Net vasodilation of efferent arterioles ↓ in glomerular pressure
↓ blood perfusing kidney tissueà↑ hypoxemia & renal ischemia
Pre-existing ↓effective arterial blood volume (EABV) from
dehydration, GI loss, diuretics, CKD, CHF, cirrhosis, etc.
Decreased EABV triggers endogenous renal autoregulation, resulting in norepinephrine (NE) mediated vasoconstriction of afferent arteriole of the nephron
Net ↓ in renal blood flow and ↓ in glomerular pressure
↓ volume of blood filtered by the glomeruli per unit time
Non-steroidal anti-inflammatory drugs (NSAIDs)
     Inhibition of Cyclooxygenase COX-1 (expressed in kidney) and COX-2 (expressed in kidney and sites of inflammation)
↓ Renal prostaglandin (PG) synthesis: local hormones involved in renal homeostasis
NSAID induced nephrotoxicity:
associated with chronic usage independent of dosage
(see Calgary Guide slide on NSAIDs and the Kidney: NSAID induced nephrotoxicity)
↓ intrarenal PG reduces inhibitory effects of PG over ADH in cortical colleting duct (CCD) of the kidneyà↓ antagonism on ADH activity
↑ ADH activity causes insertion of more
aquaporins (water channels) in the collecting duct of renal tubules
Net ↑ in volume of water reabsorbed into the blood
           ↓ vasodilatory effect of PG at the afferent arteriole of the nephron
↓ Glomerular filtration rate (GFR)
↓ PG signalling results in ↓ renin secretion at juxtaglomerular apparatus
Low renin levelsà↓ conversion of angiotensinogen into its AT1 form and, by extension, ACE mediated conversion of AT1 to AT2
↓ ACE 2 signalling leads to ↓ levels of aldosterone in the serum
↓ Na+ and K+ channel insertion on apical surface and ↓ Na/K ATPase activity on basolateral surface of principal cells
↓ K+ excretion into urine, and ↓ Na+ reabsorption
back into the blood, at the late DCT and collecting duct of the kidney
               Pre-renal Acute Kidney Injury (AKI): kidney injury due to renal hypoperfusion
Prolonged and/or severe ischemia causes cell death and aggregation of tubular epithelial cells of the kidney with subsequent inability to reabsorb luminal Na+
          Renal dehydration predisposes
precipitation of uromodulin protein
Renal tubules mold uromodulin into cylindrical structures known as “casts”. Casts that contain only uromodulin protein are known as “hyaline casts”
Hyaline cast seen on urinalysis
Hypoperfusion of the kidney activates the renin- angiotensin- aldosterone system (RAAS)
↑ amount of sodium (Na) reabsorbed from the filtrate (less Na excreted)
Fractional excretion of Na <1%
Papillary necrosis: ureteral passage of sloughed ischemic tissue causing ureteral obstruction
Acute tubular necrosis: a type of kidney injury causing damage to the tubules
Hypertension
        Post-renal AKI: a type of kidney injury due to
obstruction of the urinary tract
Distal distal obstruction of the urinary tract causes fluid to accumulate within the kidneysàenlarging the kidneys
Renal Ultrasound shows hydronephrosis (enlarged kidneys)
Damaged tubule epithelial cells slough into the tubular lumen
Epithelial cell breakdown in the tubular lumen
releases uromodulin & other proteins, which aggregate into “casts” (cylindrical imprints of the renal tubule). The varied protein content of these casts result in them having a coarse, granular appearance
Damaged tubular epithelial cells are unable to properly reabsorb sodium
↓ amount of sodium (Na) reabsorbed from filtrate into the blood (more Na excreted)
Fractional excretion of Na >2%
True excess of free water relative to Na+ in the blood
Excess free water ↑ venous hydrostatic pressure (see Calgary Guide slide on edema for full mechanisms)
Pitting Edema
          Hyperkalemia
Hyponatremia
     Coarse granular casts seen on urinalysis
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published January 29, 2022 on www.thecalgaryguide.com

adult-pneumonia-pathogenesis-and-clinical-findings

Adult Pneumonia: Pathogenesis and clinical findings
Author: Laura Byford-Richardson Reviewers: Tara Shannon, *Yan Yu, Sadie Kutz , Natalie Morgunov, *Kerri Johannson, *Julie Carson *MD at time of publication
  Smoking à suppressed neutrophil function and damaged lung epithelium
Chronic lung conditions e.g. COPD, asthma, lung canceràdestroys lung tissue and offers pathogen more niduses for infection
Immune suppression e.g. HIV, sepsis, glucocorticoids, chemotherapyà suppression of immune response
Systemic inflammatory response towards invading microbe
Systemic cytokine release leads to a disruption in hypothalamic thermoregulation
Exposure to a pathogen via
inhalation, aspiration, contiguous Notes:
  or hematological mechanism
Susceptible host and/or virulent pathogen
Proliferation of microbe in lower airways and alveoli
Local response by alveolar epithelial cells release chemokines into surrounding tissue to recruit neutrophils to the site of inflammation
• Pathogens can be bacteria, viruses, fungi and parasites
• Pneumonia is a lower respiratory tract infection (in contrast to
upper respiratory tract infections such as bronchitis) and can be further classified by location of exposure: community, health- care, hospital acquired
Inflammatory response varies depending on type of invading pathogen (i.e. S. Pneumonia causes a lobar pattern and Influenza A & B cause an interstitial pattern)
           LOBAR: Accumulation of neutrophils and plasma exudate from capillaries into alveoli specific to a lung area/lobe
INTERSTITIAL: Accumulation of infiltrates (i.e. inflamed cellular debris) in the alveolar walls (i.e. space between the alveolar spaces and bloodstream)
             Fever
Notes:
• Other signs and symptoms
of pneumonia exist such as chest pain, accessory muscle use, crackles on auscultation and fatigue
• These signs and symptoms are less specific to the ones outlined on this slide
Irritation and attempted clearance of airways
Fluid infiltrates are inside alveoli, airway clearance leads to phlegm production
Productive Cough
Fluid build up does not allow X-rays to pass through à white opacity on plain film at site of fluid buildup
Consolidation on CXR
Alveolar sacs blocked by fluid accumulation
Thickening of alveolar walls ↑ diffusion distance between alveoli & capillaries
Irritated alveolar walls trigger cough reflex
Since fluid infiltrates are NOT in the alveoli, attempts to empty the alveoli through coughing doesn’t lead to production of fluid
Dry Cough
Chills/Rigors
      ↓ Exchange of CO2 and O2
Hypoxemia
Triggers peripheral and central chemoreceptors to ↑ respiratory drive
Dyspnea
        Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Sept 26 2016, updated Feb 9, 2022 on www.thecalgaryguide.com

hip-osteoarthritis-pathogenesis-and-clinical-findings

Hip Osteoarthritis: Pathogenesis and clinical findings
Authors: Ebrahim Alawadhi, Alyssa Federico Reviewers: Mehul Gupta, Tara Shannon, Yan Yu*, Richard Ng* * MD at time of publication
  Primary causes Aging
↓ Synovial fluid in the hip joint
Secondary causes
           Gender
Females > males
Genetics
Family history of osteoarthritis
High-impact sports, activities, and occupations
Repetitive stress on the hip joints
Obesity
↑ Loading on hip joints
Inflammatory disease (e.g. Rheumatoid arthritis )
Trauma
Infection (e.g. septic arthritis)
Anatomical abnormality (e.g. developmental hip dysplasia)
         ↑ Cartilage stiffness and degeneration ↑ Friction in the hip joint with movement
Changes in normal hip architecture Abnormal load on the hip joint
Radiographic changes
See Osteoarthritis (OA): X-Ray Features slide
Repeated attempts to repair cartilage damage and bone
Bone spur (bony growth) formation in joint
Joint fluid accumulation from mild inflammation of the joint
Stiffness
Limited joint movement
    Hip Osteoarthritis
Multifactorial condition that manifests as degeneration of cartilage, bone, and synovium in the hip joint
↓ Cartilage between femoral head and acetabulum ↑ Bone on bone contact
            Mechanical symptoms (crepitus, locking, clicking, catching)
Bones rubbing together activates nociceptors within the hip joint
Nociceptors further aggravated by actions that ↑ bone on bone contact (e.g. prolonged loading, movements that ↓ joint space)
Worsening pain in the groin region
Favour the affected leg to avoid pain while walking
↓ Space for the femoral head to move
↓ Range of motion of affected hip joint
Limited range of motion when walking
Antalgic gait
↓ Use of muscles around the affected hip
Muscular atrophy around the hip
       C Sign: patient identifies location of pain by cupping lateral hip with one hand, which creates a “C” shape with that hand
Pain in groin region
↑ Sensitivity of surrounding nerves
Radiating pain to buttocks and knee
Inactivity
Hip instability when walking
Muscle contractures
↑ Falls
                 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published March 6, 2022 on www.thecalgaryguide.com

cough-physiology

Cough: Physiology
Mass or particulate matter
in the throat & airways
↑ pressure on mucosa Pressure on rapidly and slowly
adapting mechanoreceptors
Mechanoreceptor activation opens ion channels on vagus neurons
Authors: Calvin Howard Reviewers: Nilani Sritharan Ciara Hanly Yonglin Mai (麦泳琳)* Yan Yu* Stephen Field* * MD at time of publication
 Systemic immune reaction
Inflammation of alveoli Cytokine release
Cytokines bind to sensory neurons
Neurons ↑ receptor sensitivity and number
↑ sensation of stimuli Ionic flux across neuronal
membrane
Vagus neurons depolarize
Thinly myelinated axons (Aδ fibers)
General chemicals, heat, cold, and hydrogen ions
Transient receptor potential vanilloid (TRPV)/Transient receptor potential ankyrin (TRPA) receptors activate
TRPV/TRPA activation opens ion channels on vagus neurons
Unmyelinated axons (C fibers)
                       carry depolarization
Vagus nerves conduct depolarization to
carry depolarization
SUMMARY
Afferent pathways activated
Cough center (medulla)
Efferent pathways Respiratory muscles
    nucleus of the solitary tract
Medullary nuclei activated, stimulates:
        Phrenic nerves
Diaphragm contraction
Spinal nerves
External intercostal contraction
Vagal nerves
Glottis closure
Spinal nerves
Thoracic, abdominal, and pelvic muscle contraction
Vagal nerves
Glottis opens, allowing forceful expiration
Intrathoracic pressure and volume decrease
             Volume in lungs increases
Intrathoracic pressure increases
Mechanical Cough
        Legend:
Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
Complications
Published Sept 1, 2019, updated Dec 5, 2021 on www.thecalgaryguide.com

isotretinoin-systemic-retinoid-mechanisms-and-side-effects

Isotretinoin (Systemic Retinoid):
Mechanisms and Side Effects
Isotretinoin
Authors: Ayaa Alkhaleefa Reviewers: Mehul Gupta, Ben Campbell Stephen Williams, Lauren Lee, Yan Yu*, Laurie Parsons* * MD at time of publication
↑ Apoptotic signaling in sebocytes
Inhibits androgen nuclear receptors responsible for sebum secretion
↓ Sebaceous lipogenesis
       ↑ Apoptotic signaling in neural crest cells during embryonic development
Alterations in hindbrain, neural crest, otic anlage, and reduced pharyngeal arch in embryo
Craniofacial, cardiac, thymic, and central nervous system malformations in fetus
Isotretinoin isomerizes to all-trans retinoic acid (ATRA)
ATRA enters cell nucleus and binds retinoic acid receptors and retinoic X receptors
ATRA induces tumour necrosis factor-related apoptosis-inducing ligand
↑ Apoptotic signaling in epidermal keratinocytes
      ↑ Expression of FoxO1
↑ Expression of p53 (tumour suppressor)
Release of caspases 3, 6, 7, and 9
        ↑ Cell cycle inhibitors p21 and p27
↓ Pro-survival proteins (Survivin)
Sebaceous gland involution
Sebum suppression
C. acnes unable to break down sebum into pro- inflammatory lipids
↓ Colonization with C. acnes
      Teratogenicity
↑ Cornification (death) of epidermal keratinocytes
↓ Corneodesmosomes (main adhesive structures of the stratum corneum)
↓ Cohesion of corneocytes (dead keratinocytes)
↓ Corneocyte buildup in pilosebaceous follicles
C. acnes unable to populate and release cytokines in corneocytes
     Epidermis
Dermis
Pilosebaceous follicle
↓ Stratum corneum thickness
↑ Trans-epidermal water loss
Dryness, peeling & inflammation
of lips (cheilitis), skin (dermatitis) & mucosa (mucositis)
↓ Comedogenesis
  Sebum
Hair follicle Dermal-
Epidermal Junction
Sebaceous gland (sebocytes)
Death of sebocytes in pilosebaceous follicles
        Reduction of number and size of acne lesions
        Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published March 20, 2022 on www.thecalgaryguide.com

proximal-biceps-tendon-rupture

Proximal Biceps Tendon Rupture: Pathogenesis and clinical findings
Author: Alyssa Federico Reviewers: Tara Shannon, Mehul Gupta, Yan Yu*, Gareth Ryan* * MD at time of publication
   Aging
Decreased tendon strength from age- related changes in extracellular matrix
Male gender
Smoking
Corticosteroid and fluoroquinolone use
Shoulder Overuse
Repetitive overhead sports like swimming & tennis
         Shoulder overuse injuries (rotator cuff tears, shoulder impingement, & tendonitis) place more stress on the proximal biceps tendon
Repetitive microtrauma and inflammation of biceps tendon Compromised biceps tendon integrity from intra-tendinous collagen degeneration, disorientation, and thinning
  Increased involvement in physical labour and hobbies
Impaired cell proliferation and healing after injury
Exact mechanisms remain unclear
       Bicep tendon involved in shoulder stabilization, elbow flexion, and forearm supination
Biceps muscle cannot contract with as much force when one head is detached
Heavy lifting or fall on outstretched handà sudden tension in the proximal biceps tendon
Proximal biceps tendon rupture
Long head of proximal biceps tendon detaches from bony attachment on supraglenoid tubercle
Audible “pop” at time of injury
No tension to hold proximal bicep tendon head in place
Proximal bicep tendon head retracts distally
             Biceps muscle fatigue and cramping
Weakness in shoulder flexion, elbow flexion & supination
Nerve damage (musculocutaneous nerve) during injury
Local inflammatory response at site of injury
Surrounding blood vessels damaged during injury
Blood collection under skin surface
Immediate bruising over rupture site
        Sudden and sharp pain at shoulder with initial injury
Inflammatory response aggravates musculocutaneous nerve
Swelling over rupture site
     + Speed test: affected elbow extended and forearm supinated. Shoulder flexed against resistanceàpain at bicipital groove
+ Neer test: on affected side, scapula stabilized while arm passively flexed and internally rotatedàpain at bicipital groove
+ Yergeson test: affected elbow flexed at 90° and forearm pronated. Forearm supinated against resistance àpain at bicipital groove
“Popeye” deformity:
mass in distal upper arm due to excessive
retraction of biceps muscle distally, away from its origin
Palpable gap
between proximal biceps tendon and bicipital groove
      Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
Published April 20, 2022 on www.thecalgaryguide.com

distal-biceps-tendon-rupture

Distal Biceps Tendon Rupture: Pathogenesis and clinical findings
Authors: Alyssa Federico Reviewers: Tara Shannon, Mehul Gupta, Yan Yu*, Gareth Ryan* * MD at time of publication
      Aging
↓ Tendon strength from age-related changes in extracellular matrix
Male gender
Overuse
Common in throwing and racquet sports
Repetitive microtrauma and inflammation of biceps tendon
Corticosteroid and fluoroquinolone use
Smoking
Hypovascularity
Area of hypovascularity between regions supplied by the brachial artery and posterior radial recurrent artery
Impaired ability to heal from microtrauma
Mechanical impingement
Compression of distal biceps
tendon by surrounding bone (radial tuberosity, proximal ulna) during forearm pronation
Repetitive compressionà tendon degeneration
          ↑ Involvement in physical labour and hobbies
Exact mechanisms remain unclear
Impaired cell proliferation
        Biceps tendon inserts at the radial tuberosity to control forearm supination and flexion
Compromised biceps tendon integrity from intra-tendinous collagen degeneration, disorientation, and thinning
Sudden eccentric force to flexed elbow (forced lengthening of a contracted bicep muscle)àtension in the distal biceps tendon
Biceps muscle cannot contract when distal tendon is detached from insertion site
Distal biceps tendon rupture
Complete detachment of distal biceps tendon at the attachment site on the radial tuberosity
Audible “pop” at time of injury
               Weak forearm supination and flexion
+ Ruland biceps squeeze test:
Affected elbow held in 60-80° of flexion, with forearm slightly pronated. Distal biceps muscle squeezed by practitioner à forearm does not supinate
Local inflammatory response at site of injury
Inflammatory response aggravates musculo- cutaneous nerve
Pain over rupture site after initial injury
Swelling over rupture site
Nerve damage (musculocut aneous nerve) during injury
Sudden and sharp pain at
elbow at initial injury
Surrounding blood vessels damaged during injury
Blood collects under skin surface
Immediate bruising over rupture site
Biceps tendon retracts proximally
High tension during tendon retraction
Fragment of bone torn off from tendon insertion site on radial tuberosity
Avulsion fracture of
radial tuberosity
    Connective tissue attachment (aponeurosis) torn during injury
    Palpable gap between distal end of biceps tendon & radial tuberosity
+ Hook test: Elbow actively flexed to 90° and forearm supinated. Index finger of examiner cannot be placed beneath distal aspect of biceps tendon (>1 cm deep)
     Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published April 20, 2022 on www.thecalgaryguide.com

irritable-bowel-syndrome-ibs-pathogenesis-and-clinical-findings

Irritable Bowel Syndrome (IBS): Pathogenesis and clinical findings
Authors: Ben Campbell Reviewers: Mehul Gupta Kiana Hampton Yan Yu* Edwin Cheng* * MD at time of publication
Note:
*Several signaling pathways in the gut are believed to play a role in IBS. The serotonin pathway is represented here for illustrative purposes, and it is a common target of therapy.
       Pathogenesis is multifactorial, not fully understood, and may arise from any one or a combination of these factors
Genetic factors
are believed to influence many of these pathways
Environmental factors
Gut infection, ischemia, injury, chronic low-level inflammation, altered microbiome
Abnormal gut chemical milieu (cytokines, lymphocytes, mast cells, hormones)
One common pathway: altered quantity or activity of serotonin-releasing enterochromaffin * cells and serotonin reuptake transporters in gut
Serotonin is a key stimulator of gut muscle contraction and sensory signaling
Altered bowel motility
Bowel motility can ↑, ↓ or alternate ↑/↓
Psychosocial factors
Anxiety, depression, adverse experiences, dysregulated stress response
    Brain can alter normal reflexes of enteric nervous system (e.g. via hypothalamic stress hormone release)
↑ Activity of sensory receptors in gut + recruitment of formerly ”silent” receptors
Altered visceral bowel sensation
Visceral hypersensitivity
Complex mechanisms
↓ Brain’s ability to modulate pain signals from gut;
↑ vigilance to pain
             Irritable Bowel Syndrome
Disorder of brain-gut interaction with gastrointestinal manifestations
Not a predominantly inflammatory condition
During times of ↑ bowel motility:
No lesions in gut mucosa
(Absence of ”red flags” for organic disease)
↑ Impacts of other factors that precipitate diarrhea
Dietary sensitivity
(e.g. to intake of fermentable oligo/di/ monosaccharides and polyols— FODMAPs—which draw water into gut lumen by osmosis)
     ↑ Impacts of other factors that precipitate constipation
Dietary sensitivity (e.g. to inadequate intake of motility- promoting fibre)
During times of ↓ bowel motility:
↑ Time for water absorption from feces and bacterial fermentation
↑ Hard stool and trapped gas in bowel
↑ Number and sensitivity of active pain receptors (nociceptors) in gut
Hyperalgesia
↑ Response to painful stimulus
Stretch receptors in gut stimulate pain in normally unconscious neural pathways
Allodynia
Pain in response to normal stimulus
↑ Peristalsis
↓ Time for water absorption from feces
↑ Water volume in gut lumen
                   ↑ Stretch / stimulation of visceral afferent nerves in gut
Absence of bloody stool and anemia
Absence of nocturnal symptoms and inflammatory markers
         Constipation
Bloating
Diffuse abdominal pain
All symptoms may exacerbate psychosocial factors
Bowel distension
Diarrhea
   Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published April 30, 2022 on www.thecalgaryguide.com

abnormal-uterine-bleeding-aub-pathogenesis-and-clinical-findings

Abnormal Uterine Bleeding: Pathogenesis and clinical findings
Authors: Joshua Yu, Karen Paik Reviewers: Ayaa Alkhaleefa, Parker Lieb,
     Hypothyroidism
Hypothalamus senses ↓ serum thyroxine
↑Thyrotropin releasing hormone (TRH) release from hypothalamus
↑Prolactin (see Feedback Loop: Prolactin)
Exogenous estrogen (e.g. estrogen-only birth control)
↑ Peripheral adipose tissue
↑ Aromatase (enzyme present in adipose tissue)
↑ Conversion of androgens to estrogens (aromatization)
Excessive stress, exercise, low body mass (mechanism unclear)
↓ Hypothalamic gonadotropin releasing hormone secretion
↓Luteinizing hormone and follicle stimulating hormone release from pituitary
↑ Estrogen
Heavier bleeding
Angiogenesis in tumour tissue
Polycystic ovarian syndrome (see slide)
Pelvic inflammatory disease
Immature hypothalamic- pituitary-ovarian axis
(an immature axis is transient in most females)
↓ Positive feedback of estrogen in late follicular stage
No LH surge
Adenomyosis (endometrial tissue grows into uterus muscular wall)
Endometrial polyps
Retained products of contraception
Infection
Inflammation of endometrium
Endometrium is more fragile
Anovulatory Bleeding
Leiomyoma (benign tumour in myometrium)
Tara Shannon, Hannah Yaphe, Dr. Sarah Glaze* * MD at time of publication
       Ovarian Scarring
Foreign bodies
Intrauterine device perforation
Physical trauma
          Impaired follicle maturation
Anovulation
Corpus luteum does not form
No ovarian progesterone production
↑ Estrogen to progesterone ratio
Proliferative effect of estrogen unopposed
Endometrial proliferation
No progesterone to organize growing endometrium
Disorganized endometrium overgrows and sloughs off
Irregular bleeding
Menopause
Premature ovarian insufficiency
                                Intermittent congestion of polyp blood supply
Transient ischemia and slight polyp necrosis
Altered growth factor production
Vascular dysregulation and leakier vessels
   Coagulopathies (e.g. von Willebrand disease)
Impaired hemostasis
Invasion of normal tissue
↓ Estrogen (hypoestrogenism)
Atrophy of endometrium and vulvovaginal tissue
Dry endometrial surfaces (↓ fluid to prevent friction)
                Endometrial carcinoma
Micro- erosions of epithelium
Inflammation
    Evidence unclear
      Heavier and more irregular bleeding
Spotting
Heavier and more irregular bleeding
Light bleeding and spotting
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published May 2, 2022 on www.thecalgaryguide.com

rhumatisme-psoriasique-pathogenese-et-resultats-cliniques

rhumatisme-psoriasique-pathogenese-et-resultats-cliniques

Psoriatic Arthritis: Pathogenesis and clinical findings
Auteur:
Payam Pournazari
Rédacteurs:
Yan Yu Scott Rapske Liam Martin* Traducteurs: Dianne Ganeswaran Stephen Williams Sylvain Coderre* * MD au moment de publication
Note:
L’arthrite rhumatoïde peut survenir avant ou après le psoriasis (la pathogenèse est la même).
Augmentation de l’activité ostéoclastique
Érosion de l’os sous- chondral mais formation de nouvelle osseuse ailleurs dans l’articulation
Radiographie: la périostite; les syndesmophytes, érosions, déformation du crayon en godets (les articulations IP), ankylose des articulations IP
   HLA B27, Cw6 et d’autres sous- types
Antécédents familiaux positifs
Activation des cellules T (mécanisme inconnu)
    Infiltration du tissu synovial de l’articulation par les cellules T et B, cellules tueuses naturelles et les macrophages
Production augmentée des molécules inflammatoires (les facteurs de nécrose tumorale [TNFs], les interleukines, etc.) qui agissent de façon systémique (dans tout le corps)
        Dans les tendons et le tissu conjonctif
Inflammation des enthèses et du tissu conjonctif provoque le gonflement
Dans la peau
Réaction immunitaire détruisant les structures de la peau et des ongles (Voir diapositive Psoriasis)
Le psoriasis en plaques sur la peau, dépressions punctiformes, onychorrhexie, taches d’huile, onycholyse, décoloration et hyperkératose
Angiogenèse et vascularite dans les vaisseaux sanguins de la membrane synoviale
Dans les articulations
Les cytokines inflammatoires stimulent les nocicepteurs locaux
1. Oligoarthrites - asymétriques
2. Arthrite des articulations IPD
3. Polyarthrite rhumatoïde -
symétrique
4. Atteinte axiale (raison
inconnue, probablement en raison de « biomécanique », « innervation », et
« vascularisation ».
5. Arthrite mutilante (grave et destructrice)
              L’enthésite
(Douleur/sensibilité à l’insertion du ligament dans l’os)
La dactylite (Inflammation de tout le doigt – les tissus mous et les articulations sont enflammés
   Légende:
 Physiopathologie
Mécanisme
Signe/Symptôme/Résultats de Laboratoire
 Complications
 Publié 10 November 2012 sur www.thecalgaryguide.com

acute-cholecystitis

Acute Cholecystitis: Pathogenesis and clinical findings
  Gallstone blocks the cystic duct, backing up bile into the gallbladder
Gallstones causing physical trauma to gallbladder wall
Irritation of adjacent diaphragm, stimulates phrenic nerve (C3-C5)
Activates stretch receptors of visceral peritoneum, stimulates foregut autonomic nerves (T5-T8)
Inflammatory mediator (i.e. prostaglandin) release by gallbladder and systemic inflammatory response
Thickened gallbladder wall on ultrasound (gold standard test)
On inspiration, the diaphragm pushes the gallbladder downward
Irritation of parietal peritoneum, stimulates somatic nerves
↑ Permeability of vessels with systemic inflammation, which leak
fluid from the blood into the interstitial space
Radiating pain to the back and right shoulder
Dull, diffuse abdominal pain referred to the epigastric region
Fever, nausea/vomiting, tachycardia
Positive Murphy’s sign (pain upon palpation of right upper quadrant [RUQ] on inspiration)
Persistent RUQ pain, abdominal guarding and peritoneal signs
Dehydration
Authors: Yan Yu, Vina Fan Reviewers: Dean Percy, Mirna Matta, Crystal Liu, Ben Campbell Maitreyi Raman* * MD at time of publication
                       Inflammation self-perpetuates
Irritation of inner gallbladder wall/mucosa
↑ Gallbladder lumen pressure
Intraluminal pressure exceeds arterial pressure
↓ Blood flow to gallbladder
Gallbladder ischemia
Local inflammation, loss of gallbladder mucosal integrity
Bacterial invasionàtransmural inflammation of gallbladder
       Without treatment, prolonged ischemia and inflammation of the gallbladder
      Gallbladder gangrene (20%)
Gallbladder perforation (20%)
 Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published August 4 2019, updated May 16 2022 on www.thecalgaryguide.com

clostridium-difficile-infection-pathogenesis-and-clinical-findings

Clostridium difficile (C. diff) Infection
Authors: Ryan Brenneis, Sravya Kakumanu Reviewers: Yoyo Chan, Sean Doherty, Vina Fan, Ben Campbell, Dr. Steve Vaughan*, Dr. Sylvain Coderre* * MD at time of publication
   Community exposure
Infected close contacts
Nosocomial exposure (most common)
Poor hand hygiene and sanitization of surfaces and medical equipment
Nosocomial risk factors
         Any antibiotic use
(Especially clindamycin, fluoroquinolones, penicillins, cephalosporins)
↑ Antibiotic resistant strains
Presence of pre-disposing risk factors
(Note: do not need to be present for infection)
Recent GI surgery
Chemotherapy that has antimicrobial and immunosuppressive effects
Usage of medications that reduce stomach acid (↑ pH)
   ↑ C. diff spores on surfaces and personnel
    Contact exposure
Environmental exposure
 to C. diff carriers
Inoculation of GI tract
Disruption of normal gut microbiome allowing C. diff overgrowth
Comorbidities
(>65 years old, cirrhosis, inflammatory bowel disease, enteral feeding, obesity)
     via fecal-oral route
  Clostridium difficile Infection of GI Tract
    Spores unaffected by antibiotics germinate post-antibiotic treatment
Infection recurrence
Pseudomembranous colitis on endoscopy
(colonic ulcerations potentially seen with severe infection)
Hypotension Acute kidney injury
Release of C. diff toxin A and B inactivates Rho and Ras GTPases in colonic epithelial cells (colonocytes)
(Rho and Ras GTPases control cytoskeletal dynamics and gene expression)
Cytoskeletal disorganization and arrest of RNA synthesis causes necrosis of colonocytes and triggers host immune response
Neutrophil chemotaxis and activation
↑ Inflammation of colon
Disruption of tight junctions between colonocytes
Release of fluid into intestinal lumen and inability of colon to reabsorb it
Toxic megacolon
Bowel perforation
Bloody stool
(<10% of patients)
Abdominal cramps
  Large bowel dilation from muscle paralysis
Inflammation and destruction of underlying smooth muscle
Breakdown of colonocyte cell membranes
Inflammation of visceral peritoneum
                 Volume depletion
Watery diarrhea: ↑ frequency, small volume
   Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published March 30, 2019, updated May 16, 2022 on www.thecalgaryguide.com

constatations-classiques-de-sclerose-en-plaques-sep-sur-irm-cerebrale

Constatations classiques de sclérose en plaques (SEP) sur IRM cérébrale
Auteurs: Evan Allarie Davis Maclean Viesha Ciura* Rédacteurs: Yan Yu* Traducteurs: Liana Martel Eddy Lang* * MD au moment de la publication
Infiltrations dans la région infratentorielle
Perte de la densité neuronale/axonale dans la région affectée, remplacée par une gliose au fil du temps
   Remarque : les constatations peuvent varier.
Les constatations présentées ici ne sont pas exhaustives, mais constituent certaines des zones les plus fréquemment mises en évidence par l'IRM du cerveau dans la SEP. Les sites les plus communément observés sont : les régions juxtacorticales, périventriculaires, infratentorielles, la moelle épinière et le nerf optique. Cependant, les lésions peuvent apparaître partout où il y a de la myéline dans le SNC.
L'inflammation et la destruction actives permettent au contraste de gadolinium de traverser la barrière hémato-encéphalique, ce qui peut être visualisé comme un marqueur de l'inflammation active dans la SEP
Lésion classique du nerf optique (CN II) en cas de névrite optique. Rehaussement par Gadolinium (↑ intensité du signal de la lésion après l’injection de gadolinium) sur cette image en T1 démontre une inflammation active
Image Credits: Dr. Viesha Ciura
Pour plus de détails sur la pathogenèse, voir la diapositive du Guide de Calgary: Multiple Sclerosis (MS): Pathogenesis and Clinical Findings
Inflammation dans le système nerveux central (SNC)
Les cellules T, les cellules B et les macrophages infiltrent le SNC
Infiltration se produit autour des veinsàinflammation locale Infiltrats périveineux autour des veines médullaires
(perpendiculaires aux ventricules)
             Inflammation et destruction de la barrière hémato-encéphalique ↑ liquide inflammatoire extravasculaire autour des lésions, qui apparaît hyperintense/brillant
      Lésions s’étendent autour des veines, créant les lésions ovoïdes charactéristiques
Plaques hyperintenses perpendiculaires périventriculaires classiques en T2/FLAIR suivant les veins médullaires – ‘Doigts de Dawson’
Lésion hyperintense du pédoncle cérebelleux moyen en T2/FLAIR dans une localisation classique de la SEP
            Légende:
 Physiopathologie
Mécanisme
Signe/Symptôme/Résultats de Laboratoire
 Complications
 Publié 25 octobre 2020 sur www.thecalgaryguide.com

pleural-effusions-pathogenesis-and-anterior-posterior-chest-x-ray-findings

Pleural Effusions: Pathogenesis and Anterior-Posterior Chest X-Ray Findings
 Decubitus chest x-ray view causes fluid to accumulate on ipsilateral side. (Most sensitive view for small effusions)
Excess fluid accumulation within pleural space that is gravity- dependent/free- flowing
Large accumulation of fluid pressing against lung tissue and mediastinum
Fluid layering (i.e. opacification) between lung and chest wall
Author: Sravya Kakumanu Reviewers: Reshma Sirajee, Tara Shannon *Stephanie Nguyen, *Shelley Spaner * MD at time of publication
     Transudative
pleural effusion
*See Pathogenesis and Clinical Findings of
Transudative Pleural Effusions slide
Exudative pleural
effusion
*See Pathogenesis and Clinical Findings of
Exudative Pleural Effusions slide
Fluid accumulation at bottom of pleural space in anterior- posterior chest x- ray
Lung atelectasis (lung collapse)
Fluid is denser than air and appears white on x-ray
↓ Pressure and occupied space in thoracic cavity
Meniscus sign (concave line above opacification)
Opacification of affected pleural space
Blunting of costophrenic angles
Diaphragmatic silhouette sign (sharp diaphragm edge obscured by fluid)
Mediastinal shift towards atelectic lung
                      Contralateral mediastinal shift (when no lung atelectasis)
 Contralateral tracheal deviation
        Pleural infections
↑ Inflammation at infection site à↑ permeability of parietal pleura endothelium
Bacterial invasion into pleural space from pulmonary parenchyma
↑ Migration of neutrophils and release of inflammatory cytokines in pleural space
Inflammation ↑ activation of coagulation cascade and ↓ fibrinolytic activity
↑ Deposition of fibrin clots/membranes within pleural space creating septations
Loculated pleural effusion/empyema (fluid stuck within septations within the pleural space)
Opacification does not follow gravity
Effusion does not move in decubitus view
Pleural thickening (whitening of lung perimeter from fibrin deposition - more visible on CT scan)
More rounded margins with chest wall/fissures
    Note: ~175-500mL of fluid may need to accumulate before being visible on an Anterior-Posterior Chest X-Ray
Image credit: https://radiopaedia.org/cases/pleural-effusion-7
   Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published May 22, 2022 on www.thecalgaryguide.com

gout-pathogenesis-of-x-ray-findings

Gout: Pathogenesis of X-Ray findings
Authors: Omer Mansoor, Nameerah Wajahat Reviewers: Reshma Sirajee, Tara Shannon *Stephanie Nguyen, *Shelley Spaner *MD at time of publication
   Hyperuricemia
See hyperuricemia slide for mechanism
↑ Uric acid concentration in blood leaks into joints as monosodium urate (MSU) crystals
Uric acid crystallizes due to lower temperature, change in pH, mechanical stress and other synovial factors
Crystals found more commonly in 1st MTP joint > ankle > wrist
MSU deposits within the bone producing
intra-osseus tophi (stone-like deposits of crystals)
MSU deposits in soft tissue and bone
↑ Inflammatory marker recruitment causes granulomatous inflammation
Osteoclasts (↑ bone resorption) are activated, and osteoblasts (↑ bone formation) are inhibited at
site of inflammation Marked localized bone loss
‘Rat Bite’ erosions
Intra-osseus bone lesion
(rare and non-specific for gout)
Joint effusions may be seen on x-ray as an early finding in the acute phase of gout
Soft Tissue Tophi
(appear cloudy and can hide other x-ray findings)
Normal bone density and preserved joint space
(until late in the disease)
Overhanging
sclerotic margins
Bone remodelled in an outward fashion to create the edge
Punched out lytic bone lesions Appear as circular ‘hole punches’ in bone
                        Note: Repeated episodes of gout must occur for 5 to 10 years before most joint changes may be seen on x-ray
Image credit: Radiopaedia
 Legend:
 Pathophysiology
Mechanism
Sign/Radiographic Findings
 Complications
Published June 7, 2022 on www.thecalgaryguide.com

Epilepsy Pathogenesis

Epilepsy: Pathogenesis
       Genetic susceptibility
Angelman syndrome, Fragile X syndrome, Tuberous sclerosis, neurofibromatosis
Chromosomal abnormalities cause abnormal neural patterns
Neural
Cerebral palsy, focal cortical dysplasia
Malformation of cortical development
Cerebrovascular
Intracranial hemorrhage, ischemic stroke
Other Acquired
Head trauma, cerebral infection (i.e., HSV1, VZV, cysticercosis)
Neoplasm
Metabolic
Inborn errors of metabolism
Inherited enzyme deficiencies
Neurodegenerative
Alzheimer’s Disease
Protein-rich plaque build-up and brain atrophy
            Inflammation and formation of scar tissue irritates neural tissue
Infiltration of mass, grey matter irritation
  Damage to the brain and subsequent alteration of neuronal circuitry
       Neurotransmitter imbalances (i.e. ↑ glutamate, ↓ GABA, ↓ serotonin, ↓ dopamine, ↓ noradrenaline)
Abbreviations:
• HSV1 – Herpes Simplex Virus 1 • VZV – Varicella Zoster Virus
↑ Inflammatory cytokines (e.g. interleukin-6, tumor necrosis factor-α)
Altered neurogenesis and gliosis
Ion channel and receptor dysfunction causes imbalance of ion channel charges
Authors: Keerthana Pasumarthi Christopher Li Reviewers: Negar Tehrani, Ephrem Zewdie, Ran (Marissa) Zhang, Carlos R. Camara-Lemarroy* * MD at time of publication
  Neurons fire in burst activity (referred to as paroxysmal depolarization shift) and often in groups (hypersynchrony)
Abnormal neural activities eventually create self-reinforcing circuits and transform neural network over time
       Injuries from
falls, bumps, operating heavy machinery
Involuntary contractions Loss of consciousness
Post-ictal confusion
Recurrent seizures
Sudden Unexplained Death of Epilepsy Patient (SUDEP)
Loss of airway reflexes
Aspiration of saliva or food contents
Aspiration Pneumonia
        Legend:
 Pathophysiology
Mechanism
 Sign/Symptom/Lab Finding
 Complications
Published July 5, 2022 on www.thecalgaryguide.com

Cervical Insufficiency

Cervical Insufficiency: Pathogenesis and clinical findings
      Cervical lacerations
Cervical loop Cone biopsy electrosurgical
procedure excision procedure
Cervical procedures
Post-procedure cervical re-modelling
↓ Structural collagen in extracellular matrix
Opened cervix allows for ↑ voiding of vaginal skin cells, bacteria, mucous and fluid
Fetal head moves lower into pelvis, exerting pressure on pelvic ligaments
Hysteroscopy dilatation and curettage
Antepartum Sterile intra-amniotic infection inflammation
Inflammatory cascade within cervix
↑ Local myometrial and cervical prostaglandins/ cytokines
↑ Collagenase
and leukocyte elastase increases break down of collagen
       Congenital abnormality of collagen synthesis
Ex. Ehler-Danlos syndrome
↑ Vaginal discharge
Change in color of vaginal discharge
Braxton-Hicks-like contractions
Menstrual-like cramps
Back ache
↓ Cervical tensile strength and rigidity
Cervical Insufficiency
Spontaneous dilation and thinning of cervix
Unknown cause
Prolapsed fetal membranes
Second trimester loss or preterm birth
Risk of recurrent second trimester loss or preterm birth
Authors: Kiera Pajunen
Reviewers: Ran (Marissa) Zhang Brianna Ghali Ingrid Kristensen* * MD at time of publication
                            Debris seen in amniotic fluid on U/S
Short cervix (<2.5cm
on trans-vaginal ultrasound)
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published July 5, 2022 on www.thecalgaryguide.com

exudative-pleural-effusions-pathogenesis-and-lab-findings

Exudative Pleural Effusions: Pathogenesis and Lab Findings
Authors: Sravya Kakumanu
Reviewers: Ben Campbell *Tara Lohmann * MD at time of publication
Chylothorax
Damage to thoracic duct
Leakage of lymphatic fluid into pleural space
      Pulmonary embolism
Clot obstructs blood flow to lung
Infarcted lung tissue
Lung infection (e.g. pneumonia, tuberculosis)
Lung infection signals inflammatory response
Systemic Lupus Rheumatoid Erythematosus (SLE) arthritis (RA)
Autoimmune antibodies localize to pleura
Pleural tumors (primary or secondary from metastatic cancer)
         Inflammatory cells migrate to affected site and release cytokines
↑ Permeability of pleural capillaries ↑ Fluid leakage across capillaries
Exudative Pleural Effusion
Cancer invades lymphatic drainage of pleural space (PS)
↓ Drainage of pleural fluid (PF) from pleural space
If infectious etiology
Tumor invasion = inflammatory response
            See Pleural Effusions: X-ray Findings and Physical Exam Findings of Lung Diseases slides
  ↑ Permeable pleural capillaries allow ↑ protein and cell leakage into pleural space
   If pleural tumour: Release of cancer cells into pleural space
Cancer cells on PF cytology 60-75% sensitive for malignancy
If rheumatoid arthritis:
Release of auto-antibodies into pleural space
Auto-antibodies initiate inflammatory response in pleural space
↑ Inflammatory cells have ↑ glucose metabolism in pleural space
Sterile PF with mildly elevated white blood cells, normal pH, normal glucose ↑ Inflammation at infection site damages endothelium of pleura
Pleural Infection Stage I: Simple Parapneumonic Effusion
        ↑ Inflammatory cells and bacterial cells in pleural space have ↑ glucose metabolism in pleural space
Bacterial invasion from infected parenchymaà pleural space
< 40mg/dL glucose in PF
↑ Activation of coagulation cascade and ↓ fibrinolytic activity
↑ Deposition of fibrin
clots/membranes within pleural
space creates loculated effusion
(compartmentalized effusion due to septations in pleural space)
       ↑ Production of
lactate
dehydrogenase
(LDH)
(LDH maintains NAD+ supply during ↑ glucose metabolism)
↑ CO2 production pH of PF < 7.20
      ↑ CO2 production pH of PF < 7.20
< 3.3mmol/L glucose in PF
Pleural Infection Stage II: Complicated Parapneumonic Effusion
Loculated effusion OR bacteria present OR ↓ pH + ↓ glucose
↑ Fibroblast proliferation creates thickened pleura ↑ Pus in pleural space Pleural Infection Stage III/Empyema: Loculated effusion and pus in pleural space
    PF/serum protein ratio ≥ 0.5
PF/serum LDH ratio ≥ 0.6
Light’s Criteria: Any criteria can be met to be an exudative pleural effusion
      PF LDH ≥ 2/3 upper limit of normal
    
Legend:
Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
Complications
Published August 9, 2022 on www.thecalgaryguide.com

presentation-of-sah

Subarachnoid Hemorrhage: Clinical Findings
Sudden bleeding into space surrounding the brain (for pathogenesis, see Subarachnoid Hemorrhage: Pathogenesis)
Authors: Jason An, M. Patrick Pankow Reviewers: Owen Stechishin, Dave Nicholl, Haotian Wang, Hannah Mathew, Ran (Marissa) Zhang, Yan Yu*, Cory Toth* * MD at time of publication
Bleed into subarachnoid space
Subarachnoid Hemorrhage (SAH)
   Posterior hypothalamus ischemia (↓ Blood flow and oxygen)
Red blood cell lysis from energy depletion or complement activation
Release of spasmogens (spasm inducing agents)
Cerebral vasospasm (narrowing of arteries from persistent contraction) ↓ blood flow
Cerebral ischemia
         Release catecholamines (hormones from the adrenal gland; e.g., epinephrine, norepinephrine)
↑ Intracellular calcium
Release of antidiuretic hormone
Antidiuretic hormone acts on the distal convoluted tubule and collecting duct in kidney to reabsorb water
Dilution of serum sodium
Hyponatremia (low blood sodium levels)
Release of epileptogenic (potential seizure causing agents) into cerebral circulation
Seizure
Products from blood breakdown in cerebral spinal fluid
Irritation of meninges (membranes surrounding the brain)
Aseptic meningitis (non-infectious inflammation)
Meningismus
(neck pain + rigidity)
Cerebral infarction (death of tissue)
Obstructs cerebral spinal fluid flow and absorption at subarachnoid granulations
Hydrocephalus (fluid build up in ventricles)
↓ Level of consciousness
Reduced cerebral blood flow
Dilation of cranial vessels to ↑ blood flow
Rapid ↑ internal carotid artery intracranial pressure
Refer to Increased Intracranial Pressure: Clinical Findings slide
Internal carotid artery
Pituitary ischemia
Hypopituitarism
[underactive pituitary gland, failing to produce 1+ pituitary hormone(s)]
Refer to hypopituitarism slides
                Myocardial disruption
Left ventricle dysfunction
↑ Pressure in left heart
Blood forced backwards into pulmonary veins
↑ Pulmonary blood pressure
Fluid from blood vessels leaks into lungs
Dysrhythmias (disturbance in rate/rhythm of heart) causing ↓ cardiac output
Syncope
(loss of consciousness due to ↓ blood flow to the brain)
Pulmonary edema
(excess accumulation of fluid in lung)
Cerebral hypoperfusion
Sudden ↑in blood volume
Vessels and meninges suddenly stretch
Thunderclap Headache (worst headache of patient's life)
                                  Shortness of breath
Reactive cerebral hyperemia (excess blood in vessels supplying the brain)
Artery specific findings:
Rapid ↑ internal carotid artery intracranial pressure
Middle cerebral artery
          Posterior communicating artery
Compression of outer CN3 Compression of inner CN3
Anterior communicating artery
                    Nonreactive pupil
Gaze palsy
(eye deviates down and out)
Diplopia
(double vision)
Ptosis
(drooping of upper eyelid)
Frontal lobe ischemia
Avolition
(complete lack of motivation)
Ischemia of motor strip pertaining to the legs
Bilateral leg weakness
Motor strip ischemia
Hemiparesis
(weakness/ inability to move one side of the body)
Ischemia of parietal association areas (brain regions integral for motor control of the eyes, the extremities and spatial cognition)
Aphasia
(impaired ability to speak and/or understand language)/ neglect
      Legend:
 Pathophysiology
Mechanism
 Sign/Symptom/Lab Finding
 Complications
Published July 1, 2014, updated August 10, 2022 on www.thecalgaryguide.com

thrombose-veineuse-profonde-soupconne-tvp-pathogenese-et-complications

Thrombose veineuse profonde soupçonné (TVP)
Auteurs: Dean Percy Yan Yu Rédacteur: Tristan Jones Julia Heighton Man-Chiu Poon* Lynn Savoie* Traducteurs: Sophia Shah Sylvain Coderre* *MD à la publication
La grossesse, contraceptifs oraux
Pathogénèse et complications
Activation plaquettaire
↑ formation de caillots
Maladies héréditaires
Anomalie congénitale de la coagulation (p.e. facteur V Leiden, facteur II muté, déficit en protéine S/C) ↑ capacité de coagulation
    Notes:
• TVP provoque une embolie pulmonaire, le thrombus artériel provoque un accident vasculaire cérébral
• TVP précédente est un facteur de risque de TVP courante
Malignité
Libération anormale de cytokines favorisant la coagulation
           Traumatisme/opération
Blessure systémiqueà activation de la cascade de coagulation
État d'hypercoagulabilité↑ capacité du sang à coaguler lors de la stimulation
Oestrogène favorise l'hypercoagulabilité, surtout en présence d'autres risques
     Hypertension Bactéries
Valve artificielle
Endommage les parois des vaisseaux
Adhèrent/ envahissent / vaisseaux
Surface anormale
Blessure de vaisseau
Expose le facteur tissulaire sur les cellules endommagées /sous-endothélium pour la liaison au FvW
Triade de Virchow
Stase veineuse
↓ débit sanguins au site de la lésion
vasculaireà concentration des facteurs de coagulation sanguine sur ce site
La graisse contient plus d'aromatase: ↑ d'androgènesà œstrogènes
mode de vie sédentaire, mauvais retour veineux
           Obésité
        Les caillots se produisent généralement dans les veines des jambes
1. Les veines larges et profondes permettent l'accumulation de sang
2. Retour veineux des jambes est contre la gravité
3. Valves dans les veines des jambes enclins au refoulement
↓ mouvement musculaire = ↓ débit sanguin
Fracture, immobilisation, alitement, long tour d’avion/ de véhicule
  Destruction de la valve veineuse par un caillot
Insuffisance veineuse
Les caillots empêchent le sang à retourner au cœur. L'accumulation de sang dans une jambe entraîne l’œdème unilatéral et l’inflammation veineuse (rougeur, chaleur, sensibilité)
Le caillot s'embolie dans les poumons
Thromboembole
-*Embolie pulmonaire (complication aiguë mortelle)
- Hypertension pulmonaire thromboembolique chronique
       Légende:
 Physiopathologie
Mécanisme
Signe/Symptôme/Résultats de Laboratoire
 Complications
Publié 15 juin 2019 sur www.thecalgaryguide.com

alcoholic-fatty-liver-disease-pathogenesis-and-clinical-findings

Alcoholic Fatty Liver Disease: Pathogenesis and clinical findings
Authors: Tara Shannon Reviewers: Ben Campbell, Yan Yu*, Samuel Lee* * MD at time of publication
↑NADH:NAD+ ratio in hepatic cells, a “high energy state” marker in the cell
When a cell is in a high energy state, metabolic processes that provide energy are inhibited
↓ β-oxidation (break down) of
triglycerides in the liver
  Abbreviations:
• CYP2E1 – Cytochrome P450, subtype
2E1
• NADH/NAD+ – Nicotinamide
Adenine Dinucleotide (in reduced and oxidized form respectively)
Ethanol upregulates hepatic cell fatty acid transporters
such as fatty acid transport protein 1 and 5 (FATP1,5)
↑ Fatty acid transporters in hepatic cells
↑ Uptake of free fatty acids from circulation into hepatic cells
Alcohol (ethanol) consumption
Ethanol is primarily metabolized in the liver by enzymes alcohol dehydrogenase (ADH) and CYP2E1 ADH converts ethanol into acetaldehyde & NADH CYP2E1 converts ethanol into acetaldehyde
Excessive alcohol consumedàthe liver cannot process ethanol and its metabolites quickly enoughà ethanol, acetaldehyde, and NADH accumulate in the liver and may enter the bloodstream
Acetaldehyde is a highly reactive and unstable molecule that can damage proteins, enzymes, and DNA
(Hepatic stellate cells (HSC) are sensitive to liver damage and release cytokines when activated)
Acetaldehyde activates HSCàHSC release tumor necrosis factor alpha (TNF-!), an inflammatory markerà↑ liver inflammation
                  Acetaldehyde upregulates the production of enzymes required for fatty acid synthesis
TNF-! stimulates fat production in liver cells
TNF-! enters circulation and stimulates adipose cells to break down, releasing triglyceride stores as free fatty acids
↑ Free fatty acids released into circulation
Liver biopsy shows liver inflammation
TNF-! causes the production of reactive oxygen species (unstable, highly reactive molecules)
Reactive oxygen species cause oxidative damage to hepatic cells’ mitochondria
Damaged mitochondria can no longer efficiently perform metabolic tasks
                ↑ Building blocks, catalysts, and direct stimulating factorsàliver cells ↑ fat production
↑ Lipogenesis in the liver (triglyceride production)
↑ Triglyceride accumulation in liver cells
Triglyceride-rich droplets in hepatic cells change the cells’ physical properties compared to normal liver cells, allowing imaging modalities to exploit these differences to detect lipid infiltration
          Fatty livers appear darker on CT scans
Liver biopsy shows triglyceride- rich droplets inside liver cells
Fatty livers appear brighter and with poorer visualization of structures (like bile ducts and vessels) on ultrasounds
Imaging such as ultrasound, CT, and MRI can visualize and quantify liver fat
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published August 21, 2022 on www.thecalgaryguide.com

patellar-tendon-rupture-pathogenesis-and-clinical-findings

Patellar Tendon Rupture: Pathogenesis and clinical findings
Author:
Molly Joffe
Reviewers:
Liam Thompson, Alyssa Federico, Tara Shannon, Gentson Leung* *MD at time of publication
    Chronic disease (e.g. renal failure, gout, rheumatoid arthritis, and diabetes)
Repetitive activities involving rapid knee flexion and extension
Tendon inflammation and micro-tearing
Fluoroquinolone (antibiotic class) use
  Smoking
Immobilization of knee
↓ Muscle and tendon flexibility and strength
Steroid use (including intraarticular injections)
Changes in collagen fibrils composing tendon (exact mechanism unknown)
        Disrupted blood supply to tendon and poor healing
  Compromised integrity and strength of the tendon
Sudden heavy load on flexed knee while foot is planted
  Quadriceps muscles contract while lengthening (eccentric contraction)àforce exceeds tendon strength
      Patellar tendon innervated by common peroneal (fibular) nerve
Nociceptors (pain receptors) within tendon activated by tendon rupture
Pain and tenderness below the patella
Collagen fibres in tendon tear
Tearing or popping sensation at time of injury
Patellar Tendon Rupture
Tendon/Ligament detaches from bony attachment on patella or tibial tubercle No connection of quadriceps to tibia via patellar tendon
↑ Force on tibial tubercle during tendon detachment
Avulsion fracture of tibial tubercle
Trauma to tendon ruptures surrounding blood vessels
Coagulation cascade and inflammatory mediators released at rupture site
    Quadriceps muscles unable to stabilize patellar tracking
Instability of knee joint
Quadriceps muscle tension pulls patella upwards with no resistance from patellar tendon
Patellar tendon cannot use distal attachment site for leverage
Knee extensor muscles unable to effectively contract
              Knee buckling
Abnormal gait
Patella alta
(high sitting patella) on X-ray
Palpable gap below the patella
Bruising below the patella
Swelling below the patella
      ↑ Risk of falls
Loss of patellar reflex
Inability to perform a straight leg raise (hip flexes and knee cannot remain straight)
 Hematoma (blood collection under the skin)
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published August 28, 2022 on www.thecalgaryguide.com

medial-epicondylitis-golfers-elbow-pathogenesis-and-clinical-findings

Medial Epicondylitis (Golfer’s Elbow): Pathogenesis and clinical findings
  Intrinsic Factors: age, body weight, nutrition, gender, anatomical variations, joint laxity, systemic disease, muscle weakness / imbalance, vascular perfusion
Micro-tears within flexor-pronator tendons initiating healing process: inflammation, proliferation, and remodeling (see acute wound healing slide)
Continued repetitive strains with inadequate recovery time between activitiesàhealing unable to meet tissue damage
Authors: Brett Lavender Reviewers: Alyssa Federico, Liam Thompson, Tara Shannon, *Gerhard Nikolaus Kiefer * MD at time of publication
Extrinsic Factors: activities involving repeated forceful use of the flexor-pronator muscle groups (sports including golf and baseball, activities such as shoveling, gardening or hammering nails)
         Ineffective revascularization of damaged tissue
MRI or Ultrasound Findings: Tendon thickening, partial tears, disrupted vascular distribution +/- edema of surrounding tissues
Disorganized collagen formation and scarring à↑ type III collagen (most common collagen involved with wound healing)
↑ Tendon thickening
Decreased tensile strength of tendon
Weakness
of the flexor-pronator muscle groups
↑ Nerve growth within damaged tissue (consequence of healing response)
Local nerves are compressed by thickened tendonànociceptors within tendon are activated
(Ulnar nerve passes through cubital tunnel adjacent to medial epicondyle)
Ulnar paresthesias
may result to structures innervated distal to the cubital tunnel
               Medial Epicondylitis
Tendinosis at the common flexor-pronator origin at the medial epicondyle of the humerus
Pain with passive wrist extension or resisted flexion
Tenderness
over the proximal wrist flexor-pronator muscles
Pain
localized to medial epicondyle
  Severity ranges from mild to severe based on the effect on patient activities
    Mild tendinopathy: patient continues most activities with minor pain
Moderate tendinopathy: patient continues some activities with modifications
Severe tendinopathy: patient’s daily activities are impacted by severe pain
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published August 30, 2022 on www.thecalgaryguide.com

ascending-cholangitis-pathogenesis-clinical-findings

Ascending Cholangitis: Pathogenesis and clinical findings
Authors: Brandon Hisey, Neel Mistry Reviewers: Alec Campbell, Vina Fan, Ben Campbell, Kelly Burak*, Eldon Shaffer* * MD at time of publication
Reflux of biliary contents into the vascular system (cholangiovenous reflux)
Bacteria ascends into biliary tract from duodenum via Sphincter of Oddi
     Gallstone in the common bile duct
Stricture of biliary/hepatic ducts
Biliary / pancreatic duct malignancy
Biliary obstruction (partial bile duct obstruction)
↓ Excretion of bilirubin into
bileà ↑ bilirubin in blood
↑ Serum bilirubin
Deposition of bilirubin in the skin and mucous membranes
Jaundice*†
Parasites in bile duct
(E.g. Clonorchis)
Complication of endoscopic retrograde cholangiopancreatography (ERCP)
        Bile accumulates in biliary tract
Bile duct dilation on ultrasound
Sludge (bile precipitants) impact bile ducts worsening obstruction
Obstruction & detergents in bile inflame ductal mucosa. Inflammation then spreads to adjacent structures
Stimulates phrenic (C3-C5) and foregut autonomic nerves (T5-T8)
Dull right upper quadrant pain*† radiating to back and right shoulder
↑ Intra-biliary pressure
Impaired forward flowà
↑ backflow of bile
Impaired bile secretion damages ductal epithelium of the biliary tract
Damaged ductal epithelium leaks ALP and GGT (enzymes) into blood
↑ Serum ALP and GGT
↑ Permeability of bile ductules
Reduced flushing of bile out to duodenum
               Inflammatory response triggered
Fever*† ↑ WBCs
Tachycardia Hypotension† Confusion† Oliguria
Bacterial translocation from biliary tract into blood
Bacteremia
Massive systemic inflammatory response
Septic Shock
(see Distributive Shock slide)
                   * Charcot’s triad ~20% of cases | † Reynolds’ pentad ~7% of cases
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
First published November 18, 2015; Updated August 31, 2022 on www.thecalgaryguide.com

acute-lower-gi-bleeds-pathogenesis-and-clinical-findings

Acute Lower GI Bleeds: Pathogenesis and Clinical Findings
      Diverticulosis
Formation of diverticula (outpouchings of the bowel wall)
Intestinal vessels are stretched over the domes of the diverticula
Angiodysplasia
Formation of
dilated, thin- walled vessels in GI tract mucosa/ submucosa
Colorectal Cancer
Infectious Colitis
Invasion of bacteria and/or bacterial toxins into intestinal wall
Cell damage and cell death
Sloughing off of intestinal epithelial cells
Ischemic Colitis
↓ Blood flow to a portion of the colon
Lack of oxygen delivery to colon wall
Necrosis (cell death) in the colon wall
Inflammatory Bowel Diseases (Note: these are chronic diseases and rarely present with acute lower GI bleed)
                    New, extremely friable blood vessels develop within the tumor
Malignant tissue invades the colon wall and disrupts colonic blood vessels
Crohn Disease
Immune- mediated full thickness inflammation of bowel wall
Ulcer formation and disruption of intestinal vessels
Ulcerative Colitis
Recurrent immune- mediated inflammation of colon mucosa
    Authors:
Miranda Schmidt Illustrator:
Mizuki Lopez Reviewers:
Vina Fan,
Ben Campbell, Kerri Novak*
* MD at initial time of publication
Acute Lower GI Bleed
↑ Risk for vessel damage and rupture
       Blood travels rapidly through GI tract
Hematochezia
(bright red blood per rectum)
May result in significant blood loss
Blood from the small intestine or right colon travels a longer distance through the GI tract
Bacteria in the GI tract has time to oxidize hemoglobin in the blood
Oxidization makes blood a darker color
Melena (rare in a lower GI bleed) (tarry black stool)
      Inferior Vena Cava
Diaphragm
Esophagus
Loss of red
blood cells results in a loss of hemoglobin (a component of red blood cells)
Fluid from the extravascular space moves into the blood vessels to maintain vascular volume
Fluid is administered in a healthcare setting to compensate for blood loss
Hypovolemic Shock (rare in a lower GI bleed) (↓ Oxygen delivery to tissues due to low blood volume)
See “Hypovolemic Shock” slide for signs and symptoms
       Lower GI Bleeds
are intra-luminal GI tract bleeds that occur anywhere distal to the ligament of Treitz (transition between duodenum and jejunum)
After 24 hours, the addition of fluid to the intravascular space dilutes hemoglobin in the blood
Normocytic Anemia
       Duodenum
Ligament of Treitz
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published August 31, 2022 on www.thecalgaryguide.com

lateral-epicondylitis-tennis-elbow-pathogenesis-and-clinical-findings

Lateral Epicondylitis (Tennis Elbow): Pathogenesis and clinical findings
Authors: Brett Lavender Reviewers: Alyssa Federico, Liam Thompson, Tara Shannon Gerhard Nikolaus Kiefe* * MD at time of publication
  Extrinsic Factors: activities involving repeated forceful use of the extensor-supinator muscle groups (sports including tennis and squash, activities such as painting, carpentry or using certain hand tools)
Intrinsic Factors: age, body weight, nutrition, gender, anatomical variations, joint laxity, systemic disease, muscle weakness / imbalance, vascular perfusion
   Micro-tears within extensor-supinator tendons initiating healing process: inflammation, proliferation, and remodeling (see acute wound healing slide)
 Continued repetitive strains with inadequate recovery time between activitiesàhealing unable to meet tissue damage
     Ineffective revascularization of damaged tissue
MRI or Ultrasound Findings:
Tendon thickening, partial tears, disrupted vascular distribution +/- edema of surrounding tissues
Disorganized collagen formation and scarringà↑ type III ↑ Nerve growth within damaged tissue collagen (most common collagen involved with wound healing) (consequence of healing response)
 ↑ Tendon thickening
Decreased tensile strength of tendon
Weakness
of the extensor- supinator muscle groups
Lateral Epicondylitis
Tendinosis at the common extensor-supinator origin at the lateral epicondyle of the humerus
Local nerves are compressed by thickened tendon ànociceptors within tendon are activated
Pain with passive wrist Pain localized to flexion or resisted extension lateral epicondyle
          Tenderness over the proximal wrist extensor-supinator muscles
  Severity ranges from mild to severe based on the effect on patient activities
     Mild tendinopathy: patient continues Moderate tendinopathy: patient continues Severe tendinopathy: patient’s daily most activities with minor pain some activities with modifications activities are impacted by severe pain
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published September 25, 2022 on www.thecalgaryguide.com

quadriceps-tendon-rupture-pathogenesis-and-clinical-findings

Quadriceps Tendon Rupture: Pathogenesis and clinical findings
Author:
Molly Joffe
Reviewers:
Liam Thompson, Alyssa Federico, Tara Shannon, Gentson Leung* *MD at time of publication
     Repetitive activities involving, rapid, active flexion and extension of the knee
Quadriceps tendon inflammation and micro-tearing
Immobilization of leg/knee
Steroid use (including intraarticular injections)
       Quadricep muscles atrophy
↓ Quadricep muscle strength
Smoking
Chronic disease (e.g. renal failure, gout, rheumatoid arthritis, and diabetes)
Fluoroquinolone (antibiotic class) use
Changes in collagen fibrils composing tendonàimpaired tendon strength (exact mechanism unknown)
Quadriceps tendon shortens
↓ Quadricep tendon flexibility
Disrupted blood supply to quadriceps tendon and poor healing
        Compromised integrity of the quadriceps tendon and ↓ strength
Sudden heavy load on flexed knee while foot is plantedàquadriceps muscles contract while lengthening (eccentric contraction)
Force on quadriceps tendon exceeds its strength
Quadriceps Tendon Rupture
Tendon detaches from bony attachment on patella
        ↑ Force on patella during quadriceps tendon detachment
Collagen fibres in quadriceps tendon tear
No connection of quadriceps to tibia via quadriceps tendon
Patellar tendon tension pulls patella downwards with no resistance from quadriceps muscles
Quadriceps tendon innervated by common peroneal (fibular) nerve
Nociceptors (pain receptors) within tendon activated by tendon rupture
Pain and tenderness above the patella
Trauma to tendon ruptures blood vessels
Coagulation cascade and inflammatory mediators released at rupture site
     Quadriceps muscles unable to stabilize patellar tracking
Knee joint instability
Patella baja (low sitting patella) on X-ray
Palpable gap above the patella
Loss of patellar reflex
Patellar tendon cannot use proximal attachment site for leverage
Knee extensor muscles unable to effectively contract
Inability to perform a straight leg raise (hip flexes while knee cannot remain straight)
Bruising above the patella
Swelling above the patella
                 Avulsion fracture of patella
Tearing or popping
sensation at time of injury
Knee buckling
Abnormal gait
   ↑ Risk of falls
Hematoma (blood collection under the skin)
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published October 2, 2022 on www.thecalgaryguide.com

felty-syndrome

Felty Syndrome: Pathogenesis and clinical findings
Authors: Anjali Arora Reviewers: Ben Campbell, Liam Martin*, Yan Yu* * MD at time of publication
Longstanding chronic Rheumatoid Arthritis
Rheumatoid Arthritis (RA): a disease of systemic autoimmunity – refer to Rheumatoid Arthritis slides for detailed pathogenesis
   Genetic susceptibility
Presence of a specific type of Human Leukocyte Antigen (HLA-DR4), a surface protein on immune cells that is known to be associated with or worsen autoimmune activity
Idiopathic autoimmunity
Neutrophil activation, apoptosis and adherence to endothelial cells in the spleen
Felty Syndrome
     An uncommon extraarticular manifestation of seropositive Rheumatoid Arthritis characterized by the triad of splenomegaly, neutropenia and arthritis
     Immunologic stress leads to proliferation of white pulp in spleen (responsible for initiating immune responses to foreign antigens)
Splenomegaly
↑ RBC and platelet sequestration within the spleen leading to a quantitative shortage of these cells in circulation
Enlarged spleen compresses stomach
Loss of appetite
Complex autoimmune phenomena lead to ↓ neutrophil production and ↑ removal from the circulating pool
Neutropenia
For a detailed mechanism, refer to Neutropenia slides
↓ Circulating neutrophils leads to ↑ susceptibility to infections
Severe, untreated Rheumatoid Arthritis will lead to systemic inflammation
See Rheumatoid Arthritis slides for detailed mechanism
Chronic erosion of synovial membranes in joints commonly affecting the hands, wrists and knees
Arthralgia (symmetric polyarthritis, usually with small joint distribution)
               Thrombocytopenia
Refer to Thrombocytopenia slides for signs and symptoms
Normocytic Anemia Refer to Normocytic
Anemia slide for signs and symptoms
Host becomes infected
Fever
Recurrent infections
       Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
Published October 18, 2022 on www.thecalgaryguide.com

microscopic-colitis-pathogenesis-and-clinical-findings

Microscopic Colitis: Pathogenesis and clinical findings Genetic
Author: Tony Gu, Kayleigh Yang Reviewers: Yoyo Chan, Vina Fan, Ben Campbell, Dr. Edwin Cheng* * MD at time of publication
Release of cytokines (e.g. interferon gamma (IFNγ))
Downregulation of intestinal tight junction proteins
↓ Epithelial barrier function
↑ Transmucosal permeability
Infiltration of bacteria or antigens
    ↑ Expression of nuclear factor kappa B (NF-kB) in colonic epithelial cells
NF-kB increases transcriptional activity in specific genes
↑ Inflammatory molecules
(histamine, prostaglandins, and nitric oxide) in epithelium
predisposition Unknown trigger
↑ Transforming growth factor beta 1 (TGF- β1)
and vascular endothelial growth factor (VEGF) expression in colonic epithelial cells
Equilibrium shift to more production of collagen
(fibrinogenesis) than breakdown (fibrinolysis)
Drug exposure (e.g., non-
steroidal anti- (+)
inflammatory drugs (NSAIDs), proton pump inhibitors (PPIs))
            Inflammation and accumulation of immature subepithelial collagen matrix
Subtype: Collagenous Colitis
Intestinal mucosal inflammation
Subtype: Lymphocytic Colitis
    characterized by a colonic subepithelial characterized by intraepithelial
collagen band on histology
lymphocytic infiltrates on histology
Microscopic Colitis
       Intestinal epithelial damage
Malabsorption of nutrients (including bile acids)
Weight loss
↑ Intestinal transmucosal permeability
Water drawn into lumen through osmosis
Ions and water back leak into intestinal lumen
Non-bloody watery diarrhea
Dehydration and electrolyte disturbances
Continuous release of inflammatory mediators
Peripheral afferent nerve sensitization
Visceral hypersensitivity
Abdominal pain
           Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published December 8, 2018, Updated October 18, 2022 on www.thecalgaryguide.com

copd-findings-on-posterior-anterior-and-lateral-chest-x-ray-findings

Authors: Shayan Hemmati COPD: Findings on Posterior-Anterior and Lateral Chest X-ray Findings Reviewers: Reshma Sirajee, Sravya Kakumanu, Tara Shannon, *Stephanie Nguyen, *MD at time of publication
   Chronic Obstructive Pulmonary Disease (COPD) See Pathogenesis of COPD Slides
Findings of COPD may not be apparent on chest X-ray early within the disease
Lung inflammation causes proteolytic destruction of lung parenchyma (part of lungs involved in gas exchange)
Alveolar walls are damagedàformation of fragile enlarged sacs of air called bullae
Rupture of bullae due to weak alveolar walls
Air leaves alveola and enters pleural space
Pneumothorax
See Primary Spontaneous Pneumothorax slide
Vasoconstriction of pulmonary arteries to shunt blood to better ventilated areas
Pulmonary hypertension
See Pathogenesis of Pulmonary Hypertension slide
↑ Pressure within pulmonary arteries
Enlargement of pulmonary arteries
Hilar enlargement Lung hyperinflation
(defined as 10 posterior ribs above the diaphragm level on the midclavicular line)
↑ Lucency of the lung
field, ↓ lung markings
(unless COPD is of chronic bronchitis phenotype)
↑ Size of retrosternal airspace
↑ Anterior – posterior (AP) diameter (“Barrel Chest”)
Flattening of hemidiaphragms
12 3 4
5 6
7
8
9 10
    Poorly ventilated areas of lung
Hyperinflation as air becomes trapped in damaged lungs
↑ Total volume of air within the chest
                Airway fibrosisà bronchial wall thickening (yellow arrow heads)
Air is less dense than soft tissue and vessels so it appears black
↑ Pressure anteriorly on sternum
↑ Pressure posteriorly on thoracic wall
↑ Pressure posteriorly on diaphragms
PA
                Bullae sometimes visible on X-ray (focal areas of lucency with spread out vascular markings)
     Image credit: Bronchial wall thickening image courtesy of Dr. Ashley Davidoff
Image credit: Radiopaedia
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published October 26, 2022 on www.thecalgaryguide.com

Granulomatosis with Polyangiitis Pathogenesis

Granulomatosis with polyangiitis: Pathogenesis
   Author:
Oswald Chen
Reviewers:
Ben Campbell
*Liam Martin
* MD at time of publication
Drugs
Thiol- and hydrazine-containing medications (e.g., hydralazine, propylthiouracil, allopurinol)
Environmental exposures
Silica dust, cigarette smoke, infections (Staphylococcus aureus)
Genetic factors
Alpha-1 antitrypsin deficiency, proteinase 3 gene mutation
  ↑ Production of cytokines and antineutrophil cytoplasmic antibodies (ANCAs) (mechanism unknown) Cytokines bind to endothelial cells (that line blood vessels) and neutrophils, priming them
Proteinase 3 (PR3), an enzyme that degrades extracellular matrix proteins, migrates from neutrophil granules to neutrophil cell surface
      Circulating ANCAs bind to PR3 on neutrophils
PR3 stimulates maturation of dendritic cells in lungs
Dendritic cells present antigen (PR3) to naïve CD4+ T cells in peripheral lymph nodes
T cells differentiate into type 1 and type 17 helper T cells (Th1 and Th17 cells)
Th1 and Th17 cells secrete cytokines (interferon γ (INF-γ) and tumor necrosis factor α (TNF-α)) in lungs
Secreted cytokines trigger macrophage maturation
Formation of granulomas (giant cells with central necrosis surrounded by plasma cells, lymphocytes, and dendritic cells) primarily in lungs and upper airways
    ANCA-activated neutrophils release proinflammatory cytokines, attracting more neutrophils to endothelium (blood vessel wall)
ANCA-activated neutrophils undergo firm adhesion to endothelium
     ANCAs stimulate ↑ secretion of proteolytic enzymes and reactive oxygen species from neutrophils
Endothelial damage and tissue injury
    Granulomatosis with polyangiitis (GPA)
ANCA-associated vasculitis affecting medium and small-sized arteries, associated with necrotizing granulomas
Constitutional symptoms with involvement of multiple organ systems
 (see slide on clinical findings)
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published December 4, 2022 on www.thecalgaryguide.com
    
Granulomatosis with polyangiitis: Clinical findings Inflammation-mediated endothelial damage and granuloma formation
(see slide on pathogenesis)
Granulomatosis with polyangiitis (GPA)
Author:
Oswald Chen
Reviewers:
Ben Campbell
*Liam Martin
* MD at time of publication
 ANCA-associated vasculitis affecting medium and small-sized arteries, associated with necrotizing granulomas
    Constitutional symptoms
Fever, unintentional weight loss, night sweats, arthralgias
Skin involvement
Inflammation of cutaneous vessels
Systemic inflammation obstructing blood flow, with granulomatous lesions primarily in upper airways and lungs
Ear, nose, and throat involvement
↑ C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) (markers of inflammation)
Necrotizing granulomas on biopsy of affected tissue
Positive PR3-ANCA/c-ANCA blood test (antibodies present in ~90% of patients, described in GPA Pathogenesis slide)
             Renal involvement
Inflammation of renal vessels
Rupture of basement membrane (layer that filters blood from glomerular capillaries into Bowman’s capsule)
Pauci-immune glomerulonephritis (see Nephritic Syndrome slide)
Rapidly progressive glomerulonephritis
Eye involvement
Inflammation of ocular tissue
Conjunctivitis
Scleritis/ episcleritis (painful red eye)
Lower respiratory tract involvement
Inflammation of pulmonary vessels
                 Vessel occlusion and ischemia
Skin necrosis
Vessels burst and blood pools under skin
Round and retiform (net- like) palpable purpura of lower extremities
Granulomatous destruction of nasal cartilage
Collapse of nasal bridge
Saddle nose deformity
Inflammation of paranasal sinus and nasal cavity vessels
↓ Perfusion of lungs
Dyspnea
Damage to interstitial capillaries
Hemoptysis
Diffuse alveolar hemorrhage
              Rhinitis/ sinusitis
Granulomatous obstruction of eustachian tube
Otitis media (see Otitis Media slide)
    Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published December 4, 2022 on www.thecalgaryguide.com

Granulomatosis with polyangiitis: Clinical findings

Granulomatosis with polyangiitis: Clinical findings Inflammation-mediated endothelial damage and granuloma formation
(see slide on pathogenesis)
Granulomatosis with polyangiitis (GPA)
Author:
Oswald Chen
Reviewers:
Ben Campbell
*Liam Martin
* MD at time of publication
 ANCA-associated vasculitis affecting medium and small-sized arteries, associated with necrotizing granulomas
    Constitutional symptoms
Fever, unintentional weight loss, night sweats, arthralgias
Skin involvement
Inflammation of cutaneous vessels
Systemic inflammation obstructing blood flow, with granulomatous lesions primarily in upper airways and lungs
Ear, nose, and throat involvement
↑ C-reactive protein (CRP) and erythrocyte sedimentation rate (ESR) (markers of inflammation)
Necrotizing granulomas on biopsy of affected tissue
Positive PR3-ANCA/c-ANCA blood test (antibodies present in ~90% of patients, described in GPA Pathogenesis slide)
             Renal involvement
Inflammation of renal vessels
Rupture of basement membrane (layer that filters blood from glomerular capillaries into Bowman’s capsule)
Pauci-immune glomerulonephritis (see Nephritic Syndrome slide)
Rapidly progressive glomerulonephritis
Eye involvement
Inflammation of ocular tissue
Conjunctivitis
Scleritis/ episcleritis (painful red eye)
Lower respiratory tract involvement
Inflammation of pulmonary vessels
                 Vessel occlusion and ischemia
Skin necrosis
Vessels burst and blood pools under skin
Round and retiform (net- like) palpable purpura of lower extremities
Granulomatous destruction of nasal cartilage
Collapse of nasal bridge
Saddle nose deformity
Inflammation of paranasal sinus and nasal cavity vessels
↓ Perfusion of lungs
Dyspnea
Damage to interstitial capillaries
Hemoptysis
Diffuse alveolar hemorrhage
              Rhinitis/ sinusitis
Granulomatous obstruction of eustachian tube
Otitis media (see Otitis Media slide)
   
Legend:
Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
Complications
Published December 4, 2022 on www.thecalgaryguide.com

Erythema multiforme (EM): Pathophysiology and clinical findings

Erythema multiforme (EM): Pathophysiology and clinical findings Infectious agents (cause >90% of EM)
Herpes simplex virus types 1 and 2
Mononuclear cells transport herpes simplex virus DNA fragments to keratinocytes in epidermis
Herpes simplex virus-specific CD4 T-cells of the immune system are recruited to the epidermis
CD4 T-cells release interferon-gamma (a pro-inflammatory cytokine) in response to viral antigens in keratinocytes in the epidermis
Lymphocytes infiltrate along the dermal-epidermal junction of the skin Lymphocytes create an inflammatory and edematous environment Keratinocytes irreversibly degenerate and undergo apoptosis and necrosis Formation of inflammatory epidermal lesions
Authors: Ayaa Alkhaleefa Reviewers: Ben Campbell Damilola Omotajo Jori Hardin* *MD at time of publication
  Drugs (a rare cause of EM)
    Non-steroidal anti- inflammatory drugs i.e. diclofenac sodium
Antibiotics i.e. TMP- SMX
Topical 5% Imiquimod, used to treat actinic keratoses
     Activate tumour-necrosis-factor- alpha, an inflammatory cytokine
    Skin
Epidermal layer
Dermal-Epidermal
Junction Dermal layer
Triggering agent (infection, drugs)
Immune-mediated inflammation and epidermal tissue damage
Erythema multiforme
           Inflammation ↑ capillary blood flow in dermis
Red colouring of skin in the area
Centre of lesions
contain a necrotic core
Inflammatory edema causes margins around necrosis to form a raised and pale ring
Inflammation stimulates cutaneous itch receptors
       Erythematous, papular, target-shaped, pruritic lesions on mucosal and acral (palms of hands, soles of feet) sites and face
   Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published November 29, 2022 on www.thecalgaryguide.com

rheumatoid-pneumoconiosis-caplans-syndrome-pathogenesis-and-clinical-findings

Rheumatoid Pneumoconiosis (Caplan’s Syndrome): Pathogenesis and clinical findings
Authors: Christopher Li Keerthana Pasumarthi Reviewers: Daniela Urrego, Ben Campbell, Liam Martin* * MD at time of publication
  Dust-exposed occupation (e.g. coal, asbestos, silica)
↑ Accumulation of dust particles in lungs
Dust (e.g. silica) mobilizes from the lungs to other organs such as the kidneys, lymph nodes, and spleen
Accumulation of silica in other organs which activate the immune system to secrete cytokines, chemokines, and lysosomal enzymes
Activation of antigen- presenting and antibody- producing cells
Increased risk to produce autoantibodies
Increased risk for autoimmune conditions such as rheumatoid arthritis
See slide: Rheumatoid Arthritis (RA): Extra- articular manifestations for mechanism of systemic inflammation
See slide Pneumoconioses: Pathogenesis and clinical findings for mechanism
            Note: Rheumatoid arthritis may precede lung nodules or develop later in the course
Rheumatoid arthritis
Systemic autoimmune inflammatory disease that mainly involves synovial joints
(+)
Alveolar macrophages and neutrophils ingest dust particles
↑ Inflammation and cytokine production in pulmonary parenchyma (e.g. interleukin-1, tumor necrosis factor-α)
Rheumatoid pneumoconiosis
Interstitial lung disease, with characteristic nodules, resulting from the interaction between dust inhalation and rheumatoid arthritis inflammation
Cytokines recruit histiocytes, neutrophils, lymphocytes, and fibroblasts to the area to produce a zone of inflammation around the dust-containing cell
Necrosis and apoptosis of dust-containing cells, macrophages, and surrounding collagen
Necrotic cells are digested by new macrophages to restart the process
Production of nodules that contain a central necrotic area surrounded by alternate layers of dust and necrotic tissue (Caplan nodules)
          Hyperactive immune response to foreign materials in lungs
Multiple concentric rings of dust seen histologically on light microscopy
0.5-5 cm rounded opacities on chest X-ray in periphery of lungs
See slide on
Pneumoconioses
for symptoms and pulmonary function test results
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published November 17, 2022 on www.thecalgaryguide.com

Acute Liver Failure: Pathogenesis and clinical findings

Acute Liver Failure: Pathogenesis and clinical findings
Authors: Juliette Hall Reviewers: Vina Fan, Ben Campbell, Mayur Brahmania* * MD at time of publication
      Acetaminophen Overdose
Accumulation of toxic NAPQI (a metabolite of acetaminophen)
NAPQI binds hepatocellular proteins
(see Acetaminophen Overdose: pathogenesis and clinical findings slide)
Drug-induced liver injury
Metabolism of drugs by the liver can produce reactive drug metabolites
Intracellular stress, mitochondrial injury, or immune response
Viral Hepatitis (i.e. HAV, HBV, HEV, HSV)
Acute infection or infection flare provokes an immune response against infected hepatocytes
Autoimmune Hepatitis
Autoimmune antibodies attack hepatocytes (see Auto-immune Hepatitis (AIH) slide)
Ischemia (i.e. from shock)
↓ O2 delivery to the liver
Hepatocellular hypoxia
Wilson’s Disease
Heritable mutation in the ATP7B gene
↓ Biliary excretion of copper
            Hepatic copper accumulation injures hepatocytes (see Wilson’s Disease slide)
       Accelerated rate of hepatocellular necrosis or apoptosis
 Hepatocyte death exceeds regeneration such that liver function is compromised within a short amount of time
Acute Liver Failure
An illness of <26 weeks duration in the absence of pre-existing cirrhosis, characterized by INR ≥1.5 and evidence of altered mentation (hepatic encephalopathy)
       Injured hepatocytes leak hepatic enzymes (AST, ALT, GGT) into circulation
↑ Liver enzymes
Hepatocellular inflammation
Stimulation of foregut
autonomic nerves
Right upper quadrant pain
↓ Toxin metabolism
Toxins build up and activate microglial cells (brain macrophage)
Oxidative stress and cerebral edema
Hepatic encephalopathy
Characteristic set of neuropsychiatric symptoms (see Hepatic Encephalopathy slide)
↓ Hepatocellular function and number
↓ Complement protein synthesis
↓ Ability to clear immune complexes and activate B cells
Accumulation of pigmented bilirubin
        ↓ Synthesis of coagulation factors
↑ INR
↓ Conjugation of bilirubin by the liver and ↓ transport into bile for excretion
            ↑ Serum bilirubin
Jaundice
Infection
 Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
Published November 15, 2022 on www.thecalgaryguide.com

Acute and Chronic Gastritis: Pathogenesis and clinical findings

Acute and Chronic Gastritis: Pathogenesis and clinical findings
Author: Oswald Chen
Reviewers: Vina Fan, Ben Campbell, Eldon Shaffer* * MD at time of publication
   Infectious
Helicobacter pylori (most common)
Noninfectious
NSAIDs, alcohol, gastric reflux
Stress
Critical illness, trauma
Gastric ulceration (see Peptic Ulcer Disease slide)
   ↓ Gastric mucosal defense
(↓ secretion of prostaglandins and mucus)
Acid erodes gastric mucosa
Inflammatory cells (primarily neutrophils) infiltrate site of injury
Acute Gastritis
Acute inflammation of gastric mucosa
Inflammatory cells (primarily lymphocytes and plasma cells) accumulate in gastric mucosa
Autoimmune Metaplastic Environmental Metaplastic Atrophic Gastritis (AMAG) Atrophic Gastritis (EMAG)
Atrophic Gastritis
Chronic inflammation of gastric mucosa
Inflammatory cells destroy gastric glandular epithelial cells
Gastric mucosal atrophy and metaplasia (replacement of gastric mucosal cells with intestinal epithelial cells, commonly goblet cells)
Hypochlorhydria
(parietal cell loss → ↓ hydrochloric acid secretion)
↓ Iron absorption
Iron deficiency anemia
(see Iron Deficiency Anemia slide)
Activation of chemoreceptors and mechanoreceptors
Activation
of visceral nociceptors
Visceral afferents stimulate chemoreceptor trigger zone of medulla
Nausea/ vomiting
      Autoimmune
Associated with human leukocyte antigens HLA-B8 and HLA-DR3, which are proteins expressed on immune cell surface
Antibodies destroy parietal cells and intrinsic factor in gastric body and fundus
Epigastric pain (typically burning or gnawing) Dyspepsia (abdominal discomfort after
eating, often with early satiety and bloating)
                      Parietal cell loss →
↓ intrinsic factor →
↓ vitamin B12 absorption
Vitamin B12 deficiency
(see Vitamin B12 Deficiency slide)
Macrocytic Peripheral anemia neuropathy
↓ Gastric acidity leads to ↓ inhibition of G cells in antrum Hypergastrinemia (↑ gastrin release from G cells)
Gastrin binds to cholecystokinin-B (CCK-B) receptors on parietal and enterochromaffin-like (ECL) cells of gastric body
↑ CCK-B signaling leads to ↑ cell proliferation and ↓ cell death (↓ apoptotic activity)
Hyperplasia (↑ number of cells)
Low-grade dysplasia (disordered growth of epithelium) High-grade dysplasia
          Carcinoid tumor (0.7% of cases) Malignancy arising from ECL cells
Gastric adenocarcinoma (0.3% of cases) Malignancy arising from gastric epithelial cells
    Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published November 15, 2022 on www.thecalgaryguide.com

Achalasia: Findings on Fluoroscopy with Barium Swallow

Achalasia: Findings on Fluoroscopy with Barium Swallow
Authors: Nameerah Wajahat, Omer Mansoor, Aly Valji Reviewers: Tara Shannon, Reshma Sirajee, Stephanie Nguyen* *MD at time of publication
 Barium swallow performed (patient swallows barium contrast, a radiopaque agent, which outlines the gastro-intestinal (GI) tract during fluoroscopy)
    Primary Achalasia
(idiopathic)
Secondary (Pseudo) Achalasia
(due to another disease process such as tumor, Chaga’s disease, diabetes mellitus)
Achalasia
Secondary disease processes can cause denervation or nerve dysfunction of esophageal myenteric plexus (EMP). Tumors can obstruct the lumen and infiltrate the EMP.
Visual differences from primary achalasia
Look for fixed abnormalities and/or mucosal irregularities and shouldering in the setting of a tumor
   Nerves controlling the lower esophagus are damaged, making it difficult for food and liquids to pass the esophagus
See “Achalasia: Pathogenesis and Clinical Findings” for full pathogenesis of primary and secondary achalasia
Inflammation and degeneration of nerves in the wall of esophagus
Dysfunction of the esophageal myenteric plexus (network of nerves in the GI tract responsible for peristalsis and sphincter relaxation)
Incomplete relaxation of the Loss of peristalsis in distal lower esophageal sphincter (LES) esophagus
          Barium has difficulty passing through the LES to the stomach
Barium outlines the GI tract, following esophageal abnormalities
Normal findings
Esophageal Dilatation
Occurs upstream of narrow LES
Bird Beak Sign / Rat Tail Sign
Smooth narrowing of the distal esophagus
Incomplete LES relaxation
        Barium Swallow may be normal in some patients with achalasia. Esophageal manometry (gold standard for diagnosis) or upper endoscopy can be considered instead.
Image Source: Radiopaedia
 Legend:
 Pathophysiology
Mechanism
Radiographic Findings
 Complications
Published November 9, 2022 on www.thecalgaryguide.com

popliteal-bakers-cyst-pathogenesis-and-clinical-findings

Popliteal (Baker’s) Cyst: Pathogenesis and clinical findings
Authors: Liam Thompson, Megan Ure Reviewers: M. Patrick Pankow, Tara Shannon, Reza Ojaghi, Usama Malik, Dr. Gerhard Kiefer* * MD at time of publication
        Repeated contraction of muscles surrounding the bursaàmicro trauma to bursa
Inflammatory response
Idiopathic
(common: kids 4-7 years old)
Bursa located between medial head of gastrocnemius and semimembranosus tendon
Accumulation of inflammatory fluid into the bursa forming a cyst
Meniscal Tear
Tear creates a channel between joint capsule and bursa posterior to the knee joint
Rheumatoid Arthritis
Auto-immune mediated inflammation of knee joint
Osteoarthritis
Destruction of articular cartilageà inflammation within knee joint See Osteoarthritis: Pathogenesis slide
   Damage to joint capsule and accumulation of inflammatory fluid
 Proteolytic enzyme dysregulation Local muscle action influences fluid flow and pressure changes
    Extra- articular Popliteal Cyst
Fluid filled sac behind knee
Usually asymptomatic
(resolves spontaneously)
Synovial joint herniates into popliteal fossa (most commonly medially)àaccumulation of fluid (synovial + inflammatory) into synovial herniation, forming a cyst
    Knee flexion
Intra-articular Popliteal Cyst
Known as “Baker’s cyst”
Knee extension
↑ Muscle tension around cyst
Channel between knee joint and cyst gradually closes with ↑ tensionàat full extension, no fluid flow between the joint and cyst (trapping the fluid in the cyst)
     ↓ Muscle tension around cyst
Channel between knee joint and cyst opens
Knee joint pressure becomes negative
↑ Fluid flow from cyst into knee joint
↓ Fluid in popliteal cyst
Knee joint pressure becomes positiveà ↑ Fluid flow from knee joint into cyst
                (Posterior tibial nerve located lateral to the popliteal cyst and enervates posterior knee, calf, and bottom of foot)
Posterior tibial nerve becomes entrapped by mass (cyst)
Enlarging mass in posterior knee
Cyst reaches maximum volume
Pressure within cyst exceeds tensile strength of surrounding sac
Fluid drains distally into calf
Popliteal vein located lateral to cyst
↑ Venous pressure distal to cyst
Popliteal vein occluded by mass
Venous pooling distal to site of occlusion
↓ Space in posterior calf compartmentàcompression of calf muscles and posterior tibial nerve
Ankle dorsiflexion ↑ pressure in compartment
Positive Homan’s sign
(discomfort with passive ankle dorsiflexion)
              Posterior plantar numbness
Pain receptors (nociceptors) activated
Posterior knee pain/pressure
Cyst Rupture Abrupt and intense pain
Stimulates inflammatory response
Fluid pushed from veins into interstitial space
Swelling and erythema (redness) in distal calf and foot
Pseudo-Thrombophlebitis
       worse with extension or physical activity
(shares symptoms with deep vein thrombosis but no associated clot)
 Legend:
 Pathophysiology
Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published Feb 10, 2018, updated Nov 19, 2022 on www.thecalgaryguide.com

hypovolemic-shock

Hypovolemic Shock: Pathogenesis, Complications, and Clinical Findings
Authors: Dean Percy Miranda Schmidt Reviewers: Yan Yu Tristan Jones Frank Spence* Ben Campbell Ayaaz Sachedina* * MD at time of publication
Progressive ↓ in level of consciousness
Pulseless Electrical Activity
Acute Kidney Injury
↑ Reabsorption of salt and water in the kidney
Oliguria
(↓ urine output)
    Inflammation (pancreatitis, cirrhosis, post-operative, etc.)
Inflammatory mediators ­ vessel permeability and fluid leaks out
Trauma
Ruptured vessels leak fluid into potential spaces
Hemorrhagic losses
(GI bleed, postpartum hemorrhage, etc.)
↓ Intravascular volume
↓ Venous return to the heart
↓ Cardiac output (blood pumped from the heart)
Hypovolemic Shock
↓ Oxygen delivery to tissues due to low blood volume
Insufficient organ perfusion
Non-Hemorrhagic losses
(dehydration, GI losses, skin losses / burns, renal losses, etc.)
         ‘Third Spacing’ of fluid
(fluid located outside the intravascular or intracellular space; large collections can occur in the pelvis, thorax, GI tract, long bones of children, intra-abdominally, retroperitoneally)
P = Q x R; less ‘flow’ in the vessels (Q), with vessels not constricting enough to maintain resistance (R)à pressure (P) will drop
↓ Blood Pressure
Caution: young, healthy individuals can maintain blood pressure during circulatory collapse with ­ cardiac output and ­ vasoconstriction; do not use blood pressure as an indicator of shock severity in children
Carotid sinus baroreceptors sense low blood pressure ↓ Carotid sinus inhibition of sympathetic nervous system Release of sympathetic catecholamines (epinephrine and
↓ Pressure in venous circulation
Brain
Heart
Kidneys
↓ Blood in the right internal jugular vein
↓ Oxygen delivery to the brain
↓ Myocardial contractility (from lactic acidosis)
↓ Blood flow to kidneys
↓ Jugular Venous Pressure
                                        Catecholamines bind to beta-1 receptors in the sinoatrial node of the heart
Beta-1 receptor activation causes ↑ heart rate
Tachycardia
norepinephrine)
Catecholamines bind to and stimulate alpha-1 receptors in peripheral vessels
Vasoconstriction of peripheral vessels
↓ Blood flow to peripheral tissue
Catecholamines bind to and stimulate beta receptors in sweat glands
Diaphoresis
(sweating)
In all body tissues
Inadequate oxygen delivery
↓ ATP production
↑ Anaerobic metabolism
↓ Body temperature
Impaired neurological functioning
Renal ischemia
Activation of the renin-angiotensin aldosterone system
↓ Glomerular filtration rate
↓ Clearance of lactic acid by the kidney
↑ Lactic acid production
↓ Rate of activity of clotting enzymes
Lactic Acidosis
Unknown mechanism
Coagulopathy Hypothermia
             Trauma Triad of Death
           ↑ Capillary Cold, mottled refill time extremities
   Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published January 24, 2013, updated December 4, 2022 on www.thecalgaryguide.com

Ischemic Stroke: Pathogenesis

Ischemic Stroke: Pathogenesis Small artery occlusion
Acute injury (<20mm diameter) of basal or brainstem penetrating arteries
Large artery atherosclerosis
Cholesterol plaque ↓ diameter of intra- or extracranial vessel
Cardiac embolism
Blood clot in heart breaks free, travels to brain
Other
E.g. volume loss, severe infection
Unknown
E.g. 2 or more mechanisms
Modest ↓ in O2 at penumbra (see figure)
Authors: Mizuki Lopez Andrea Kuczynski Illustrator: Mizuki Lopez Reviewers: Sina Marzoughi Usama Malik Hannah Mathew Ran (Marissa) Zhang Andrew M Demchuk* Gary M. Klein* * MD at time of publication
       Significant ↓ in O2 at ischemic core (see figure)
↑ Anaerobic metabolism ↓ ATP
Production
Dysfunction of Na+/K+ ATPase pump (for 1 ATP molecule, 3 Na+ moved out of cell, 2 K+ moved into cell)
H2O influx following Na+ Cerebral edema
Compression of vessels and surrounding tissue damages blood-brain barrier
↑ Permeability of damaged blood-brain barrier
Infiltration by peripheral immune cells
Immune cells release inflammatory cytokines
↓ Cerebral Blood Flow
     Penumbra Ischemic core
          Metabolic demands are greater than supply of ATP
Cell death
Microglia (resident neural immune cells) activate to clean dead cell debris
Microglia release inflammatory cytokines (TNFα, IFγ, IL-1β)
Cytokines lead to astrocyte activation (support cells for neurons)
Astrocytes release more inflammatory cytokines
Inflammation of brain tissue
↑ Na+, Ca2+ influx, K+ outflux
↓ Glutamate (excitatory neurotransmitter) reuptake by astrocytes (support cells for neurons)
↑ Glutamate in extracellular fluid
Spreading depolarization from core (unclear mechanism)
Activate biochemical pathways including glutamate receptor activation
↑ Glutamate activity
Activate glutamate receptors that conduct Ca2+
↑ Ca2+ influx into neuron
Activation of catabolic proteases, lipases, nucleases in neuron
Dysfunction of neuronal protein synthesis and activity
Neuronal cell death
↑ Volume of dead (infarcted) brain tissue
                 Neurons depolarize and release glutamate
Reversal of Na+ Dependent Glutamate Reuptake Transporters on astrocytes (normally 3 Na++ 1 H+ + 1 glutamate into cell, for 2 K+ out)
            ↑ Glutamate in extracellular fluid
      Stroke symptoms (e.g. weakness, slurred speech, visual field losses, autonomic dysfunction)
(see Ischemic Stroke: Impairment by Localization stroke slide)
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published November 14, 2017; updated November 6, 2022 on www.thecalgaryguide.com

Cirrhosis

Cirrhosis:
pathogenesis and complications
Author:
Chunpeng Nie
Yan Yu
Reviewers:
Paul Ratti, Amy Maghera, Vina Fan, Ben Campbell,
Sam Lee*, Mayur Brahmania* *MD at time of publication
Chronic viral hepatitis (B, C, or D) Hepatitis D occurs with hepatitis B
Alcohol related liver disease
Non-alcoholic steatohepatitis
Autoimmune: autoimmune hepatitis, primary biliary & sclerosing cholangitis
Genetic: hemochromatosis, Wilson’s disease, α1- antitrypsin deficiency
Toxic: drugs may rarely cause chronic liver disease or cirrhosis
         Severe or chronic hepatocyte injury overrides regenerative capacity → Fibrosis
Cirrhosis
Mild fibrosis may reverse with treatment of underlying cause
Nodular/shrunken liver on ultrasound
↑ Liver stiffness on transient elastography Diffuse fibrosis with nodular regeneration on biopsy
            Fibrotic liver provides ↑ resistance to blood flow
Portal hypertension
Irreversible formation of fibrosis, within which hepatic cell regeneration is restricted to form nodules of poorly-functioning cells
Inflammation → epigenetic changes, oncogene mutations
Hepatocellular carcinoma
↑ Serum α-fetoprotein (sensitivity 50%, specificity 99%)
Fibrosis disrupts normal function of hepatic lobules
Hepatic insufficiency
↓ Liver synthetic function
↓ Thrombopoietin synthesis
Thrombocytopenia
↓ Conjugation of bilirubin,
↓ secretion of bilirubin into bile ducts, and
↓ drainage of bilirubin into hepatic duct
Accumulation of serum bilirubin >30 μmol/L
Jaundice
Scleral icterus, jaundiced frenulum
     ↑ Pressure in portal venous circulation
          Portosystemic shunts
Increased flow to esophageal, rectal, and splenic veins
Vascular stretch → endothelial vasodilator (e.g. NO) release
Vasodilators enter systemic circulation
Pulmonary vasodilation
Blood cells in pulmonary vasculature have ↓ time for gas exchange
Hepatopulmonary syndrome (rare)
Dyspnea or hypoxemia, worsened when upright
↓ Synthesis of clotting factors (V, VII, IX, X, XI, XII, fibrinogen, prothrombin) and anticoagulant proteins (antithrombin, proteins C/S)
Unpredictable imbalance of hemostatic and anticoagulant factors
Elevated INR
± coagulopathy
Impaired metabolism of waste products and toxins
Toxins (mainly ammonia) accumulate and cross the blood- brain barrier
Hepatic encephalopathy
Day-night reversal, asterixis, delirium
      Varices
(dilated veins in the esophagus, stomach, or rectum)
Variceal bleed
GI bleed and hypovolemia
Backflow of blood into spleen
Splenomegaly
Enlarged spleen sequesters (traps) blood cells
Cytopenias
(eg. thrombocytopenia or anemia), petechiae, easy bruising
↓ Blood flow to kidneys, which sense ↓ effective blood volume
Kidneys retain water and Na+
↑ Hydrostatic pressure in splanchnic vessels
↓ Albumin synthesis
↓ Capillary oncotic pressure
                 Net fluid flux out of vasculature into interstitial space
          Ascites
(fluid in peritoneal cavity)
Peripheral edema
  Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published August 7, 2022, updated November 6, 2022 on www.thecalgaryguide.com

Hidradenitis Suppurativa

   Genetic mutations causing impaired function of gamma-secretase (NCSTN, PSEN1 or PSENEN genes)
Defective notch signaling pathway (a regulator of many cell processes)
Hormones (excess androgen activity)
Smoking (nicotine exposure)
Skin to skin friction
Systemic inflammation
Hidradenitis Suppurativa:
Pathogenesis and Clinical Findings
     Other unknown genetic factors
Follicular occlusion
HS nodule
Epidermal layer
Dermal- Epidermal Junction
Dermal layer
Inflammatory cytokine- mediated activation of nociceptors
Inflammatory cytokines
Sebaceous gland
Apocrine sweat gland
Hair follicle
Pilosebaceous- apocrine unit
                             Changes in gene expression of hair follicle and apocrine gland anti-microbial peptides
Accumulation of corneocytes (dead keratinocytes) and sebum àfollicular occlusion and rupture
↑ Interleukin-36 (IL-36, a cytokine) Altered keratinocyte differentiation
Hair follicle hyperkeratinization
Abnormal structure and function of the pilosebaceous- apocrine unit
Infiltration of normal skin flora microbes, macrophages, dendritic cells, and Th17 cells
Increased density of nicotinic acetylcholine receptors in pilosebaceous-apocrine unit (see figure)
                   Mechanism not fully understood
Hyperhidrosis
Pain
     Innate immune cells produce inflammatory cytokines: tumour necrosis factor alpha (TNF-α) and various interleukins (IL-1, IL-6, IL-12, IL-17, IL-22 and IL-23)
IL-17 promotes neutrophil migration into the skin
Formation of neutrophil extracellular traps (NETs)
B-cell activation and IgG autoantibody formation against skin tissue antigens
Scarring
Ongoing inflammation impairs wound healing
           Granulocyte infiltration and activation, release of granulocyte colony-stimulating factor
Accumulation of keratin, purulent and/or serosanguinous fluid in dermis
Inflammatory papules, nodules, and abscesses
Pro-inflammatory cytokine-mediated response, leading to epithelial hyperplasia with increased fibrosis and collagen remodelling in the dermis
Formation of epithelial tissue tunnels in the dermis with two cavities on either end that open to the skin surface and fill with fluid or keratin
     Hidradenitis Suppurativa
Chronic Inflammatory skin disease that occurs in areas with a high density of apocrine sweat glands, including the axilla, underneath the breasts, groin, and buttocks
Sinus tracts (dermal connections between lesions)
Double-ended comedones
Authors: Leah Johnston Reviewers: Mehul Gupta Lauren Lee Stephen Williams Ben Campbell Laurie Parsons* * MD at time of publication
        Wound drainage and odour
Psychological distress
 Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
Published January 30, 2023 on www.thecalgaryguide.com

Inhalational Injury

Inhalation Injury: Pathogenesis and complications
Authors: Marshall Thibedeau, W Fraser Hill, Paloma Arteaga Juarez Reviewers: Spencer Yakaback, Tony Gu, Dami Omotajo, Ben Campbell, Yan Yu*, Michael Liss*, Duncan Nickerson*, Donald McPhalen* * MD at time of publication
 Smoke Inhalation
Suspect if patient found unconscious, history of fire in a confined space and presence of facial burns
       Convective heat transfer
Exposure to chemical irritants
Tracheobronchial injury
Injury stimulates vasomotor and sensory nerves of the trachea and bronchi
Neuropeptides from neurons are released into local circulation and activate nitric oxide
Inhalation of toxins
Lack of O2 in an enclosed space
Asphyxiation
(O2 deprivation in lungs)
Acute hypoxemia
Loss of consciousness
Death
             Upper airway injury (above vocal cords)
Cell lysis & necrosis
Local release of inflammatory substances
↑ Vascular permeability à edema of tissues in upper airway
Damage to lower respiratory tract cilia
↓ Mucus clearance from alveoli
Parenchymal injury
(delayed reaction dependent on severity of burn)
Alveolar epithelial and endothelial barrier irritation/damage
Inflammatory response
Carbon monoxide poisoning
Cyanide poisoning
Cyanide binds to mitochondrial cytochrome oxidase a3
     CO binds more strongly to hemoglobin than O2
↑ Carboxyhemoglobin in blood, ↓ free hemoglobin available to bind O2 in the lungs
↑ Affinity for O2 on remaining binding sites in hemoglobin (i.e. hemoglobin binds O2 more strongly and is slow to release it)
↓ O2 delivered to tissues
Inhibition of cytochrome c oxidase
          Mucous obstructs airways
Air retained
distal to the obstruction is resorbed from nonventilated alveoli
Regions without gas collapse i.e atelectasis
↑ Risk of infection
↓ Mitochondrial respiratory chain function
Impaired oxidative phosphorylation & cellular energy production
Cellular dysfunction in high metabolic tissues
             Nitric oxide acts as a vasodilator in alveolar arterioles
Loss of hypoxic vasoconstriction (constriction of arterioles in alveoli due to ↓ O2)
Reactive inflammation & bronchoconstriction
       Stridor
Complete airway obstruction
↑ Vascular permeability leading to fluid leakage into interstitium and alveoli
           Blood flow to poorly ventilated alveoli is maintained
Ventilation/perfusion (V/Q) mismatch (regions of lung not effectively ventilated despite being well perfused by blood)
Acute Respiratory Distress Syndrome See ARDS Pathogenesis slide
Hypoxemia
Lower airway edema
Wheezing Coughing
Impaired brain function
Muscle weakness
Impaired heart function
         Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 First published November 10, 2019; Updated on February 12, 2023 on www.thecalgaryguide.com

Central Retinal Vein Occlusion

Central Retinal Vein Occlusion: Pathogenesis and clinical findings
Authors: Graeme Prosperi-Porta Mina Mina Reviewers: Stephanie Cote Usama Malik Mao Ding Johnathan Wong* *MD at time of publication
       Hypercoagulable state
(pathologic state where there is an exaggerated tendency for the blood to clot)
↓ Anticoagulation (e.g., Protein C or S def) and/or ↑ coagulation (e.g., malignancy, Polycythemia Vera)
Non-ischemic (perfused) CRVO
Glaucoma
(a disease characterized by ↑ intraocular pressure)
Optic disc drusen
(calcified nodules located within the optic disk – the most anterior part of the optic nerve)
Diabetes
Dyslipidemia
Hypertension
Vasculitis
(inflammation of blood vessels; e.g., sarcoid, Systemic Lupus Erythematosus)
Endothelial damage
       Pupillary responses do not vary between both eyes when a light is shone in one eye at a time
Relative afferent pupillary defect (RAPD) mild or absent
Lost vascular wall integrity
Normal retinal electrical response to a light stimuli
Normal
electro- retinography (ERG) with b- wave >60%
Few scattered Intra-retinal hemorrhages
↓ In retinal electrical response to a light stimuli
Pupils respond differently to a light stimulus shone in one eye at a time
Severe ↓ in visual acuity (Snellen acuity of <20/400)
Visual field deficits
↓ b-wave amplitude (<60%) on ERG
RAPD present
↑ Pressure compromises retinal vein outflow
Central retinal atherosclerosis (build-up of fat & cholesterol plaque in arteries) & hardening
Atherosclerotic changes in the central retinal artery compress the central retinal vein (since they are both held together in region of the optic disc)
Venous stasis (stagnation of blood flow) ↑ Likelihood of thrombus formation
Central Retinal Vein Occlusion (CRVO)
      A common retinal vascular disease characterized by blockage of the main vein that drains blood from the retina
CRVO classified based on the degree of perfusion (as seen through retinal angiography)
Ischemic (nonperfused) CRVO
              ↑ Intraluminal venous pressure
Dilated tortuous retinal veins
Normal visual fields
Vascular wall integrity lost
Four-quadrant hemorrhages described as “blood and thunder” on fundus exam
Obstruction due to thrombus
↓ Retinal capillary perfusion causes ischemia
Hypoxia in the retinal tissues
↑ Release of vascular endothelial growth factor to revascularize diseased tissue & overcome hypoxic conditions
Angiogenesis (formation of new blood vessels)
Intraretinal infarcts
“Cotton wool spots” on fundus exam
                 Venous capillary fluid/protein leakage
Mild retina, macula and disc edema
Moderate ↓in visual acuity often (Snellen acuity >20/400)
New vessel proliferation can occur in the anterior chamber of the eye
This change can block the outflow path of the aqueous humor in the eye
Neovascular glaucoma
Neovascular vessels are structurally different in comparison to regular retinal vasculature
          Neovascular vessels are more fragile
↑ Likelihood of rupture Vitreous hemorrhage
Neovascular vessels lack tight junctions
    ↑ Fluid leakage
Retina, macula and disc edema
    Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 First Published June 28, 2017, updated March 4, 2023 on www.thecalgaryguide.com

Douleur somatique aigue

Douleur somatique aigue:
Auteurs: Lisa Murphy, Yan Yu* Rédacteurs: Mackenzie Gault, Melinda Davis* Traducteur/Traductrice: Ovidiu Croitoru Sylvain Coderre* * MD au moment de la publication
Nocicepteurs: Neurones qui détectent les stimulis nocifs ou douloureux et transmettent ces informations à la moelle épinière. Il existe deux grands types:
- A∂ fibres: Myélinisés, sensation initiale nette et rapide.
- C fibres: amyélinisés, sensation retardée, terne, brûlante.
Vers l’hypothalamus
Les neurones de 2e ordre
synapsent dans l’hypothalamus
Les neurones hypothalamiques
coordonnent la réponse viscérale du corps à la douleur
 Pathophysiologie
Lésions tissulaires aigues, trois types de causes:
Mécanique (ex: piqure) Thermique (ex: poêle chaude activé) Chimique (ex: inflammation)
Nocicepteurs activités au site de blessure (Neurones sensoriels de 1er ordre)
Fibres nociceptives (A∂ et C) transportent l’information sensorielle nocive vers la corne dorsale ipsilatérale de la moelle épinière
Les neurotransmetteurs excitateurs sont libérés et stimulent les neurones sensoriels de 2ème ordre.
Les neurones sensoriels de 2ème ordre décussent immédiatement dans la moelle épinière et remontent via les voies antérolatérales (spinothalamiques) du côté opposé, se terminant à divers endroits
          Vers le thalamus
Les neurones de 2e ordre synapsent dans le thalamus
Dans le thalamus, les neurones sensoriels de 2ème ordre synapsent avec les neurones sensoriels de 3e ordre, qui transmettent le signal au cortex cérébral
Vers le tronc cérébral
Les neurones de 2e ordre
synapsent dans la formation réticulaire du tronc cérébral
Vers le mésencéphale
Les neurones de 2e ordre
synapsent dans la zone grise périaqueducale (PGA) du mésencéphale
         Localisation et sensation douleur
Réponse émotionnelle et comportementale
Stimule les voies descendantes pour moduler le signal de douleur
Diminution ou augmentation de la perception de la douleur
↑ Rythme cardiaque
Nausée
    Légende:
 Pathophysiologie
Mécanisme
Signes/Symptôme/Labo
 Complications
 Publié 25 avril 2019 sur www.thecalgaryguide.com

Pyogenic Brain Abscess on MRI

Pyogenic Brain Abscess on MRI: Findings and Pathogenesis Common pathogens such as staphylococcus and streptococcus bacteria exist in the
outside environment or on the skin
Hematogenous Spread Direct Spread
Pathogen travels to brain by bloodstream Pathogen travels to brain by ears or sinus
Pathogen enters the brain parenchyma by crossing the blood brain barrier (BBB) transcellularly, paracellularly or by a Trojan Horse mechanism
Authors: Omer Mansoor, Aly Valji, Nameerah Wajahat Reviewers: Mao Ding, Reshma Sirajee, James Scott* *MD at time of publication
R
              Transcellular
Pathogen invades BBB cell directly to enter
Paracellular
Pathogen goes between BBB cells by disrupting tight junctions
Trojan Horse
Pathogen bypasses BBB by hiding inside a macrophage cell
Ring Enhancing Lesion Sensitive, but not specific for a Brain Abscess
     Pathogen causes parenchymal inflammation and progresses through four stages of infection
Axial T1 + Gadolinium MRI Head. Pyogenic Brain Abscess showing infection at Stage 3 or 4. T1 highlights fat, whereas T2 highlights fat and water*.
R
     Stage 1: Early Cerebritis Focal infection (2-3 days) with no pus formation
Stage 2: Late Cerebritis Progressive (1 week) infection of abscess
Stage 3: Early Encapsulation
In 1-2 weeks, pus is formed from the pathogen
Stage 4: Late Encapsulation After 2 weeks, abscess shrinks in size as it necroses
      Pathogen causes acute inflammatory changes of swelling (edema) and vascular congestion
No fibrous capsule is formed yet and infection is not well localized on a T1 MRI
Increased edema could be seen on T2 MRI
Poor Demarcation and Vasogenic Edema
Abscess is not well-defined with minimal enhancement on T1 but can have increased signal on T2
Fibrous capsule is formed by surrounding healthy brain tissue that walls off abscess
Capsule is made of granulation tissue and fat, which lights up non-specifically on a T1 MRI
Use Diffusion Weighted Imaging (DWI) to distinguish abscess from other brain lesions
DWI measures water diffusion in different directions, specifically diseased areas where water movement is restricted
Restricted Diffusion Edema and inflammation allow water to move freely (hyperintense signal)
         *Imaging Source: Feraco et al., 2020: https://jptcp.com/index.php/jptcp/article/download/688/685?inline=1
DWI Sequence Axial MRI Head. Same Pyogenic Brain Abscess as above. DWI is the mainstay imaging sequence for diagnosing a pyogenic brain abscess*.
 Legend:
 Pathophysiology
Mechanism
Radiographic Findings
 Complications
 Published Mar 25, 2023 on www.thecalgaryguide.com

Carpal Tunnel Syndrome

Carpal Tunnel Syndrome: Pathogenesis and clinical findings
Diabetes Mellitus (See Pathogenesis Slide)
↑ Blood sugar: deposition of advanced glycation end (AGE) products (proteins or lipids glycated when exposed to sugar)
AGE attaches to and prevents tendons from moving properly
Authors: Amanda Eslinger Yvette Ysabel Yao Reviewers: Matthew Harding Owen Stechishin Mao Ding, Cory Toth* * MD at time of publication
Wrist trauma (distal radius fractures, carpal/metacarpal fractures, tendon ruptures)
      Repetitive Strain Injury (repetitive hand & wrist movements)
Irritation, swelling & thickening of tendons in carpal tunnel
Calcium deposits
Calcifica tion
Deposition Amyloidosis
Amyloid
(protein aggregates) deposition
Gout
(See Gout Slide)
Uric acid crystal deposition
Pregnancy
↑ Concentration of hormones & uterine pressure on inferior vena cava
Backup of blood into systemic circulation
Autoimmune
(i.e. Rheumatoid arthritis, scleroderma, lupus, Sjogren’s syndrome)
↑ Inflammatory cytokines causing inflammation
Hypo- thyroidism
Myxedema (swelling of skin and underlying tissues) in carpal tunnel
                      Idiopathic or Congenital
Edema
Narrowed carpal tunnel leads to ↑ internal pressure
Carpal Tunnel Syndrome
Vascular: median artery thrombosis in carpal tunnel
    Median nerve compression inside the carpal tunnel Mechanical disruption of median nerve
Compression exacerbated with flexed wrists (i.e. sleep, driving, holding phone/cup)
Disruption of daily activities and sleep
Ischemia (↓ blood supply) to median nerve
Hypoxia (↓ oxygenated blood flow)
Metabolic conduction block (impaired axonal transport due to ischemia)
Nerve conduction study
(show sensory nerve impulses slowing across the wrist, followed by mild / moderate / severe loss of sensory nerve amplitude
       Damage to the myelin sheath
↓ Saltatory conduction (action potential propagation along myelinated axons)
Neuropraxia (nerve compression blocks conduction)
Interruption in axonal continuity
Axonotmesis (endoneural tube stays intact but myelin & distal axon degenerates)
Recovery possible
Full disruption of myelin, axon & nerve sheath
Neurotmesis (axons no longer have an endoneural tube to guide regrowth)
Recovery impossible
               ↓ Ability to contract and use abductor pollicis brevis muscle
↓ Signals through median nerve
Interference with signals to the brain causes unusual sensations
Hypoalgesia (↓ pain Dysesthesia (tingling, burning, or sensitivity at 1st 3 1⁄2 digits) painful sensation at 1st 3 1⁄2 digits)
           Thenar muscle wasting
Reduced hand dexterity
Weak thumb abduction
 Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
Published December 2nd, 2013, updated March 22, 2023 on www.thecalgaryguide.com

Acute Pulmonary Embolism on CTPA

Acute Pulmonary Embolism: Computed Tomography Pulmonary Angiogram (CTPA/CTPE)
 Virchow’s Triad:
Hypercoagulability, venous stasis, vascular endothelial injury
Image Source: European Society of Radiology
Image: Polo mint sign on axial CTPA.
Image Source: Journal of The Indian Academy of Echocardiography
Image: Railway sign on axial CTPA. Image Source: Moore et al. 2018
Image: Pleural effusion and pulmonary infarction on axial CTPA.
Authors: Aly Valji, Nameerah Wajahat, Omer Mansoor Reviewers: Reshma Sirajee, Sravya Kakumanu, Victória Silva, Mao Ding Vincent Dinculescu* *MD at time of publication
 Deep Vein Thrombosis (DVT): Majority of pulmonary embolism (PE) arise from DVT: Clot travels via inferior vena cava → right atrium → right ventricle → pulmonary arteries/arterioles
See “Virchow’s Triad and Deep Vein Thrombosis” for full pathogenesis
    Polo Mint Sign
Clot visualized in short axis, Filling defect entirely surrounded by IV contrast creating a circle (polo mint)
Railway Sign
Clot visualized in long axis, Filling defect surrounded by IV contrast on two sides creating the appearance of a railway track
Type I or II Ventilation/Perfusion respiratory failure (V/Q) mismatch
Reverse Halo Sign
Pulmonary infarction leads to wedge shaped opacity with a rim of consolidation (black arrows) surrounding “ground glass” (red arrows)
Pulmonary Embolism: Clot in pulmonary arteries
See “Signs and Symptoms of Pulmonary Embolism” for full presentation
Saddle Embolus: Large clot over the pulmonary trunk bifurcation Lobar/Segmental/Subsegmental Embolus: Clot within the pulmonary arteries of the lungs
Blockage of pulmonary arteries = ↓ Blood flow, ↑ Right heart pressure
              ↓ Gas exchange b/w lungs and blood
Ischemia of lung tissue → infarction → inflammation of dead tissue
↑ Right heart filling and expansion
Left heart filling impaired
↓ Cardiac output due to ↓ Left heart filling
“Massive PE” = sustained systemic hypotension or bradycardia (SBP < 90 mmhg, HR < 40 bpm)
Saddle Embolus
Filling defect due to blockage of bifurcation. IV contrast appears white, and embolus appears grey
               Pleural Effusion
Exudative Pleural Effusion
Tissue inflammation → ↑ blood vessel permeability = Leakage of fluid into pleural space
Transudative Pleural Effusion
↑ Hydrostatic pressure from right heart congestion = Pushes fluid into pleural space
  Image Source: Samra et al. 2017
Image: Saddle embolus on axial CTPA.
 Legend:
 Pathophysiology
Mechanism
Radiographic Findings
 Complications
Published March 28, 2023 on www.thecalgaryguide.com

Coronary Artery Bypass Graft CABG Indications

Coronary Artery Bypass Graft (CABG): Indications
Author: Breanne Gordulic Reviewers: Miranda Schmidt Ben Campbell Sunawer Aujla Angela Kealey* * MD at time of publication
  Symptomatic multivessel (≥ 3 vessels) coronary artery disease (MVCAD) or complex MVCAD
Acute coronary syndrome (ACS)
Left main coronary artery disease
Multivessel (≥ 3 vessels) CAD and diabetes
Cardiac surgery required for other pathology
Multivessel CAD, LV dysfunction and congestive heart failure (CHF)
Complex CAD includes stenosed vein grafts, bifurcation lesions, calcified lesions, total occlusions, ostial lesions
STEMI initial treatment is PCI/thrombolysis
CABG outcomes compared to percutaneous coronary intervention (PCI) in MVCAD include ↑ survival in diabetes, ↑ survival with LV dysfunction, ↓ repeat revascularization, ↓ myocardial infarction, ↓ stroke
↑ Risk of failure in complex CAD with PCI
     Rapid reperfusion to myocardium most important in STEMI to decrease myocardial damage
CABG can be considered for residual stenoses 6-8 weeks later
   NSTEMI or unstable angina (UA) with MVCAD involving at least three vessels including the proximal left anterior descending (LAD)
     Left main coronary artery divides into left anterior descending (LAD) and left circumflex (LCx) which supplies 2/3 of myocardium
↑ Survival Myocardial infarction from left main artery occlusion Death
        Left main stenosis
Ventricular dysrhythmias
Ongoing ischemia
LV dysfunction Hemodynamic instability
↑ risk of PCI
CABG has mortality benefit
↓ Number of operations
       ↑ Risk of cardiovascular disease in diabetes
Revascularization indicated along with other cardiac surgery
Multivessel CAD with >90% stenosis and CHF
LV ejection fraction <35%
↑ Risk of atherosclerosis from hyperglycemia and dyslipidemia
CABG bypasses several atherosclerotic plaques in coronary arteries
↑ Durability
↑ Complete perfusion
     Valve stenosis or regurgitation Septal defect
Aortic root or arch pathology
Combination procedure
          Evidence of ischemia at rest
Evidence of
impaired LV function at rest
↓ All-cause mortality in CABG vs medical management
Chronic obstructive pulmonary disease
Abbreviations:
• ACS- acute coronary syndrome. Acute reduction in
blood flow to heart muscle resulting in cell death. • CAD- coronary artery disease. Narrowing or blockage of the coronary arteries by plaque
• NSTEMI- myocardial infarction (heart attack) with no ST elevation on electrocardiogram
• PCI- percutaneous coronary intervention. A balloon tipped catheter is used to open blocked coronary arteries; a stent may be placed.
• STEMI- myocardial infarction with ST segment elevation on electrocardiogram
       Consider revascularization (restore blood flow to blocked or narrowed blood vessels) of coronary arteries to increase perfusion to myocardium (heart muscle)
Coronary artery bypass graft recommended
Assess surgical risk and comorbidities with evaluation by heart team that includes both a cardiac surgeon and interventional cardiologist (SYNTAX Trial)
Individual management plan for patients with comorbidities that increase mortality
Frailty
Chronic Renal Failure
↑ Inflammation and deregulated angiogenesis affects all organ systems
↓ Physiologic reserve and ↓ ability to recover from acute stress
↑ Pneumonia
↑ Respiratory and Renal Failure
↑ Stroke
↑ In hospital mortality
↓ Survival two years after surgery
                      Use Society of Thoracic Surgeons Score, EuroSCORE, or SYNTAX II Score to predict patient outcome with anatomy, disease severity, and preoperative characteristics
Coronary Artery Bypass Graft
Surgery to take healthy blood vessels from the body and connect them proximally and distally to blocked coronary arteries
Cardiopulmonary bypass, fluid overload, ↑ renal vasoconstriction and ↓ renal oxygenation from rewarming
Kidney injury
↑ End stage kidney disease
    Blood flow restored to
ischemic myocardium
↓ Angina
↑ Quality of life ↑ LV function ↑ Survival
      Legend:
 Pathophysiology
Mechanism
 Sign/Symptom/Lab Finding
 Complications
Published April 12, 2023 on www.thecalgaryguide.com

Telogen Effluvium

Authors: Ayaa Alkhaleefa Telogen effluvium (TE): Causes, pathophysiology, and clinical findings Reviewers: Elise Hansen, Sunawer Aujla, Dr. Jori Hardin* *MD at time of publication
Telogen effluvium: non-scarring alopecia characterized by diffuse shedding of telogen-phase hair due to a reactive process
        Hypothyroidism
↓ Thyroid hormones (T3,T4)
↓ Binding of thyroid hormone to receptors in the skin and hair
Cell division ceases in keratinocytes
The catagen phase of the hair cycle is triggered (involuting phase where hair enters apoptosis)
Delayed re-entry of telogen (resting)
hair into the anagen (growing) phase
Post-partum hair loss (telogen gravidarum)
↑ Circulating placental estrogen
Prolonged anagen phase
↑ Hair growth during pregnancy
Baby is delivered
↓ Estrogen and other trophic
hormones postpartum
The increased amount of anagen
hairs from pregnancy all enter catagen phase simultaneously
Nutritional deficiencies i.e. iron deficiency
Critical illness
Fever triggers various pro- inflammatory cytokines (tumor necrosis factor, interleukin 1b, interleukin 6, and interferon types 1 and 2)
Premature entry into catagen phase (the body induces cell-cycle arrest in all non-essential structures)
Hair follicle keratinocytes undergo apoptosis in response to inflammation
         Ribonucleotide reductase (an enzyme involved in DNA synthesis) cannot utilize iron as a co-factor
↓Iron stores
↓ Expression of iron-
dependent genes (CDC2, NDRG1, ALAD, and RRM2)
↓ Expression of matrix genes of a healthy hair follicle (Decorin and DCT)
       ↓ Production of matrix keratinocytes (cells that form the hair shaft of growing hair)
     Arrest of matrix proliferation
Hair shedding commonly occurs in the bitemporal areas 2-3 months after triggering event
          Hair shaft Hair follicle
Telogen phase
Skin
Epidermis
Dermal- Anagen phase
  Epidermal Junction Dermis
      Catagen phase
         Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published May 10, 2023 on www.thecalgaryguide.com

OA Clinical findings

Osteoarthritis: Pathogenesis and clinical findings
Ehlers-Danlos Syndromes (connective tissue disorders, e.g., Familial Hypermobility Syndromes)
Damage of normal cartilage under abnormal loading
Authors: Sean Spence Modhawi Alqanaie Reviewers: Yan Yu Jennifer Au Mao Ding Gary Morris* * MD at time of publication
   Single large traumatic event or repeated microtrauma
Damage to normal articular cartilage under normal loading (force put on a joint)
Genetic anomalies in cartilage production and inborn errors of cartilage metabolism
Damage of abnormal cartilage under normal joint loading
Destruction/attrition of articular cartilage
Osteoarthritis
       A degenerative joint disease that can affect both load bearing joints (knee, hip) as well as in smaller joints (proximal inter-phalangeal, carpometacarpal joints in hand)
       Repeated physical joint trauma
Aberrant bone deposition secondary to wear on subchondral bone
Formation of osteophytes (bony projections) and subchondral sclerosis (bone thickening)
Lack of cartilage
Direct contact between bony processes with movement of the joint
Inflammation alters the chemical milieu of joint tissue
        Synovial fluid forced into bone
Damage to subchondral (under cartilage) blood vessels
Subchondral fractures
cytokines regulate hyaluronic acid synthase
Synovium makes lower molecular weight hyaluronic acids (a potent proinflammatory molecule)
↓ synovial fluid viscosity
↑ risk of infection
Increased secretion of proteolytic enzymes
↑ joint fluid production
Joint effusion
          Disruption of normal joint architecture
Palpable bony hypertrophy
(ex. Bouchard’s nodes (bony bumps on the middle joints of the finger))
Impaction of osteophyte with normal joint structures during movement
Physical disability
Crepitus
(grating sound in a joint)
Joint movement with reduced lubrication stimulates joint nociceptors
    Stimulation of joint nociceptors (sensory receptors that detect damaging stimuli) in subchondral bone
Pain with use of joint
Pain with motion
↓ Range of motion
           Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published November 1, 2012, updated May 18, 2023 on www.thecalgaryguide.com

Acute Laryngitis

Acute Laryngitis: Pathogenesis and clinical findings Infectious
Author: Charmaine Szalay-Anderson Reviewers: Shayan Hemmati, Sunawer Aujla, Derrick Randall*,
             Viral (most common)
Malaise Fever
Fungal
Atopy (asthma, allergy)
Non-infectious
Gastroesophageal Reflux
Trauma or damage to larynx
Smoking
Yan Yu*
* MD at time of publication
Environmental Pollution/Inhalants
Bacterial (S. pneumoniae,
H. influenzae, M. catarrhalis)
Systemic immune response
Spread of infection to larynx through upper respiratory tract
Infection of the vocal folds and surrounding tissue
Mechanical
(vocal misuse/ trauma)
     (Area in the neck that contains the structures for voice production, anatomically anterior to the esophagus, inferior to the pharynx and superior to the trachea)
  Irritation of the vocal folds and surrounding tissue
       Inflammatory cascade triggered
Acute Laryngitis
Symptoms for <3 weeks
Acute injury to vocal folds
Vocal fold
lesions (i.e., vocal polyps)
    Laryngeal inflammation
Neutrophils and macrophages release inflammatory cytokines
     Local laryngeal inflammationà↑ vascular permeability ↑ Secretion of mucous leading to airway congestion Cough reflex initiated to clear airway congestion Cough
Edema of vocal folds and surrounding tissue
      Dysphagia (difficulty swallowing)
Dysphonia (difficulty speaking)
Odynophagia (painful swallowing)
Swelling impairs vocal cord vibration
Frank aphonia (loss of voice)
      Progressive worsening of edema
  Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published May 24, 2023 on www.thecalgaryguide.com

diverticulosis-vs-diverticulitis-distinguishing-features

Diverticulosis vs. Diverticulitis: Distinguishing features
Authors: Sahil Prabhnoor Sidhu, Vadim Iablokov, Vina Fan Reviewers: Brandon Hisey, Laura Byford-Richardson, Raafi Ali Dr. Sylvain Coderre* * MD at time of publication
Local inflammation
Diverticulitis
Inflammation of diverticuli
  Conditions causing inherent
weakness in bowel wall, i.e. aging, Ehlers-Danlos syndrome, Marfan syndrome
Risk factors (i.e. low fiber diet, obesity, inactivity, smoking) contributing to reduced gut motility
↑Intraluminal pressure in the colon
   Herniation of colonic mucosa and submucosa through circular muscle at points of weakness to form outpouchings
Diverticulosis
Presence of outpouchings in the colon (diverticuli)
Note: In Western populations, most diverticulosis is left-sided, whereas in Asian populations, these outpouchings are more often right-sided.
Mucosal abrasion or micro-perforation by ↑ intraluminal pressure or dense food particles
Bacterial overgrowth,
dysbiosis and passage into the lamina propria
         Feces collects in diverticuli
Gut bacteria metabolize undigested material and produce gas
Stretching of colon Bloating wall irritates and
visceral afferent flatulence nerves
Episodic abdominal discomfort and cramping
Blood vessels in
the mucosa and submucosa (vasa recta) are stretched over the diverticuli, and may rupture from ↑ pressure
Diverticular bleed
Painless hematochezia (passage of fresh blood from the rectum)
Inflammatory cytokines activate clotting factors
Clotting of blood in vessels supplying diverticula
Inflammatory cytokine release (IL-6, TNF-α)
Cytokines enter systemic circulation
Edema in the bowel wall
Irritation of adjacent parietal peritoneum and somatic nerves
Left lower quadrant (LLQ) pain, guarding
Small abrasions are walled off by pericolic fat and mesentery
                    ↑WBC
Fever
Inflammation may spread to nearby organs, leading to ulceration and abnormal connections between organs
Pericolic abscess
        No hematochezia
Local ischemia and focal necrosis resulting in loss of integrity of the bowel wall
Perforation
Generalized peritonitis
         Colonic obstruction (rare)
Fistulae (rare): i.e. colovesical, coloenteric
Stricture/fibros is formation with healing
  Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published October 12, 2016; Updated July 30, 2023 on www.thecalgaryguide.com

Knee Osteoarthritis

Authors: Jared Topham Knee Osteoarthritis: Pathogenesis and clinical findings Reviewers: Liam Thompson, Raafi Ali Yan Yu*, Kelley DeSouza* * MD at time of publication
  Primary Causes
Secondary Causes
          Aging
↓ Synovial fluid in joint
Gender
Females > males
Genetics
Family history of osteoarthritis
Race
Black > Caucasian
Joint malposition (e.g. valgus or varus)
Articular trauma
Inflammatory disease or infection (e.g. Rheumatoid or septic arthritis)
Obesity and ↑leptin ↑ Chondrocytes,
inflammatory mediators, and metalloproteinases
Extracellular matrix degradation
↑ Knee joint loading forces
Metabolic syndromes (e.g. diabetes mellitus)
↑ Oxidative stress and insulin resistance
Low-grade systemic inflammation
              ↓ Elasticity and ↑ degradation of cartilage
↑ Friction in knee joint with movement
↓ Cartilage along femoral groove and posterior surface of patella
Pain, catching, and crepitus (crackling/clicking sound) in the patellofemoral joint
Inability/difficulty with kneeling or climbing stairs
Abnormal distribution of forces accumulate and stress articular surface
↑ Damage/laxity to soft tissue structures stabilizing knee joint
Knee Osteoarthritis
(Multifactorial entity characterized by cartilage breakdown, deterioration of connective tissue, and bone deformities)
↓ Cartilage between distal femur and proximal tibia ↓ Joint spaceàto articular dysfunction
Radiographic changes
See Osteoarthritis (OA): X-Ray Features slide
Repeated attempts to repair cartilage and joint disruption
Subchondral bone thickening (sclerosis) under joint cartilage and bone spur (osteophyte) formation around joint line
Rotational/antero-posterior instability and ↑ external adduction moments during walking
Alterations in proteoglycans, fiber arrangement, and collagen composition in soft tissue structures within/around knee joint
↑ Shear forces and medial compartment narrowing erode and pinch soft tissue structures within the knee joint
Cruciate ligament degeneration
Weakened passive stabilizers of the knee joint
Knee giving way and instability (falls)
                        Meniscal tears, if large àprevents knee extension/flexion
Locking of the knee
Joint line tenderness:
Patient points to area of tenderness/pain reproducible upon palpation
Anatomical axis of hip, knee, and ankle joints ↑ loading medially
Medial > lateral joint line tenderness
↑ Joint friction activating nociceptors in the surrounding anatomical tissues
Injury and inflammation ↑ nociceptive responses in soft tissue structures and subchondral bone within knee joint
Nociceptive feedback to brain inhibits activity of motor cortex neurons controlling muscles around the knee
↓ Motor output and muscle activation over time
↓ Muscle strength/endurance, lower limb muscle use, functional ability (walking, stairs, etc.)
Joint inflammationà accumulation of fluid within joint
Stiffness, swelling, redness, and pain
Limited joint space reduces range of motion for femur to roll/slide on tibia
↓ Knee flexion and extension
          Flexion contracture and antalgic gait
Reduced weight acceptance of the joint and surrounding muscles/tendons
↓ Mobility and physical dysfunction
Muscular atrophy
Reduced function of active stabilizers of the knee joint (quadriceps, adductors, hamstrings)
                Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published July 30, 2023 on www.thecalgaryguide.com

Rotator Cuff Disease

Rotator Cuff Disease: Pathogenesis and clinical findings
Authors: Jared Topham Reviewers: Raafi Ali, Yan Yu*, Kelley DeSouza* * MD at time of publication
       Aging
Collagen fiber disorientation and myxoid degeneration
Tendons, ligaments, and connective tissue are replaced by gelatinous and/or mucoid substance
Obesity
↑ Loading on shoulder structures
↓ Static stability (from glenoid labrum and ligamentous components) of glenohumeral joint
Tensile forces
Repeated eccentric tension from overhead activities
Trauma, sports, and occupation
↑ Torque, compression, and translational stresses
Metabolic syndromes
Reactive oxygen species
interact with ↑ glucose forming advanced glycation end-products (AGEs) which accumulate in soft tissues
Smoking
Impingement syndromes
Vessel damage, ischemia, tenocyte apoptosis
                     Macro-trauma causing an acute, complete tear in the rotator cuff muscle(s)
↓Dynamic stability (from rotator cuff and periscapular muscles) and range of motion of the shoulder at the glenohumeral joint
↑ Bone on bone contact of proximal humeral head and boney structures of the scapula
Subacromial bursa degeneration
↓ Protection of underlying supraspinatus muscle from attrition between humeral head and acromion
Rotator Cuff Syndrome
(Inflammation, impingement, or tearing of one or more of the four muscles/tendons of the rotator cuff: supraspinatus, subscapularis, infraspinatus, teres minor)
Repetitive loading and micro-tearing of tendon/muscle fibers
↑ Oxidative stressors and inflammatory cascades
↓ Vascularity of rotator cuff structures
Radiographic changes: See Rotator Cuff Disease: X-ray and ultrasound features slide, in addition to: calcific tendonitis, calcification of in the coracohumeral ligament, and hooked acromion (calcification from tendon pulling)
     In some cases, soft tissues enclose/surround shoulder joint capsule thicken (fibrose) and tighten
        Degenerative joint disease and rotator cuff arthropathy
Proximal humeral head migration and ↓ subacromial space
Inflammation and insufficient healing of rotator cuff structures, which may lead to:
Supraspinatus (shoulder abduction) degeneration
Pain, shoulder stiffness, ↓ active AND passive range of motion
Adhesive Capsulitis (frozen shoulder)
Infraspinatus and teres minor (external rotation) degeneration
  Rotator cuff tendons become inflamed and irritated as they rub against acromion
Subacromial impingement
Subscapularis (internal rotation) degeneration
+Lift-off test: Inability to hold dorsum of the hand off lumbar spine while internally rotating shoulder
↓ Shoulder strength and muscular atrophy
                 Pain with passive shoulder flexion beyond 90°
Winging of the scapula during arm adduction
+Empty-can test: Weakness and/or arm depression with resisted abduction with arm internally rotated in 90°
+Drop-arm test: Inability to maintain shoulder in abducted position at 90° and/or adduct the arm in a controlled manner (resulting in ”dropping”)
Weakness to resisted external rotation with elbow in 90° flexion, inability to keep arm externally rotated (infraspinatus)
+Hornblower’s sign: decreased external rotation strength in arm abduction (suggests additional teres minor tear)
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published July 30, 2023 on www.thecalgaryguide.com

Low Ankle Sprain

Low Ankle Sprain: Pathomechanics and Clinical Findings
Authors: Parker Lieb, Joseph Kendal Reviewers: Liam Thompson, Tara Shannon, Sunawer Aujla, Stephanie de Waal, Amanda Eslinger, Dave Nicholl, Maninder Longowal Gerhard Kiefer* * MD at time of publication
Talus subluxation with anterior force on heel
+ Anterior drawer test
Recurrent sprains
Synovial inflammation & hypertrophy
Remodeling of collagen and bone
Degenerative changes
(e.g., osteophytes, subchondral sclerosis)
   Ankle eversion beyond normal range (less common)
Excessive stress to medial ankle deltoid ligament
Ankle plantar flexion and inversion beyond normal range (most common)
Excessive stress to lateral ankle ligament(s)
      Associated fracture of malleolus or subtalar joint
Bone pain, inability to weight bear
Recruitment of inflammatory cells to damaged area
↑ Local pro- inflammatory cytokines
↑ Capillary permeability & vasodilation to damaged area
Collagen fibers in ligament(s) rupture
Low Ankle Sprain
(medial or lateral)
Local blood vessels tear
Blood leaks into surrounding tissue
Grade I
Mild injury
Grade II
Moderate injury
Grade III
Severe injury
Minimal ligament disruption
Incomplete ligament tear
Complete ligament tear
No joint laxity
Joint laxity
Gross joint laxity
Mechanical and functional instability
Decreased ankle joint space
Ankle Impingement
(compression of soft and/or bony structures in joint)
Chronic ankle instability
                        Damage to mechanoreceptors in muscles surrounding ankle joint
       Bruising
Swelling
Focal tenderness over torn ligament(s)
Pain on injury and with weight bearing
 Impaired proprioception & neuromuscular control
          Nociceptors activated by trauma and inflammatory factors
Falls Antalgic gait
  Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Nov 9, 2014; updated Aug 15, 2023 on www.thecalgaryguide.com

Rotator Cuff Disease Xray and Ultrasound Features

Rotator Cuff Disease: X-ray and ultrasound features
Rotator cuff tears can affect each of the muscles making up
the rotator cuff individually, or in combination
Authors: Jared Topham Reviewers: Raafi Ali, Kelley DeSouza* * MD at time of publication
    Supraspinatus tear (most common)
Chronic (>3 months) tear with degenerative-type changes
Rotator cuff insufficiency, loss of supporting structures holding humeral head inferiorly
Displacement of the humeral head anterosuperiorly and instability of joint
Microtrauma affecting superior aspect of glenohumeral joint
“Acetabularization” or coracoacromial arch: concave acromial erosion and increased sclerosis (hardening)
Subscapularis tear (second most common)
Teres minor tear
Infraspinatus tear
   Rotator Cuff Syndrome
(Inflammation, impingement, or tearing of one or more of the four muscles/tendons of the rotator cuff: supraspinatus, subscapularis, infraspinatus, teres minor)
Acute (partial or full thickness) tear of rotator cuff tendons
          Humeral subluxation (partial displacement of humeral head relative to glenoid)
High riding humerus: decreased acromial humeral distance
Decreased acromial humeral interval/space
(impinging tendons of rotator cuff)
Full: Defect extends from the subacromial bursa (fluid filled sack beneath the acromion and above the rotator cuff tendons) to the articular surface of the glenohumeral joint
Tendon/muscle fibers completely separated from bone and/or muscle fiber connections severed
Partial: Focal defect affecting a portion of the tendon which may involve the bursa or glenohumeral articular surface
Non-visualization of the tendon
          Acetabularization of glenoid
Fluid replaces empty space of tendon tear
Overlying fat around the sub acromial bursa falls into tendon gap
Sagging peribursal fat sign on ultrasound
            “Femoralization” of the humerus: bone erosion (destruction) and rounding of greater tuberosity
Osteoarthritis of glenohumeral joint: See Osteoarthritis (OA): X-ray features slide
Hyperechoic (brightened) line between articular cartilage of humeral head and muscle tendon on ultrasound
Cartilage interface sign on ultrasound
Hypoechoic (darkened) tendon outline discontinuity on ultrasound imaging
  Femoralization (rounding) of greater tuberosity
Subluxation of the humeral head relative to the glenoid
  Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published September 6, 2023 on www.thecalgaryguide.com

Acute Respiratory Distress Syndrome

Acute Respiratory Distress Syndrome: Pathogenesis and clinical findings Acute respiratory distress syndrome (ARDS) is a clinical syndrome involving acute lung injury. It results in severe hypoxemia and bilateral
Authors: David Olmstead Mao Ding Reviewers: Midas (Kening) Kang Usama Malik Kevin Solverson* * MD at time of publication
↓ PaO2 (Partial pressure of oxygen in arterial blood ↓SpO2 (Peripheral oxygen saturation)
Tachypnea (↑ RR) Tachycardia (↑ HR)
Dyspnea
Bilateral Opacity on chest radiograph
↓ PaO2, ↓SpO2
↑ PaCO 2
↑ PaO2, ↓PaCO2 Eupnea (normal
breathing)
↓ O2 Requirements Depression, Anxiety, PTSD Neuromuscular Weakness
Chronic Respiratory Dysfunction
airspace disease in the absence of elevated left-heart pressures.
Direct Lung Injury
Causes include pneumonia and pulmonary sepsis (community- acquired, hospital-acquired, aspiration, viral), drowning, and chemical pneumonitis from aspiration or direct inhalational injury
Indirect Lung Injury
Causes include sepsis with a non-pulmonary source, trauma, severe burns, transfusion- related acute lung injury (TRALI) and pancreatitis
        Lung Tissue Inflammation
Exudative: Neutrophils migrate into the alveoli in response to inflammatory stimulus
Note: While the three phases of ARDS take place in sequence, all areas of the lung may not be in the same phase at the same time. For this reason, the processes can be thought of as overlapping.
Proliferative: Body attempts to heal damage. If it is not successful, the tissue transitions to the fibrotic phase
Neutrophil-containing pulmonary exudate interferes with surfactant function
Neutrophil infiltration and proinflammatory cytokines lead to tissue edema, dysfunction and subsequent destruction of pulmonary epithelium
Residual debris in alveoli are cleared by phagocytic cells
Restoration of alveolar epithelial cells.
Alveoli collapse in absence of working surfactant
Damaged epithelium impairs gas exchange
Pulmonary capillaries do not adequately absorb fluid
The body’s attempts to heal lung tissue result in deposition of hyaline membranes in the alveoli
Ventilation- Perfusion Mismatch
Pulmonary Edema
Impaired Gas Diffusion
                              Functional epithelium is able to absorb fluid back into circulation
↑ useful surface area for gas exchange
Clearing of CXR
       Impaired Function After Prolonged Illness
Pulmonary Hypertension
      Fibrotic: Inadequate healing results in long-term pulmonary damage (rare)
Fibroblast activity leads to deposition of collagen in alveoli and alveolar capillaries
Fatigue Pulmonary Fibrosis
Nail Clubbing (nails appear wider & swollen) Cough/Dyspnea
     Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Feb 6, 2018, updated Oct 10, 2023 on www.thecalgaryguide.com
  
Acute Respiratory Distress Syndrome: Note: Acute respiratory distress syndrome is a clinical
Authors: David Olmstead Reviewers: Midas (Kening) Kang Usama Malik Kevin Solverson* * MD at time of publication
 Pathogenesis and clinical findings
Direct Lung Injury
Causes include pneumonia and pulmonary sepsis (community-acquired, hospital-acquired, aspiration, viral), drowning, and chemical pneumonitis from aspiration or direct inhalational injury
Indirect Lung Injury
syndrome involving acute lung injury. It results in severe hypoxemia and bilateral airspace disease in the absence of elevated left-heart pressures.
  Causes include sepsis with a non-pulmonary source, trauma, severe burns, transfusion-related acute lung injury (TRALI) and pancreatitis
        Lung Tissue Inflammation
Exudative: Neutrophils migrate into the alveoli in response to inflammatory stimulus
Note: While the three phases of ARDS take place in sequence, all areas of the lung may not be in the same phase at the same time. For this reason, the processes can be thought of as overlapping.
Proliferative: Body attempts to heal damage. If it is not successful, the tissue transitions to the fibrotic phase
Neutrophil-containing pulmonary exudate interferes with surfactant function
Neutrophil infiltration and proinflammatory cytokines lead to tissue edema, dysfunction and subsequent destruction of pulmonary epithelium
Abbreviations:
PaO2: Partial pressure of oxygen in arterial blood
SpO2: Peripheral oxygen saturation.
CXR: Chest radiograph.
Residual debris in alveoli are cleared by phagocytic cells
Restoration of alveolar epithelial cells.
Alveoli collapse in absence of working surfactant
Damaged epithelium impairs gas exchange
Pulmonary capillaries do not adequately absorb fluid
The body’s attempts to heal lung tissue result in
deposition of hyaline membranes in the alveoli
Ventilation- Perfusion Mismatch
Pulmonary Edema
Impaired Gas Diffusion
↓ PaO2, ↓SpO2 Tachypnea
Tachycardia
Dyspnea
Bilateral Opacity on CXR
↓ PaO , ↓SpO 2 2
↑ PaCO2
↑ PaO2, ↓PaCO2 Eupnea
↓ O2 Requirements
Clearing of CXR
Depression, Anxiety, PTSD
Neuromuscular Weakness
Chronic Respiratory Dysfunction
                                 ↑ useful surface area for gas exchange
Functional epithelium is able to absorb fluid back into circulation
            Impaired Function After Prolonged Illness
      Fibrotic: Inadequate healing results in long-term pulmonary damage (rare)
Fibroblast activity leads to deposition of collagen in alveoli and alveolar capillaries
Pulmonary Fibrosis
Pulmonary Hypertension
Cough/Dyspnea Nail Clubbing Fatigue
        Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
Published February 06, 2018 on www.thecalgaryguide.com

Approach To Dementia

Approach to Dementia/Major Neurocognitive Disorder (NCD)
Authors: Iqra Rahamatullah Mahrukh Kaimkhani
Reviewers: Yvette Ysabel Yao Mao Ding Gary Michael Klein* *MD at time of publication
1) Changes noticed?
Modest ↓cognitive performance from previous, DOES NOT interfere with daily independence
MILD COGNITIVE IMPAIRMENT
More pronounced ↓cognitive performance from previous, DOES interfere with daily independence
MILD TO MODERATE DEMENTIA
↓Cognitive performance, difficulty with ≥1 basic activities of daily living (ADL) or ≥2 instrumental ADLs
MODERATE TO SEVERE DEMENTIA
DEMENTIA
Fluctuating course, acute onset, inattention WITH either disorganized thinking or altered level of consciousness
DELIRIUM
     2) Is it dementia?
Normal, age-related: ↓focus, ↓cognitive speed, ↓reaction time, ↓memory
NORMAL COGNITIVE DECLINE
      3) What is the cause of the dementia? (main causes discussed here)
Loss of cognitive functioning, including memory, language, problem solving, and other thinking abilities, that interferes with independence in everyday activities
      Beta-secretase cleaves beta amyloid protein
Atherosclerosis or thrombosis
Misfolded alpha- synuclein
Toxic beta amyloid plaque and tau tangle (sticky) formation
Ischemia to areas of brain (strokes)
Build ups and deposition within neurons (Lewy bodies)
Disrupted signaling, inflammation, hippocampal and cerebral impairment
Necrosis of brain tissue in areas impacted by strokes
Neuronal impairment and atrophy (especially in substantia nigra)
Neuronal atrophyàfrontal + temporal lobe atrophy
Progressive atrophy of basal ganglia and dorsal striatum + lateral ventricles expanding
Death of dopaminergic neurons in substantia nigra
Alzheimer’s Dementia
Vascular Dementia
Lewy Body Dementia
Frontotemporal Dementia
Huntington’s Disease
Parkinson’s Disease
↓Memory, ↓learning, ↓language skills, disorientation, inattention
Total debilitation, fatal infections
Findings vary depending on area
Step-wise worsening impairment
Parkinsonism, hallucinations, REM- sleep behavior disorder
Total debilitation, dependence
Personality and behavioral changes
Mental status changes
Chorea, ↓cognition, mood changes
Aspiration, dementia, suicide
Resting tremor, rigidity, anosmia
Depression, dementia, falls
              Abnormal protein inclusions and tangles (usually tau) form in neurons
Autosomal dominant disease (with anticipation) with ↑CAG repeats in Huntingtin gene
Genetic mutations, environmental exposures, or idiopathic cause
          Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published September 17, 2023 on www.thecalgaryguide.com
   
Approach to Dementia/Major Neurocognitive Disorder (NCD)
Authors: Iqra Rahamatullah Mahrukh Kaimkhani Reviewers: Yvette Ysabel Yao
Fluctuating course, acute onset, inattention WITH either disorganized thinking or altered level of consciousness (LOC)?
DELIRIUM
    1) Changes noticed?
2) Is it dementia?
Normal, age-related: ↓focus, ↓cognitive speed, ↓reaction time, ↓memory
NORMAL COGNITIVE DECLINE
Modest ↓cognitive performance from previous, DOES NOT interfere with daily independence
MILD COGNITIVE IMPAIRMENT
More pronounced ↓cognitive performance from previous, DOES interfere with daily independence
MILD TO MODERATE DEMENTIA
↓Cognitive performance, difficulty with ≥1 basic activities of daily living (ADL) or ≥2 instrumental ADLs
MODERATE TO SEVERE DEMENTIA
        3) What is the cause of the dementia? (main causes discussed here)
Beta-secretase cleaves beta amyloid protein
Atherosclerosis or thrombosis
Misfolded alpha-synuclein
Toxic beta amyloid plaque and tau tangle (sticky) formation
Ischemia to areas of brain (strokes)
Build ups and deposition within neurons (Lewy bodies)
Disrupted signaling, inflammation, hippocampal and cerebral impairment
Necrosis of brain tissue in areas impacted by strokes
Neuronal impairment and atrophy (especially in substantia nigra)
Neuronal atrophyàfrontal + temporal lobe atrophy
Progressive atrophy of basal ganglia and dorsal striatum + lateral ventricles expanding
Death of dopaminergic neurons in substantia nigra
Alzheimer’s Dementia
Vascular Dementia
Lewy Body Dementia
Frontotemporal Dementia
Huntington’s Disease
Parkinson’s Disease
↓Memory, ↓learning, ↓language skills, disorientation, inattention
Total debilitation, fatal infections
Findings vary depending on area
Step-wise worsening impairment
Parkinsonism, hallucinations, REM- sleep behavior disorder
Total debilitation, dependence
Personality and behavioral changes
Mental status changes
Chorea, ↓cognition, mood changes
Aspiration, dementia, suicide
Resting tremor, rigidity, anosmia
Depression, dementia, falls
DEMENTIA
Loss of cognitive functioning, including memory, language, problem solving, and other thinking abilities, that interferes with independence in everyday activities
                    Abnormal protein inclusions and tangles (usually tau) form in neurons
Autosomal dominant disease (with anticipation) with ↑CAG repeats in Huntingtin gene
Genetic mutations, environmental exposures, or idiopathic cause
          Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published September 17, 2023 on www.thecalgaryguide.com

Mitral Regurgitation Pathogenesis and clinical findings

Mitral Regurgitation: Pathogenesis and clinical findings Coronary Artery Disease
Authors: Juliette Hall, Victoria Nkunu
Reviewers: Raafi Ali, Jack Fu, Usama Malik, Sina Marzoughi, Jason Waechter* * MD at time of publication
 (Ischemic Heart Disease & Myocardial Infarction)
Myocarditis
         Left ventricular dilation displaces papillary muscles
Dilation of the Tethering of mitral valve annulus chordae tendineae
↑ Volume and pressure in left atrium
↑ Volume pushed back into left ventricle
Dilated left ventricle
Apical impulse on palpation and auscultation
↓ Forward flow of blood out of heart
Blood backs up into pulmonary circulation
↑ Intravascular hydrostatic pressure in pulmonary vessels
Fluid extravasates out of vessels and into the lungs
Papillary muscle rupture
Mitral valve leaflets flail
Mitral valve prolapse
Structurally abnormal valve
Connective tissue disorders
Weak valve leaflets
Rheumatic heart disease
Dilatation of the mitral valve annulus, inflammation of leaflets
Infective endocarditis
Vegetations form on valve leaflets
             Mitral Regurgitation
Blood consistently flows backward throughout systole
Holosystolic murmur, radiates to axilla, ↑ with afterload (e.g. making a fist)
  Backflow of blood from left ventricle to left atrium due to impaired mitral valve closure
     S3 heart sound
Myocardial remodeling
↓ Muscle efficiency
↓ Left ventricle systolic function
↓ O2 saturation, tachypnea, wheeze, ↑ work of breathing, crackles, frothy sputum (if severe)
Congestive heart failure
↓ Stroke volume ejected into aorta
      ↓ Cardiac output
↓ Organ perfusion       ↓ O2 to kidney
       Activation of renin-angiotensin- aldosterone system
↑ Reabsorption of water by kidneys
↑ Intravascular hydrostatic pressure systemically
Peripheral edema
Injury to kidney parenchyma
↓ ability for kidney to clear creatinine
↑ Serum creatinine
            Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Feb 3, 2018, updated Oct 15, 2023 on www.thecalgaryguide.com

Statins Mechanisms and Side Effects

Statins: Mechanisms of action & side effects
Authors: Rupali Manek, Julia Iftimie Reviewers: Gurreet Bhandal, Raafi Ali, Joshua Dian, Laura Byford-Richardson Samuel Fineblit*, Alexander Ah-Chi Leung* * MD at time of publication
         Competitive inhibitors of HMG-CoA reductase (rate-limiting enzyme in cholesterol synthesis)
↓ Coenzyme Q10 (ubiquinone)
↑ Mitochondrial superoxide
↓ Hepatic membrane stability
↓ Conversion of HMG-CoA to mevalonic acid
↑ Hepatic VLDL uptake
↓ Hepatic apolipoprotein B-100 secretion
↓ Oxidative phosphorylation
Impaired mitochondrial function
↑ Liver enzyme leakage
↓ Mitochondrial ATP production
↑ Aminotransferases enzymes in liver
↑ Clearance of
LDL cholesterol from bloodstream
Myopathy/ myalgias (muscle aches)
Hepatotoxicity
↓ Circulating LDL cholesterol
↓ LDL, ↑ HDL, ↓ TG
           ↓ Hepatic cholesterol synthesis
↓ VLDL synthesis
↑ Cell surface LDL receptor expression
     Statins
First line therapy for treating hypercholesterolemia (↑ LDL cholesterol in blood) Common examples: rosuvastatin, atorvastatin, simvastatin, pravastatin, etc.
↑ Apolipoprotein AI production & ↑ hepatic HDL neogenesis
↓ TG
↑ HDL
↑ Vasodilation
↓ C-reactive protein
↓ Impacts of coagulation cascade
↓ Atherosclerosis (plaque along walls of blood vessels)
↓ Cardiovascular disease & mortality
                   Pleotropic effects (i.e. non lipid related effects)
Abbreviations:
HMG-CoA – Hydroxymethylglutaryl-CoA LDL – Low-density lipoprotein
VLDL – Very low density lipoprotein HDL – High density lipoprotein
TG – Triglycerides
Inhibition of synthesis of isoprenoid intermediates in the mevalonate pathway
↑ Nitric oxide activity ↓ Inflammation
↓ Tissue factor expression
↓ Macrophage proliferation
↓ Tissue factor (promotes macrophage mediated thrombus formation)
↑ Blood flow & endothelial function
↓ Thrombin generation
       ↓ Metalloproteinases expression
↑ Inhibition of metalloproteinase-1
↓ Thrombogenicity (production of blood clot/thrombus)
Plaque stabilization (↓ risk of atherosclerotic plaque rupture, myocardial infarction, and stroke)
       Legend:
 Pathophysiology
Mechanism
 Sign/Symptom/Lab Finding
 Physiological Outcome
 Published July 9, 2017, updated Nov 6, 2023 on www.thecalgaryguide.com

Pharmacotherapy for Dyslipidemia Overview

Pharmacotherapy for Dyslipidemia: General overview Dyslipidemia
Authors: Rupali Manek Reviewers: Gurreet Bhandal, Raafi Ali, Yan Yu*, Samuel Fineblit* *MD at time of publication
Hypolipidemia (↓ HDL or ↓ apoB containing lipoproteins like LDL)
Bile-acid sequestrants
Bind bile acids in intestinal lumen to prevent reabsorption by enterohepatic (gut-liver) circulation
↑ Excretion of bile acids and cholesterol in stool
↓ LDL in blood
Side effects: GI disturbances, commonly interact with other drugs by interfering with absorption
See Calgary guide slide on “Bile-acid sequestrants: Mechanisms of action & side effects” for complete description of mechanism and side effects
(clinical imbalance of lipids)
     Hypertriglyceridemia (if VLDL mediated & in need of treatment for pancreatitis prevention)
Hypercholesteremia
(↑ LDL in blood)
Ezetimibe
Inhibits cholesterol absorption via NPC1L1 transporter
↑ Hepatic (liver) LDL receptor expression
↑ LDL clearance from blood
↓ LDL in blood
Avoid in pregnancy
See Calgary guide slide on “Ezetimibe: Mechanisms of action & side effects” for complete description of mechanism and side effects
Combined hyperlipidemia (↑ Triglycerides and ↑ cholesterol)
Statins (ex. rosuvastatin, atorvastatin, simvastatin, pravastatin)
Competitive inhibitors of HMG-CoA reductase (rate-limiting enzyme in cholesterol synthesis)
  Fibrates (ex. fenofibrate, gemfibrozil)
Activate PPAR! (nuclear receptor)
↑ Lipolysis (breakdown of lipids) and free fatty acid oxidation
↓ Triglycerides in blood
Side effects: GI discomfort, rash, pruritis
Contraindicated in pregnancy, renal failure, liver & gallbladder disease
See Calgary guide slide on “Fibrates: Mechanisms of action & side effects” for complete description of mechanism and side effects
PCSK9 inhibitors (ex. evolocumab and alirocumab which are monoclonal antibodies)
Inhibit PCSK9 (holds the LDL:LDL receptor complex together as it is internalized into the cell for destruction of LDL)
LDL receptor returns to surface without being destroyed
↑ LDL receptor expression
↑ LDL clearance from blood
↓ LDL in blood
See Calgary guide slide on “PCSK9 Inhibitors: Mechanisms of action & side effects” for complete description of mechanism and side effects
↓ Cholesterol synthesis in liver
↑ LDL receptor expression in liver
LDL receptor recognizes apoB100 (structural protein on LDL) and apoE (structural protein found on chylomicron, VLDL, IDL)
↑ Clearance of LDL cholesterol from bloodstream
↓ LDL cholesterol in blood ↑ HDL in blood
↓ Triglycerides in blood
↓ Atherosclerosis (plaque along walls of blood vessels)
                                  Abbreviations: HDL – High density lipoprotein; HMG-CoA – Hydroxymethylglutaryl- CoA; LDL – Low-density lipoprotein; PCSK9 – Proprotein convertase subtilisin/kexin type 9; PPAR! – Peroxisome proliferator-activated receptor alpha; NPC1L1 – Niemann-Pick C1-Like 1; VLDL – Very low-density lipoprotein
Side effects: Myalgias (muscular aches), rhabdomyolysis (muscle breakdown), transaminitis (liver inflammation), liver failure, ↑ risk of diabetes mellitus
Contraindicated in pregnancy
See Calgary guide slide on “Statins: Mechanisms of action & side effects” for complete description of mechanism and side effects
  Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published Nov 6, 2023 on www.thecalgaryguide.com

Death Cardiovascular Respiratory and Neurologic Mechanisms

Death: Cardiovascular, Respiratory and Neurologic Mechanisms
Mitochondria in tissues unable to utilize O2
Reduced hemoglobin in blood to carry O2
Low oxygen content in blood (CaO2)
Hypoxemia (Type I Respiratory Failure): low dissolved oxygen in blood (PaO2)
Lungs can’t oxygenate blood fast enough
Lungs can’t rid blood of CO2 fast enough
Hypercapnia / hypercarbia (Type II Respiratory Failure): elevated dissolved CO2 in blood (PaCO2)
Cerebral vasodilation
Toxins: e.g. cyanide, pesticides, arsenic Severe anemia
        Distributive problems:
Systemic inflammation (sepsis, anaphylaxis, pancreatitis), adrenal insufficiency, vasodilatory drugs
Obstructive problems: Cardiac tamponade*, tension pneumothorax* or massive pulmonary embolism*
Hypovolemic* problems (low blood volume): Hemorrhage, dehydration, widespread skin disruption or burns
Cardiac valve dysfunction
Myocardial infarction* or cardiomyopathy
Cardiac arrhythmia or heart block
Disturbed electrical activity in cardiomyocytes
Peripheral metabolic disturbances
Hypokalemia*, Hyperkalemia* Acidosis* (including renal failure) Hypothermia*
Toxins* (e.g. cocaine, beta blockers, tricyclics) Severe thyroid derangement
Inappropriate systemic vasodilation
Adjacent forces impair heart filling
Low cardiac preload
Low stroke volume (SV; depends on valves, contractility, preload)
Decreased systemic vascular resistance (SVR)
Low blood pressure (BP = CO x SVR)
Decreased cardiac output (CO = SV x HR)
Disseminated intravascular coagulationàwidespread thrombi that occlude blood flow (also causes hemorrhage, see relevant box at left)
Methemoglobinemia: some hemoglobin gets stuck in a state that can’t carry O2
Hemoglobin has reduced capacity to carry or release O2
Drugs: e.g. dapsone, nitrates
Carbon monoxide poisoning
            Circulatory collapse / shock: inadequate perfusion of tissue with blood
Respiratory collapse: blood has insufficient useable O2 content
                                Ventricular fibrillation (VF) or pulseless ventricular tachycardia (VT)
Hypoxia*: inadequate O2 delivery or utilization in tissues
Hypoxia creates metabolic disturbances that impair cardiac cells. Alternatively, any of the preceding conditions marked with (*) can directly trigger cardiac arrest first
Pulseless Electrical Activity (PEA): organized activity on ECG with no cardiac output (can be preceded or mimicked by pseudo-PEA, in which there is still some output on ultrasound)
Low atmospheric pressure or oxygen content Severe lung disease
Asthma, COPD, interstitial lung disease, congestive heart failure, pulmonary hypertension, pulmonary embolism, lung collapse / atelectasis
Acute respiratory distress syndrome
Pneumonia, aspiration pneumonitis, inhalational injury, systemic inflammation, drowning
Severe hypoventilation
Respiratory fatigue, advanced COPD, chest wall disorders, neuromuscular disorders, upper airway obstruction, toxins (e.g. opioids, botulism)
       Can degenerate at any time
   Asystole: no cardiac electrical activity or output
Death
Respiratory arrest: cessation of breathing
Inability to protect airway
Decreased level of consciousness
          Note
This is a broad overview of the many scenarios that can result in death. For detailed explanations of the various disease mechanisms, refer to the corresponding slides.
* = reversible causes of cardiac arrest (Hs and Ts)
Author:
Ben Campbell
Reviewers:
Yan Yu*
Huma Ali*
* MD at time of publication
Bradycardia
(low heart rate, HR)
Unopposed parasympathetic stimulation of heart (can also cause vasodilation, see Distributive problems)
Disruption of spinal cord sympathetic control
Injury to cervical or upper thoracic spinal cord
Irreversible cessation of cardiac, respiratory, and brain function
      Prolonged seizure initially causes increased cardiovascular activity, until the system fatigues
Disruption of respiratory control center in medulla
Expanding skull contents squeeze brainstem (herniation)
Increased intracranial pressure
Edema from intracranial hemorrhage, trauma, brain mass
Edema, inflammation, hypoxia and/or metabolic derangements cause diffuse neuron dysfunction
Central nervous system infection
Dementia, particularly with delirium
Massive ischemic stroke
    Seizure
activity prevents or alters breathing
Metabolic disturbances that affect the central nervous system Hypoglycemia Hypocalcemia, hypercalcemia Hyponatremia, hypernatremia Uremia
Acute liver failure (hyperNH4) Many drugs / toxins Withdrawal (e.g. EtOH)
          Status epilepticus
Brainstem lesion (e.g. stroke, neoplasm, inflammatory)
 Nervous System Insult
 Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published November 11, 2023 on www.thecalgaryguide.com
 Respiratory System Insult
Cardiovascular System Insult Cardiogenic problems

Non-Alcoholic Fatty Liver Disease

Non-Alcoholic Fatty Liver Disease: Pathogenesis and clinical findings Diagnosis of Metabolic Syndrome when ≥ 3 out of the 5 preceding risk factors are present
Authors: Stephanie Happ Reviewers: Obesity Hypertension Diabetes Hypertriglyceridemia Hypercholesterolemia Iffat Naeem Sunawer Aujla Edwin Cheng* * MD at time of publication
        Insulin resistance develops in adipose tissue and hepatocytes
   ↓ Ability of insulin to suppress lipolysis of adipose tissue
↑ Delivery of free fatty acids from adipocytes to the liver
↑ De-novo lipogenesis in the liver
        Hepatic Steatosis: accumulation of fat in the liver (in the absence of alcohol consumption, termed Non-Alcoholic Fatty Liver (NAFL))
Steatohepatitis: chronic inflammatory and apoptotic climate in the hepatocytes (in the absence of alcohol consumption, termed Non-Alcoholic Steatohepatitis (NASH))
Fibrosis of the Liver: excessive scarring of liver tissue resulting from chronic inflammation, although liver architecture is largely intact
Fat droplets form and grow in the hepatocytes
Hepatic mitochondria increase their workload in attempt to break down the excess free fatty acids through beta-oxidation
↑ in cellular workload creates more reactive oxygen speciesà Inflammation and apoptosis of hepatocytes
    On-going inflammation damages hepatic stellate cells (the primary extracellular matrix–producing cells of the liver) causing the release of fibrinogenic cytokines
Cirrhosis of the liver: normal lobular structure distorts and is replaced by regenerating nodules and bridging septa, disrupting normal liver blood flow
Deposition of fibrotic
material and collagen within the perisinusoidal spaces of the liver
Decompensated Cirrhosis Hepatocellular carcinoma
       Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published November 25, 2023 on www.thecalgaryguide.com

RICE mechanism of action

Rest, Ice, Compression, Elevation (RICE): Mechanism of action
Healing of an injury requires pain and inflammatory control to encourage activity. The goal of Rest, Ice, Compression, Elevation (RICE) is to
decrease inflammation. Though activity itself will contribute to pain and inflammation, it is integral to the rehabilitation process.
Authors: Matthew Roberts Emma Windfeld Reviewers: Alexander Arnold Amanda Eslinger Shyla Bharadia Mao Ding Bradley Jacobs* * MD at time of publication
    Rest: (weight bearing or stressful motion discontinued)
Further damage to affected tissues from mechanical stress is prevented
Ice: (applied to injury)
Compression:
(wrap applied to injured area)
Mechanical force applied to tissue
Excess fluid is pushed back into capillaries and lymph network
Elevation:
(limb raised above heart)
          Blood flow to tissue is constricted
Mechanism not well understood
      ↓ delivery of inflammatory mediators such as polymorphonuclear neutrophils and macrophages to injured site
↓ production of inflammatory cytokines (pro-inflammatory substances) such as Tumor Necrosis Factor-α, Platelet Derived Growth Factor, Epidermal Growth Factor, and Transforming Growth Factor-β
↓ Inflammation
Gravity ↑ venous blood return to systemic circulation
     ↓ edema (accumulation of fluid in interstitium)
Early initiation of injury-specific rehabilitative exercises improves range of motion, strength, and proprioception
Stress to targeted area induces inflammation that, when tightly regulated, is integral to repair
Injured muscle, tendon, bone, or ligament is strengthened
      ↓ Pain
↑Range of motion and therefore function
   Early recovery from injury
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Dec 9, 2013, updated Oct 15, 2023 on www.thecalgaryguide.com
   
Rest, Ice, Compression, Elevation (RICE): Mechanism of action
Healing of an injury requires pain and inflammatory control to encourage activity. The goal of Rest, Ice, Compression, Elevation (RICE) is to decrease
inflammation. Though activity itself will contribute to pain and inflammation, it is integral to the rehabilitation process.
Authors: Matthew Roberts Emma Windfeld Reviewers: Alexander Arnold Amanda Eslinger Shyla Bharadia Bradley Jacobs* * MD at time of publication
     Rest: (weight bearing or stressful motion discontinued)
Further damage to affected tissues from mechanical stress is prevented
Ice: (applied to injury)
Compression: (wrap applied to injured area)
Mechanical force applied to tissue
Excess fluid is pushed back into capillaries and lymph network
Elevation: (limb raised above heart)
Gravity ↑ venous blood return to systemic circulation
      Blood flow to tissue is constricted
↓ delivery of inflammatory mediators such as polymorphonuclear neutrophils and macrophages to injured site
Mechanism not well understood
              ↓ production of inflammatory cytokines (pro- inflammatory substances) such as Tumor Necorsis Factor-α, Platelet Derived Growth Factor, Epidermal Growth Factor, and Transforming Growth Factor-β
↓ Inflammation
↓ Pain
↓ edema (accumulation of fluid in interstitium)
↑Range of motion and therefore function
Early initiation of injury- specific rehabilitative exercises improves range of motion, strength, and proprioception
Stress to targeted area induces inflammation that, when tightly regulated, is integral to repair
Injured muscle, tendon, bone, or ligament is strengthened
Early recovery from injury
              Legend: Published MONTH, DAY, YEAR on www.thecalgaryguide.com
Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
Complications
  
RICE: Mechanism of action
Authors: Matthew Roberts Emma Windfeld Reviewers: Alexander Arnold Amanda Eslinger Shyla Bharadia Bradley Jacobs* * MD at time of publication
 Rest
(weight bearing or stressful motion discontinued)
Ice
(applied to injury)
Compression
(wrap applied to injured area)
Elevation
(limb raised above heart)
Constricts blood flow to tissue
Mechanism not well understood
Mechanical force applied to tissue
↓ Delivery of polymorphonuclear neutrophils and macrophages to injured site
Excess fluid pushed back into capillaries and lymph network
Gravity ↑ venous blood return to systemic circulation
Prevents further damage to affected tissues from mechanical stress
↓ Production of inflammatory cytokines
↓ Edema (accumulation of fluid in interstitium)
↓ Inflammation
↓ Pain
                 ↑Range of motion and therefore function
   Early initiation of injury-specific rehabilitative exercises to improve range of motion, strength, and proprioception
Stress to targeted area induces inflammation that, when tightly regulated, is integral to repair
Injured muscle, tendon, bone, or ligament is strengthened
Early recovery from injury
            Legend: Published MONTH, DAY, YEAR on www.thecalgaryguide.com
Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
Complications

Aspiration Pneumonia

Aspiration Pneumonia: Pathogenesis and clinical findings
        Intractable vomiting
↑ Likelihood of oropharyngeal and gastric contents exiting the esophagus, entering the trachea to the lung
If the acidic gastric contents are sterile, then aspirating this results in inflammation and lung injury without development of infection
Aspiration pneumonitis
Alveolar macrophages recruit neutrophils to local site of infection. Subsequent cytokine release compromises the vascular endothelial cell wall barrier and ↑ alveolar-capillary permeability
↑ inflammation due to fluid and cellular debris build-up in alveoli
overdose
(e.g. opioids) (e.g. stroke)
Altered level of consciousness and impaired cough/clearance
Tube Poor Alcohol and Substance Medications Neurologic diseases
Esophageal and gastric motility disorders
Impaired swallowing
Chronic obstructive pulmonary disorder
feeding oral health
Bacteria adhere to epithelial surfaces and ↑ risk of airway and lung bacterial colonization
Aspirated oropharyngeal and gastric contents can also contain bacteria
↓ Elimination and clearance of foreign bacteria from airway and lung
Macroaspiration (large volume aspiration) of oropharyngeal bacteria, during eating and drinking
                    Bacteria and fluid fill bronchi and alveolar space
Aspiration Pneumonia
Alterations to lung microbial flora
  An infectious lung process caused by inhalation of foreign bacterial and oropharyngeal and gastric contents
   Aspiration of acidic fluid and pneumonia causative pathogen (typically anaerobes or bacteria in normal oral flora) with resultant inflammation
Infiltrate develops in a gravity-dependent pattern in patches around bronchi segments.
Produces proinflammatory cytokines, (e.g. tumor necrosis factor-alpha, and interleukin-1)
Hypothalamic production of prostaglandin E2 results in thermogenesis
Fever
Authors: Luiza Radu
Reviewers: Mao Ding, *Yan Yu, *Jonathan Liu *MD at time of publication
    Aspiration to the right lung more common due to large diameter and more vertical orientation of the right main bronchus
          Crackles and ↑ lung vibrations (fremitus) on auscultation
Productive Cough
Impaired alveolar gas exchange
Chemoreceptor detection of ↓ pO2 triggers
↑ ventilation
Hypoxemia
Dyspnea
Consolidation in lower lobes (particularly superior segments) and posterior segments of upper lobes
If untreated, a
pus-filled lung cavity develops (e.g. abscess)
  Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Jan 11, 2024 on www.thecalgaryguide.com

Shoulder Impingement Syndrome

Shoulder Impingement Syndrome: Signs and Symptoms
      Calcific deposition
Abnormal shoulder morphology
Static subacromial narrowing
Abnormal scapular rotation and tilt
Scapular winging Weakness
Degenerative changes to rotator cuff tendons
Weakness
Dynamic subacromial narrowing
Repetitive overhead activity/shoulder overuse
Muscle fatigue
Repetitive compression forces on subacromial space
Glenohumeral instability or stiffness
                Authors:
Janelle Wai
Dalal Awwad Reviewers:
M. Patrick Pankow Reza Ojaghi Usama Malik Sunawer Aujla Ryan Shields*
* MD at time of publication
Internal/ Posterior Impingement
Compression of rotator cuff tendons between humeral head and posterosuperior glenoid edge during end stage of throwing
Pain with passive extension and lateral rotation
+ Posterior Internal Impingement Test
Instability:
Laxity of glenohumera l joint
Stiffness: Scapular winging with downward tilt, shoulder protraction
  Primary/ Structural Impingement
Any anatomical abnormalities
Secondary/Functional Impingement
Normal anatomy with motion abnormalities
  Impingement of underlying rotator cuff muscle-tendon unit and inflammation of subacromial bursa
Rotator Cuff Syndrome
External/ Subacromial Impingement
Compression of subacromial bursa and rotator cuff (i.e. supraspinatus tendon) on the anterolateral acromion and coracoacromial ligament
X-Ray: Normal Ultrasound: +/- Tendinopathy, muscle atrophy
Pain with
overhead movement
Pain at night
(e.g., sleep position, gravity)
Pain with lifting
(e.g., weight- training, groceries)
                Pain (between 60°-120°) with passive shoulder abduction
+ Painful arc Test
Pain with passive shoulder flexion
+ Neer’s Test
Pain with passive shoulder flexion (to 90°) + internal rotation
+ Hawkins-Kennedy test
      Legend:
 Pathophysiology
Mechanism
 Sign/Symptom/Lab Finding
 Complications
 First published May 27, 2018; updated Jan 11, 2024 on www.thecalgaryguide.com

Arachnoid Cysts MRI Findings

Arachnoid Cysts: Findings on MRI
Imaging source:
radiopaedia.org
Authors: George S. Tadros Reviewers: Matthew Hobart, Shahab Marzoughi, James Scott* * MD at time of publication
   Extra-Axial Location
Cyst is visualized outside of brain parenchyma
Clear Demarcation
Since the arachnoid cyst is bound by arachnoid membrane, it has well-defined margins
CSF collection contained within a split arachnoid membrane that occurs during embryological development (primary)
Cerebrospinal Fluid (CSF) collection within arachnoid membrane adhesions following trauma, infection, inflammation or surgery (secondary)
    Arachnoid cyst (fluid-filled sac) formation within the layers of the arachnoid membrane, outside of brain parenchyma (for full pathogenesis, see Calgary Guide slide Arachnoid Cysts: Pathogenesis and clinical findings)
Use Diffusion Weighted Imaging (DWI) and Fluid-Attenuated Inversion Recovery (FLAIR) to distinguish from other cysts
Isointense to CSF on T1 and T2
Arachnoid cyst contents should appear isointense to CSF on each MR sequence, including diffusion-weighted imaging
Axial T2 MRI Head. Clearly demarcated hyperintense (bright) arachnoid cyst is seen (red arrows)
Axial T1 MRI Head. Clearly demarcated hypointense (dark) arachnoid cyst is seen (red arrows)
        FLAIR allows for suppression of free water signal to enhance fluid with ↑ protein concentration
Arachnoid cysts contain CSF-like fluid with very little to no protein
Complete suppression on FLAIR
Cysts are mostly fluid and contains no protein, so it is suppressed and appears darker in FLAIR images.
Axial FLAIR MRI Head. Hypointense cyst is seen on FLAIR (red arrows), showing low protein content in CSF-like fluid inside arachnoid cyst
DWI measures water diffusion in different directions and whether there is restriction on the direction of flow
Fluid within the cyst can flow freely, with no restriction on the direction of movement
Non-restricted diffusion
There is no directional restriction on flow within the cyst (unrestricted diffusion), so it appears dark on DWI.
Axial DWI MRI Head. Hypointense cyst is seen on DWI (red arrows), showing unrestricted diffusion within arachnoid cyst
        Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Jan 20, 2024 on www.thecalgaryguide.com

Arachnoid Cysts Pathogenesis and clinical findings

Arachnoid Cysts: Pathogenesis and clinical findings
Authors: George S. Tadros Reviewers: Yvette Ysabel Yao Shahab Marzoughi Gary Michael Klein* * MD at time of publication
  Failure in the embryological duplication or division of the arachnoid membrane
Cerebrospinal fluid (CSF)-like fluid is trapped within the erroneous membrane
Formation of a primary, congenital arachnoid cyst (most common cause)
Head trauma, intracranial hemorrhage, or infection
Inflammation and deposition of cellular matrix Adhesion of the arachnoid membrane
CSF accumulates in the subarachnoid space (space between the arachnoid mater and pia mater)
Formation of a secondary arachnoid cyst (less common)
            Cyst remains stable in size and does not expand (most common)
Patients are asymptomatic
Arachnoid cyst is diagnosed incidentally on unrelated neuroimaging (see Calgary Guide slide Arachnoid Cysts: Findings on MRI)
Arachnoid Cyst
Cyst grows in size and expands (rare but more common in children under four years of age)
Cyst exerts pressure on other structures (mass effect)
Suprasellar region
Cyst ruptures into the subdural space (rare)
CSF-like fluid accumulates in the subdural space
Subdural hygroma (collection of non-bloody CSF)
Intracranial hypertension (↑ intracranial pressure)
Generalized symptoms
               Middle fossa
Compression and irritation of the temporal cortex
Seizures
Focal symptoms depending on cyst location
Cerebellopontine angle
          Compression of vestibulocochlear nerve (Cranial Nerve VIII)
Compression and interruption of cochlear blood supply
Cyst presses on the third ventricle and aqueduct
buildup of CSF in the ventricles
Obstructive hydrocephalus
Cyst presses on the optic chiasm, hypothalamus, and pituitary
Visual impairments and endocrinopathies
Headache (most common)
Vomiting
Nausea
         Progressive hearing loss
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published Jan 25, 2024 on www.thecalgaryguide.com

Avascular Necrosis AVN of the Femoral Head Findings on MRI

 Avascular Necrosis (AVN) of the Femoral Head: Findings on MRI
Traumatic or atraumatic disruption of blood supply to the proximal femur (for full pathogenesis, see Calgary Guide slide Avascular Necrosis: Pathogenesis and clinical findings)
Ischemia of the femoral head (usually unilateral for traumatic and bilateral for non-traumatic)
Prolonged anoxia (total oxygen deprivation) within the femoral head
Cell death (necrosis) of osteocytes and marrow cells in the femoral head, forming a focal lesion (sequestrum)
Histiocytes and giant cells (immune cells) aggregate around the sequestrum, forming a “reactive zone” around the periphery of the sequestrum
Apoptotic osteocytes in the anoxic reactive zone cannot be phagocytosed leading to dysregulated bone remodeling and osteosclerosis (hardening of bone and ↑ bone mineralization and density due to ↓ resorption and ↑ bone formation)
Femoral head becomes progressively weaker while the mechanical load on it remains the same
Progressive femoral head/subchondral bone collapse Osteoarthritis
Areas with ↑ fluid content appear darker on T1w images
Areas with ↑ fluid content appear brighter on T2w images
Areas with ↑ bone density and ↓ fat content appear darker on T1w images
Areas with ↑ bone density and ↓ fat content appear darker on T2w images
Basic MRI Physiology
Edema and inflammation increases fluid content
Sclerotic areas have ↑ bone density and thus ↓ fat content
Location of Signs
Inflamed reactive zones are darker on T1w images
Inflamed reactive zones are brighter on T2w images
Sclerotic areas are darker on both T1w and T2w images
Authors: George S. Tadros Reviewers: Matthew Hobart, Mao Ding Shahab Marzoughi David Cornell* * MD at time of publication
              T1w
  Signs on both T1-weighted and T2-weighted images are most commonly seen on the superior anterolateral aspect of the femoral head
Single dark band on T1-weighted MRI
A single band-like crescentic lesion of low signal intensity is seen on T1-weighted MRI images (white arrows). This band represents the edematous reactive zone between the necrotic and normal tissue, and typically extends to the subchondral plate
      Double Line Sign on T2-weighted fat-saturated MRI
An outer distal low signal intensity line is seen (white arrows) representing reactive bone sclerosis
Image source: radiologymasterclass
T2w
  An inner proximal high signal intensity line is also seen (red arrows) representing vascular and repair tissue at the periphery of the sequestrum
   Image source: radiologymasterclass
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
Published Jan 27, 2024 on www.thecalgaryguide.com

Bacterial Infections from Transfusion

Bacterial Infections from Transfusion: Signs and Symptoms
Caused by bacterial contamination of any blood product, most commonly platelets due to room-temperature storage
      Bacteria enters bloodstream directly through transfusion
Immune cells (ex. macrophages, dendritic cells, monocytes, neutrophils) recognize bacteria
Immune cells release pyrogens (either bacterial components or signalling molecules) upon recognition
Pyrogens bind to receptors on the hypothalamus
Hypothalamus ↑ body temperature set point
Body generates and conserves heat to reach set point
Immune cells release pro- inflammatory cytokines (messenger protein)
Cytokines activate sympathetic nervous system via hypothalamus
Adrenal glands release stress hormones (epinephrine and norepinephrine) into bloodstream
Stress hormones bind to receptors on cardiomyocytes (heart muscle cells)
↑ Heart contractility
↑ Heart rate
Cytokines circulate throughout the body
Cytokines interact with endothelial cells of the blood vessels
↑ Nitric oxide production
Relaxes smooth muscle cells of blood vessel walls
Vasodilation (↑ blood vessel diameter)
Hypotension
Systemic inflammation
Stimulate nerve endings in muscles
Stimulate nerve endings in gastrointestinal tract’s lining
Blood brain barrier’s integrity is compromised
Nitric oxide reacts with oxygen to form reactive nitrogen species
Tissue damage
Fatigue
Weakness Muscle aches
Nausea and vomiting
Immune and inflammatory cells enter the brain
                             Authors:
Arzina Jaffer
Kayleigh Yang
Reviewers:
Nimaya De Silva
Raafi Ali
Michelle J. Chen
Yan Yu*
Kareem Jamani*
* MD at time of publication
Inflammation in the brain
Activates pain processing centers in the brain
Headache
Damages neurons and disrupts cell communication
Confusion and altered mental state
           Shivering
Fever
 Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published January 30, 2024 on www.thecalgaryguide.com

Acute Wound Healing

  ↓ Blood supply and oxygenation to local skin tissue
Ischemia
Degradation of intact skin
Abrasion (damage by scrape/rub)
Puncture (small piercing caused by sharp object)
Acute injury to the skin
Crush (damage by compression)
Acute Wound Healing:
Pathogenesis and clinical findings
Author: Amanda Eslinger Mina Youakim Reviewers: Heena Singh Shahab Marzoughi Yan Yu Laurie Parsons* * MD at time of publication
8 – 365+ days post-injury
Remodeling
(↓ blood vessels & organized collagen)
Extensively cross-linked type 1 collagen replaces the disorganized
collagen laid down in the proliferative phase
↑ Protein content in collagen
Scarring (fibrotic tissue replaces previously healthy tissue)
            Disruption of structure and function of dermis, epidermis and subdermal tissues
Subendothelial and endothelial damage activates the coagulation pathway
Formation of a platelet plug
Bleeding is slowed or stopped by
hemostatic plug (hemostasis)
Clot unifies wound edges
0 – 7 days post-injury
Inflammation
(In disrupted skin layers)
4 - 14 days post-injury
Proliferation of collagen, extracellular matrix & blood vessels
TGF-β attracts fibroblasts to the site of the wound
Fibroblast & macrophage stimulate tissue growth &
angiogenesis which replaces hemostatic plug
Scabbing (protective crust overlying damaged tissue)
Re-epithelialization beneath the scab sloughs it off
Healing (newly replaced tissue replaces damaged one)
        In response to irritant, mast cells release histamine
Complement activation causing nearby endothelial cells to release prostaglandins
      Vasodilation occurs around the wound area
Localized ↑ vascular supply (reception of blood and fluid from vessels)
↑ Hydrostatic pressure forces fluid from vessels into surrounding tissue
Edema (swelling from fluid buildup)
Erythema (redness)
             Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 First Published Sept 19, 2013; updated Jan 30, 2024 on www.thecalgaryguide.com

Acute Otitis Externa Complications

Acute Otitis Externa (Swimmer’s Ear): Complications
Acute Otitis Externa (AOE)
Authors: Charmaine Szalay-Anderson Vaneeza Moosa Reviewers: Shayan Hemmati Shahab Marzoughi Ben Campbell Justin Lui* * MD at time of publication
Spread to subcutaneous tissue
 Chronic otitis externa (>6 weeks)
Chronic inflammation of the outer ear
Fibroblast activation for collagen and extracellular matrix components production for tissue repair
Excess accumulation of tissue
Ear canal fibrosis (thickening)
Ear canal stenosis (narrowing)
Damage/obstruction to ear canal structures with impaired fluid drainage & pressure buildup
Inflammation of the outer ear
Recurrent or non-resolving acute otitis externa Dissemination of infection
          Spread to connective tissue and cartilage
Perichondritis (inflammation of ear cartilage)
Spread of Pseudomonas aeruginosa
in an immunocompromised host or due to antibiotic resistance
Rapid infectious spread through soft tissue to mastoid and/or temporal bone
Malignant (necrotizing) otitis externa *can be life threatening
Inflammation of connective tissue and bony structures
Spread to
tympanic membrane
Myringitis (inflammation of tympanic membrane)
Swelling and thinning of tissue
Tympanic membrane perforation (tear)
Immune reaction with inflammation
Dead white blood cell, bacteria & tissue debris accumulation in the ear canal
Pus formation with purulent otorrhea (discharge from ear)
Localized pus accumulation
Abscess
Ear canal blockage
Periauricular/ pinna (outer ear) cellulitis
Facial cellulitis
                       Erosion of temporal bone decreasing bony sound conduction
Permanent conductive hearing loss
Direct toxicity of pathogens to surrounding nerves
Cranial nerve (CN) VII (facial) palsy (+/- CN X, XI, XII)
Out-of- proportion primary otalgia (ear pain)
Sensation of fullness in the ear
Temporary hearing loss
        Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published Dec 4, 2022; updated Feb 7, 2024 on www.thecalgaryguide.com

Infective endocarditis

Infective Endocarditis: Pathogenesis, complications, and clinical findings
Subacute Endocarditis
Pre-existing valvular stenosis or regurgitation
Non-laminar flow across valve damages valve endothelium
A sterile thrombus forms
Thrombus forms on the surface of a cardiac valve
Acute Endocarditis
Poor dental hygiene/recent dental procedure
Invasive procedure/indwelling device
Positive blood cultures
Activation of immune system
      Valve Trauma
Invading bacterium
Intravenous (IV) drug use (mostly causes right sided endocarditis)
Bacteria enter the bloodstream (bacteremia)
In subacute cases, valvular abnormality usually present beforehand
In all cases, vegetation forms on affected valve
Immune complex deposit in kidney
Immune complexes cause vasculitis in retinal vessels
Immune complexes deposit subcutaneously
↓ Blood flow to organs perfused by the obstructed arteries
     Valve endothelium is damaged
           Bacteria adhere to thrombi on the cardiac valve endothelium
Infective Endocarditis
Infection of the thrombus typically produces a vegetation on the flow surface of a valve
Immune complexes (complexes of antibody bound to antigen) form secondary to infection
Generalized immune response
Malaise Chills Fever (> 38°C)
                      Author:
Sean Spence
George S. Tadros Reviewers:
Yan Yu
Jason Baserman
Danny Guo
Steve Vaughan*
*MD at time of publication
Parts of vegetation embolize systemically, obstructing arteries
Infection destroys infected valve
Smaller emboli block smaller vessels on hands/feet
Microinfarctions
Mitral regurgitation, Aortic stenosis, Aortic insufficiency
(Valve involvement: Mitral > Aortic > Tricuspid)
Vegetation seen on ultrasound/echocardiogram Damage to Glomerulonephritis
glomeruli (Inflammation of glomeruli) Roth’s spots (retinal hemorrhages with pale centers
due to coagulated fibrin)
Osler nodes (tender, raised, red lesions found on the hands and feet)
Organ infarction (tissue death)
Splinter hemorrhages (small red streaks under nails)
Janeway lesions (non-tender, red macules/nodules on palms/soles – only a few millimeters wide)
Regurgitation (blood leaks back through the insufficient valve despite it being closed)
        Valve unable to fulfill normal functions (valve insufficiency)
 Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published Aug 20, 2013, updated Feb 24, 2024 on www.thecalgaryguide.com

Onychomycosis

Dermatophyte Onychomycosis: Pathogenesis and clinical findings
Authors: Holly Zahary Loreman Reviewers: Mina Youakim Elise Hansen Shahab Marzoughi Jodi Hardin* * MD at time of publication
Host Risk Factors
Environmental Risk Factors
         Immuno- compromised
↓ Immune response to infection
Older age
Peripheral vascular disease
Reduced blood circulation
Diabetes
Pre-existing nail dystrophy
Previous nail trauma
Integrity of nail unit is compromised
Micro- traumatic pressure on nail
Dark, warm shoe environment
Optimal conditions for fugal growth
Exposure to tinea pedis or onychomycosis
Direct spread of infection to nail unit
          High blood sugar favoring infection
      Dermatophytes invade corneocytes on stratum corneum, the uppermost non- living layer of keratinized skin
Compromise/breaking of hyponychial seal or cuticle (connection between hyponychium and nail plate)
      Proximal Subungual
White Superficial
Tinea infection (e.g. Tinea Pedis, Corporis, Capitis)
Infection spreads to distal hyponychial space
Dermatophytes colonize local tissue in nail plate and nail bed
Dermatophytes feed on keratinized tissue
General Symptoms (All Subtypes)
Spongiosis (Intercellular edema)
Acanthosis (Thickening of stratum spinosum layer of epidermis)
Hyperkeratosis (Thickening of stratum corneum In effort to rid infection)
Papillomatosis (Projections of dermal papillae)
Secondary damage to nail matrix
Loss of nail
        Keratinocytes produce an acute, low-grade inflammatory cytokine response
Onychomycosis
Dermatophytic infection of the nail bed
Inflammation promotes ↑ fluid to tissues for ↑ immune cell delivery
Widespread inflammation thickens parts of the epidermis in efforts to shed the infection
Inflammation and epidermal hyperplasia (↑ growth of cells) influence local dermal papillae (group of cells just beneath the hair follicle) to proliferate and project above the skin
 Distal Subungual
Superinfecting bacteria or other fungi proliferate beneath the compromised nail imparting a yellowish appearance
Distal Subungual Subtype
(Thick yellow nails, keratin and debris accumulate distally underneath nail plate)
Dermatophytes invade the proximal end of the nail plate
Dermatophytes penetrate through the cuticle to the newly forming nail plate moving distally
Proximal Subungual Subtype (Whitish discolouration of nail plate that begins proximally and moves distally, indicative of immunosuppression)
Fungi predominantly invade various areas of the superficial nail plate layers eventually joining together
White Superficial Subtype (Chalky white scale that spreads slowly beneath nail plate, well-defined “white islands” that coalesce as disease progresses)
The entirety of the nail plate is infected by the dermatophytes
Widespread inflammation thickens the nail plate as well as beneath the nail (subungual hyperkeratosis) in efforts to shed the infection
Total Dystrophic Subtype (End-stage nail disease, entire nail becomes thick and dystrophic)
                       Local spread of infection Dermatophytes spread causing cracks in the skin deeper into toe
Abnormal keratinization in hyponychium
Keratin accumulates between nail plate and hyponychium
     Fissure (splits in the skin)
Bacteria enters lymphatics and bloodstream
  Cellulitis Sepsis
Onycholysis (nail plate separates from nail bed)
 Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published Mar 13, 2024 on www.thecalgaryguide.com
 
Dermatophyte Onychomycosis: Pathogenesis and clinical findings
Authors: Holly Zahary Loreman Reviewers: Mina Youakim Elise Hansen Shahab Marzoughi Jodi Hardin* * MD at time of publication
Host Risk Factors
Environmental Risk Factors
         Immuno- compromised
↓ Immune response to infection
Older age
Peripheral vascular disease
Reduced blood circulation
Diabetes
Pre-existing nail dystrophy
Previous nail trauma
Integrity of nail unit is compromised
Micro- traumatic pressure on nail
Dark, warm shoe environment
Optimal conditions for fugal growth
Exposure to tinea pedis or onychomycosis
Direct spread of infection to nail unit
           High blood sugar favoring infection
      Dermatophytes invade corneocytes on stratum corneum, the uppermost non-living layer of keratinized skin
Compromise/breaking of hyponychial seal or cuticle (connection between hyponychium and nail plate)
       Proximal Subungual
White Superficial
Distal Subungual
Superinfecting bacteria or other fungi proliferate beneath the compromised nail imparting a yellowish appearance
Distal Subungual Subtype
(Thick yellow nails, keratin and debris accumulate distally underneath nail plate)
Infection spreads to distal hyponychial space
Dermatophytes colonize local tissue in nail plate and nail bed
Dermatophytes feed on keratinized tissue
Keratinocytes produce an acute, low-grade inflammatory cytokine response
Onychomycosis
Dermatophytic infection of the nail bed
Inflammation promotes ↑ fluid to tissues for ↑ immune cell delivery
Widespread inflammation thickens parts of the epidermis in efforts to shed the infection
Inflammation and epidermal hyperplasia (↑ growth of cells) influence local dermal papillae (group of cells just beneath the hair follicle) to proliferate and project above the skin
General Symptoms (All Subtypes)
Spongiosis (Intercellular edema)
Acanthosis (Thickening of stratum spinosum layer of epidermis)
Hyperkeratosis (Thickening of stratum corneum In effort to rid infection)
Papillomatosis (Projections of dermal papillae)
Secondary damage to nail matrix
Loss of nail
Tinea infection (e.g. Tinea Pedis, Corporis, Capitis)
             Dermatophytes invade the proximal end of the nail plate
Dermatophytes penetrate through the cuticle to the newly forming nail plate moving distally
Proximal Subungual Subtype (Whitish discolouration of nail plate that begins proximally and moves distally, indicative of immunosuppression)
Fungi predominantly invade various areas of the superficial nail plate layers eventually joining together
White Superficial Subtype (Chalky white scale that spreads slowly beneath nail plate, well-defined “white islands” that coalesce as disease progresses)
The entirety of the nail plate is infected by the dermatophytes
Widespread inflammation thickens the nail plate as well as beneath the nail (subungual hyperkeratosis) in efforts to shed the infection
Total Dystrophic Subtype (End-stage nail disease, entire nail becomes thick and dystrophic)
               Dermatophytes spread deeper into toe
Bacteria enters lymphatics and bloodstream
Abnormal keratinization in hyponychium
Keratin accumulates between nail plate and hyponychium
 Local spread of infection causing cracks in the skin
     Fissure (splits in the skin)
 Cellulitis Sepsis
Onycholysis (nail plate separates from nail bed)
  Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published MONTH, DAY, YEAR on www.thecalgaryguide.com
 
 Dermatophyte Onychomycosis: Pathogenesis and clinical findings
Authors: Holly Zahary Loreman Reviewers: Mina Youakim Elise Hansen Shahab Marzoughi Jodi Hardin* * MD at time of publication
Host Risk Factors
Environmental Risk Factors
         Immuno- Older compromised age
↓ Immune response to infection
Peripheral vascular disease
Reduced blood circulation
Diabetes
Pre-existing nail dystrophy
Previous nail trauma
Integrity of nail unit is compromised
Micro- traumatic pressure on nail
Dark, warm shoe environment
Optimal conditions for fugal growth
Exposure to tinea pedis or onychomycosis
Direct spread of infection to nail unit
           High blood sugar favoring infection
      Dermatophytes invade corneocytes on stratum corneum, the uppermost non-living layer of keratinized skin
Compromise/breaking of hyponychial seal or cuticle (connection between hyponychium and nail plate)
     Tinea infection (e.g. Tinea Pedis, Corporis, Capitis)
Infection spreads to distal hyponychial space
Dermatophytes colonize local tissue in nail plate and nail bed
Dermatophytes feed on keratinized tissue
Keratinocytes produce an acute, low-grade inflammatory cytokine response
Onychomycosis
Dermatophytic infection of the nail bed
General Symptoms (All Subtypes)
Spongiosis (Intercellular edema)
Acanthosis (Thickening of stratum spinosum layer of epidermis)
Papillomatosis
(Projections of dermal papillae)
Hyperkeratosis (Thickening of stratum corneum In effort to rid infection)
Secondary damage to nail matrix
Loss of nail
 Proximal Subungual
White Superficial
    Distal Subungual
Distal Subungual Subtype
(Thick yellow nails, keratin and debris accumulate distally underneath nail plate)
Proximal Subungual Subtype (Whitish discolouration of nail plate that begins proximally and moves distally, indicative of immunosuppression)
White Superficial Subtype (Chalky white scale that spreads slowly beneath nail plate, well-defined “white islands” that coalesce as disease progresses)
Total Dystrophic Subtype (End-stage nail disease, entire nail becomes thick and dystrophic)
             Local spread of infection causing cracks in the skin
Dermatophytes spread deeper into toe
Abnormal keratinization in hyponychium
Keratin accumulates between nail plate and hyponychium
Onycholysis (nail plate separates from nail bed)
      Fissure (splits in the skin)
Bacteria enters lymphatics and bloodstream
Cellulitis Sepsis
    Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Legend:
Published MONTH, DAY, YEAR on www.thecalgaryguide.com

 Dermatophyte Onychomycosis: Pathogenesis and clinical findings
Authors: Holly Zahary Loreman Reviewers: Mina Youakim Elise Hansen Shahab Marzoughi Jodi Hardin* * MD at time of publication
Host Risk Factors
Environmental Risk Factors
         Immuno- Older compromised age
↓ Immune response to infection
Peripheral vascular disease
Reduced blood circulation
Diabetes
Pre-existing nail dystrophy
Previous nail trauma
Integrity of nail unit is compromised
Micro- traumatic pressure on nail
Dark, warm shoe environment
Optimal conditions for fugal growth
Exposure to tinea pedis or onychomycosis
Direct spread of infection to nail unit
           High blood sugar favoring infection
      Dermatophytes invade corneocytes on stratum corneum, the uppermost non-living layer of keratinized skin
Compromise/breaking of hyponychial seal or cuticle (connection between hyponychium and nail plate)
     Tinea infection (e.g. Tinea Pedis, Corporis, Capitis)
Infection spreads to distal hyponychial space
Dermatophytes colonize local tissue in nail plate and nail bed
Dermatophytes feed on keratinized tissue
Keratinocytes produce an acute, low-grade inflammatory cytokine response
Onychomycosis
Dermatophytic infection of the nail bed
 Proximal Subungual
White Superficial
General Symptoms (All Subtypes)
Spongiosis (Intercellular edema)
Acanthosis (Thickening of stratum spinosum layer of epidermis)
Papillomatosis
(Projections of dermal papillae)
Hyperkeratosis (Thickening of stratum corneum In effort to rid infection)
Secondary damage to nail matrix
Loss of nail
    Distal Subungual
Distal Subungual Subtype
(Thick yellow nails, keratin and debris accumulate distally underneath nail plate)
Proximal Subungual Subtype (Whitish discolouration of nail plate that begins proximally and moves distally, indicative of immunosuppression)
White Superficial Subtype (Chalky white scale that spreads slowly beneath nail plate, well- defined “white islands” that coalesce as disease progresses)
Total Dystrophic Subtype (End-stage nail disease, entire nail becomes thick and dystrophic)
                 Local spread of infection causing cracks in the skin
Dermatophytes spread deeper into toe
Abnormal keratinization in hyponychium
Keratin accumulates between nail plate and hyponychium
Onycholysis (nail plate separates from nail bed)
      Bacteria enters lymphatics and bloodstream
 Fissure (splits in the skin)
Cellulitis
Sepsis
    Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Legend:
Published MONTH, DAY, YEAR on www.thecalgaryguide.com

 Dermatophyte Onychomycosis (Tinea Unguium): Pathogenesis, clinical findings,
Authors: Holly Zahary Loreman Reviewers: Elise Hansen Name Name* * MD at time of publication
and complications
Host Risk Factors
Environmental Risk Factors
         Immuno- compromised
↓ immune response to infection
Older age
Peripheral vascular disease
Diabetes
Pre-existing nail dystrophy
Previous Nail Trauma
Integrity of nail unit is compromised
Micro-traumatic pressure on nail
Dark, warm shoe environment
Optimal conditions for fugal growth
Exposure to tinea pedis or onychomycosis
Direct spread of infection to nail unit
          Reduced blood circulation
High blood sugar, favoring infection
    Tinea pedis infection (see ‘Tinea Capitis, Tinea Corporis, and Tinea Pedis’)
Infection spreads to distal hyponychial space Dermatophytes colonize local tissue in nail plate and nail bed Dermatophytes feed on keratinized tissue
Proximal Subungual
White Superficial
Dermatophytes invade corneocytes on stratum corneum, the uppermost non-living layer of keratinized skin
Compromise/breaking of hyponychial seal or cuticle (connection between hyponychium and nail plate)
         Keratinocytes produce an acute, low-grade inflammatory cytokine response
Onychomycosis (Tinea Unguium)
(dermatophytic infection of the nail bed)
Distal Subungual
General Symptoms (All Subtypes)
Spongiosis
Intercellular edema
Acanthosis
Thickening of stratum spinosum layer of epidermis
Papillomatosis
Projections of dermal papillae
Hyperkeratosis
Thickening of stratum corneum In effort to rid infection
Secondary damage to nail matrix
Loss of nail
         Distal Subungual Subtype
Thick yellow nails, keratin and debris accumulate distally underneath nail plate
Proximal Subungual Subtype
Whitish discolouration of nail plate that begins proximally and moves distally, indicative of immunosuppression
White Superficial Subtype
Chalky white scale that spreads slowly beneath nail plate, well-defined “white islands” that coalesce as disease progresses
Total Dystrophic Subtype End-stage nail disease, entire nail becomes thick and dystrophic
      Local spread of infection causing cracks in the skin
Dermatophytes spread deeper into toe
Abnormal keratinization in hyponychium
Keratin accumulates between nail plate and hyponychium
Onycholysis (nail plate separates from nail bed)
         Tissue Damage
Cellulitis
Sepsis
Bacteria enters lymphatics and bloodstream
     Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Legend:
Published MONTH, DAY, YEAR on www.thecalgaryguide.com

Neonatal Necrotising Enterocolitis in Premature Neonates

Neonatal Necrotising Enterocolitis (NEC) in Premature Neonates:
Pathogenesis and clinical findings
Prematurity risk factors ↓ Intestinal motility
↑ Intestinal stasis allows bacteria more time to proliferate
Bacterial overgrowth in gut
       ↓ Goblet cells in intestinal epithelium
↓ Intestinal mucus layer production leads to impaired mechanical defense against pathogenic bacteria
Immature tight junctions in intestinal epithelium
↑ Permeability of intestinal epithelial barrier
↑ Toll-like receptor 4 (TLR4) expression on intestinal epithelial cells
Aberrant bacterial colonization of gut
          Impaired gut barrier allows for ↑ bacterial translocation across intestinal epithelium
TLR4 on intestinal mesentery endothelial cells bind lipopolysaccharides (LPS) on Gram-negative gut bacteria
Immune cells release proinflammatory mediators (TNF, IL-12, IL-18)
Cytokines mediate ↑ enterocyte apoptosis (including enteric stem cells) and ↓ enterocyte proliferation
Intestinal mucosa healing is impaired, leading to local inflammation & injury
TLR4 on intestinal epithelial cells binds LPS from Gram-negative gut bacteria
Authors: Rachel Bethune Naima Riaz Reviewers: Nicola Adderley Michelle J. Chen Kamran Yusuf* Jean Mah* * MD at time of publication
      Endothelial nitric oxide synthase expression is reduced
Vasoconstriction from ↓ NO reduces blood flow to intestines
Prolonged ↓ in O2 perfusion results in irreversible intestinal mucosal cell death (necrosis)
Gas escapes into abdominal cavity
Leakage of intestinal contents irritates parietal peritoneum
Bacteria enter bloodstream
Pneumo- peritoneum
Abdominal distention
Peritonitis
Sepsis
                 Blood from tissue damage mixes with intestinal contents
Bloody stool
Intestinal sensory neurons detect damage and send signals to medullary vomiting centre
Bilious vomiting
Damaged intestinal cells are unable to absorb nutrients
Short gut syndrome
Persistent intestinal mucosal injury creates penetrating lesions through intestinal wall
Intestinal perforation
         Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published May 6, 2019; updated Mar 21, 2024 on www.thecalgaryguide.com

Lichen Sclerosus

Lichen Sclerosus (genital manifestation): Pathogenesis and clinical findings
Authors: Mina Youakim Reviewers: Elise Hansen Sunawer Aujla Shahab Marzoughi Jori Hardin* * MD at time of publication
Histamine receptor binding stimulates sensory nerve endings
Pruritus (itching)
    Unknown triggers
Genetic predisposition (HLA-DQ7 and HLA-DR12)
Chronic inflammation (i.e. chronic infection, chronic toxin exposure) and trauma
Medications (e.g. carbamazepine, pembrolizumab, nivolumab, ipilimumab)
  ↑ Activation of CD4+ and CD8+ T cells released from macrophages (white blood cell) in the perineum and genital skin tissue infiltrate into the dermal-epidermal junction
T-cells proliferate in a horizontal linear formation Pro-inflammatory response activation
       Fibroblasts (contributes to the formation of connective tissue) proliferate and persist producing altered collagen under the epidermis
Collagen deposits and hyalinizes (transforms into acellular translucent material) beneath the epidermal layer
Sclerotic plaques (localized areas of thickened skin)
T cells release of pro-inflammatory cytokines (interleukins and transforming growth factor β)
↑ Oxidative stress and cell damage
Progressive basal layer degeneration thins the overall skin thickness
Mast cells respond to increased need for immune cell flow to area
Nitric oxide is released when histamine binds to vascular receptors
Localized area appears red
Erythema (reddening of the skin)
Erosion/ulceration
Skin fissures (linear cleavage of skin)
 Localized histamine release
Nitric oxide induces localized vasodilation (↑ blood flow)
            Normal Skin
Lichen Sclerosus
Epidermal layer
Basal layer
Dermal-Epidermal Junction Dermal layer
Hypopigmented patches (localized, pale areas of skin)
Epidermal atrophy (crinkling paper-type skin appearance)
Thinned skin is weakened and prone to physical stress or trauma
↑ fluid in the extracellular space due to capillary leakage from ↑ blood flow
Superficial dermal edema (swelling)
Fibrotic tissue deposition following healing of damaged tissue
            Atrophic Epidermal layer
Hyalinized collagen deposits
Basal layer degeneration Band of T-cell infiltrate
Dermal layer
Scarring
          Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Mar 25, 2024 on www.thecalgaryguide.com
   
Lichen Sclerosus (genital manifestation): Pathogenesis and clinical findings
Authors: Mina Youakim Reviewers: Elise Hansen Sunawer Aujla Shahab Marzoughi Jori Hardin* * MD at time of publication
Histamine receptor binding stimulates sensory nerve endings
Pruritus (itching)
    Unknown triggers
Genetic predisposition (HLA-DQ7 and HLA-DR12)
Chronic inflammation (i.e. chronic infection, chronic toxin exposure) and trauma
Medications (e.g. carbamazepine, pembrolizumab, nivolumab, ipilimumab)
  ↑ Activation of CD4+ and CD8+ T cells released from macrophages (white blood cell) in the perineum and genital skin tissue infiltrate into the dermal-epidermal junction
T-cells proliferate in a horizontal linear formation Pro-inflammatory response activation
       Fibroblasts (contributes to the formation of connective tissue) proliferate and persist producing altered collagen under the epidermis
Collagen deposits and hyalinizes (transforms into acellular translucent material) beneath the epidermal layer
Sclerotic plaques (localized areas of thickened skin)
T cells release of pro-inflammatory cytokines (interleukins and transforming growth factor β)
↑ Oxidative stress and cell damage
Mast cells respond to increased need for immune cell flow to area
Nitric oxide is released when histamine binds to vascular receptors
↑ fluid in the extracellular space due to capillary leakage from ↑ blood flow
Superficial dermal edema (swelling)
Epidermal atrophy (crinkling paper- type skin appearance)
Thinned skin is weakened and prone to physical stress or trauma
Localized histamine release
Nitric oxide induces localized vasodilation (↑ blood flow)
Localized area appears red
Erythema (reddening of the skin)
Erosion/ulceration
Skin fissures (linear cleavage of skin)
           Normal Skin
Lichen Sclerosus
Epidermal layer
Basal layer
Dermal-Epidermal Junction Dermal layer
Atrophic Epidermal layer
Hyalinized collagen deposits
Basal layer degeneration Band of T-cell infiltrate
Dermal layer
Progressive basal layer degeneration thins the overall skin thickness
Hypopigmented patches (localized, pale areas of skin)
Fibrotic tissue deposition following healing of damaged tissue
                        Scarring
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published MONTH, DAY, YEAR on www.thecalgaryguide.com
   
Chronic inflammation (i.e. chronic
Reviewers:
    Dermal layer
     Unknown triggers
Genetic predisposition (HLA-DQ7 and HLA-DR12)
infection, chronic toxin exposure) and trauma
Medications (e.g. carbamazepine, pembrolizumab, nivolumab, ipilimumab)
Elise Hansen Sunawer Aujla Shahab Marzoughi Jori Hardin* * MD at time of publication
  ↑ Activation of CD4+ and CD8+ T cells released from macrophages (white blood cell) in the perineum and genital skin tissue infiltrate into the dermal-epidermal junction
T-cells proliferate in a horizontal linear formation
Pro-inflammatory response activation
↑ Expression of MicroRNA-155 (enhances pro-inflammatory response and ↓ expression of tumor suppression genes)
     Fibroblasts (contributes to the formation of connective tissue) proliferate and persist producing altered collagen
Collagen deposits and hyalinizes (transforms into acellular translucent material) beneath the epidermal layer
Sclerotic plaques (localized areas of thickened skin)
T cells release of pro-inflammatory cytokines (interleukins and transforming growth factor β)
↑ Oxidative stress and cell damage
Mast cells respond to increased need for immune cell flow to area
Nitric oxide is released when histamine binds to vascular receptors
↑ fluid in the extracellular space due to capillary leakage from ↑ blood flow
Superficial dermal edema (swelling)
Epidermal atrophy (crinkling paper- type skin appearance)
Thinned skin is weakened and prone to physical stress or trauma
Lichen Sclerosus
Localized histamine release
Nitric oxide induces localized vasodilation (↑ blood flow)
Localized area appears red
Erythema (reddening of the skin)
Erosion/ulceration
Skin fissures (linear cleavage of skin)
Atrophic Epidermal layer
Hyalinized collagen deposits
Basal layer degeneration Band of T-cell infiltrate
Histamine receptor binding stimulates sensory nerve endings
Pruritus (itching)
                           Normal Skin
Progressive basal layer degeneration thins the overall skin thickness
Hypopigmented patches (localized, pale areas of skin)
Epidermal layer
Basal layer
Dermal-Epidermal Junction
Fibrotic tissue deposition following healing of damaged tissue
Scarring
         
Chronic inflammation (i.e. chronic
Reviewers:
    Dermal layer
     Unknown triggers
Genetic predisposition (HLA-DQ7 and HLA-DR12)
infection, chronic toxin exposure) and trauma
Medications (e.g. carbamazepine, pembrolizumab, nivolumab, ipilimumab)
Elise Hansen Sunawer Aujla Shahab Marzoughi Jori Hardin* * MD at time of publication
  ↑ Activation of CD4+ and CD8+ T cells released from macrophages in the perineum and genital skin tissue infiltrate into the dermal-epidermal junction
T-cells proliferate in a horizontal linear formation Pro-inflammatory response activation
↑ Expression of MicroRNA-155 (short segment of RNA which enhances pro- inflammatory response and ↓ expression of tumor suppression genes)
        Fibroblasts proliferate and persist producing altered collagen
Collagen deposits and hyalinizes (transforms into acellular translucent material) beneath the epidermal layer
Sclerotic plaques (localized areas of thickened skin)
T cells release of pro-inflammatory cytokines (interleukins and transforming growth factor β)
↑ Oxidative stress and cell damage
Progressive basal layer degeneration thins the overall skin thickness
Hypopigmented patches (localized, pale areas of skin)
Epidermal layer
Basal layer
Dermal-Epidermal Junction
Mast cells respond to increased need for immune cell flow to area
Nitric oxide is released when histamine binds to vascular receptors
Superficial dermal edema (swelling)
Epidermal atrophy (crinkling paper- type skin appearance)
Localized histamine release
Nitric oxide induces localized vasodilation (↑ blood flow)
Localized area appears red
Erythema (reddening of the skin)
Histamine receptor binding stimulates sensory nerve endings
Pruritus (itching)
                  Normal Skin
Lichen Sclerosus
Skin fissures (linear cleavage of skin)
Erosion/ulceration
   Fibrotic tissue deposition following healing of damaged tissue
Scarring
Atrophic Epidermal layer
Hyalinized collagen deposits
Basal layer degeneration Band of T-cell infiltrate
         
Reviewers:
    Dermal layer
     Unknown triggers
Genetic predisposition (HLA-DQ7 and HLA-DR12)
Chronic inflammation (i.e. chronic infection, chronic toxin exposure) and trauma
Medications (e.g. carbamazepine, pembrolizumab, nivolumab, ipilimumab)
Elise Hansen Sunawer Aujla Shahab Marzoughi Jori Hardin* * MD at time of publication
  ↑ Activation and infiltration of CD4+ and CD8+ T cells into the dermal-epidermal junction
T-cells proliferate in a band (horizontal linear) formation Pro-inflammatory response activation
   ↑ Expression of MicroRNA-155 (short segment of RNA which enhances pro-inflammatory response and ↓ expression of tumor suppression genes)
     Fibroblasts proliferate and persist producing altered collagen
Collagen deposits and hyalinizes (transforms into acellular translucent material) beneath the epidermal layer
Sclerotic plaques (localized areas of thickened skin)
T cells release of pro-inflammatory cytokines (interleukins and transforming growth factor β)
↑ Oxidative stress and cell damage
Progressive basal layer degeneration thins the epidermis
Hypopigmented patches (localized, pale areas of skin)
Epidermal layer
Basal layer
Dermal-Epidermal Junction
Mast cells respond to increased need for immune cell flow to area
Nitric oxide is released when histamine binds to vascular receptors
Superficial dermal edema (swelling)
Epidermal atrophy (crinkling paper- type skin appearance)
Localized histamine release
Histamine receptor binding stimulates sensory nerve endings
Pruritus (itching)
                    Skin fissures (linear cleavage of skin)
Nitric oxide induces localized vasodilation (↑ blood flow)
Localized area appears red
Erythema (reddening of the skin)
Scarring
Atrophic Epidermal layer
Hyalinized collagen deposits
Basal layer degeneration Band of T-cell infiltrate
 Erosion/ulceration
   Fibrotic tissue deposition following healing of damaged tissue
 Normal Skin
Lichen Sclerosus
        
Chronic inflammation (i.e. chronic
Elise Hansen
      Unknown triggers
Genetic predisposition infection, chronic toxin exposure) Medications (e.g. carbamazepine, (HLA-DQ7 and HLA-DR12) and trauma pembrolizumab, nivolumab, ipilimumab)
↑ Activation and infiltration of CD4+ and CD8+ T cells into the dermal-epidermal junction
Sunawer Aujla Shahab Marzoughi Name Name* * MD at time of publication
   T-cells proliferate in a band (horizontal linear) formation
Pro-inflammatory response activation (increase in pro-inflammatory cytokines such as Interleukins 1-alpha and 1-beta)
↑ Expression of MicroRNA-155 (short segment of RNA which enhances pro-inflammatory response and ↓ expression of tumor suppression genes)
      Fibroblasts proliferate and persist producing altered collagen
T cells release of pro-inflammatory cytokines (interleukins and transforming growth factor β)
↑ Oxidative stress and cell damage
Progressive basal layer degeneration thins the epidermis
Hypopigmented patches (localized, pale areas of skin)
Histamine receptor binding stimulates sensory nerve endings
Localized area appears red
 Localized histamine release
Localized vasodilation (↑ blood flow)
Superficial dermal edema (swelling)
Pruritus
Erythema
          Collagen deposits and hyalinizes (transforms into acellular translucent material) beneath the epidermal layer
Normal Skin
Sclerotic plaques (localized areas of thickened skin)
Epidermal layer
Basal layer
Dermal-Epidermal
Junction Dermal layer
Skin fissures (linear cleavage of skin)
Bleeding
Erosion/Ulceration
Epidermal atrophy (crinkling paper- type skin appearance)
           Lichen Sclerosus
Atrophic Epidermal layer
Hyalinized collagen deposits
Basal layer degeneration
Band of T-cell infiltrate
Dermal layer
Fibrotic tissue deposition following healing of damaged tissue
Scarring
             
 Lichen Sclerosus (genital manifestation): Pathogenesis and clinical findings
Authors: Mina Youakim Reviewers: Elise Hansen Sunawer Aujla Shahab Marzoughi Name Name* * MD at time of publication
Pruritus
Histamine receptor binding stimulates sensory nerve endings
Localized area appears red
Erythema
    Unknown triggers
Genetic predisposition (HLA-DQ7 and HLA-DR12)
Chronic inflammation and trauma
Medications (e.g. carbamazepine, pembrolizumab, nivolumab, ipilimumab)
  ↑ Activation and infiltration of CD4+ and CD8+ T cells into the dermal-epidermal junction
T-cells proliferate in a band formation Pro-inflammatory response activation
   ↑ Expression of MicroRNA-155 (short segment of RNA which enhances pro-inflammatory response and ↓ expression of tumor suppression genes)
     Fibroblasts proliferate and persist producing altered collagen
T cells release of pro-inflammatory cytokines (interleukins and transforming growth factor β)
Localized histamine release
Localized vasodilation (↑ blood flow)
   Superficial dermal edema (swelling)
     Collagen deposits and hyalinizes beneath the atrophic epidermal layer
Hypopigmented patches (localized, pale areas of skin)
↑ Oxidative stress and cell damage
Progressive basal layer degeneration thins the epidermis
Skin fissures
Lichen Sclerosus
Epidermal atrophy (crinkling paper- type skin appearance)
          Sclerotic plaques (localized areas of thickened skin)
Bleeding Scarring
Erosion/Ulceration
  Normal Skin
Epidermal layer
Basal layer
Dermal-Epidermal
Junction Dermal layer
Atrophic Epidermal layer
Hyalinized collagen deposits
Basal layer degeneration
Band of T-cell infiltrate
Dermal layer
            Legend:
Published MONTH, DAY, YEAR on www.thecalgaryguide.com
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
Complications

Post-Renal Acute Kidney Injury AKI

Post-Renal Acute Kidney Injury (AKI): Pathogenesis and clinical findings
          Blood clot or cellular debris
Foreign body
Neurogenic bladder
Obstruction intrinsic to urine excretion system
Nephrolithiasis (kidney stones)
Anatomical defect(s)
Intra- abdominal adhesions
Retroperitoneal fibrosis (scar-like tissue)
Benign or malignant masses
Prostate cancer
Benign prostatic hyperplasia
      ↓ Urine flow across point of obstruction Obstruction extrinsic to urine excretion system
         Urine buildup distends urine collecting system (hydronephrosis)
Compression of renal vasculature due to mass effect
↑ Volume/pressure proximal to obstruction
↑ Intratubular pressure
↓ Pressure gradient between glomerular afferent arteriole and Bowman’s space
Casts occlude tubules
↓ Filtration of plasma into nephrons
↓ Glomerular Filtration Rate (GFR)
Obstruction is relieved ↓ Intratubular pressure Rapid GFR ↑
Rapid diuresis of fluid and electrolytes
  Dilated pelvicalyceal system on ultrasound
↓ Urine output
          ↓ Venous drainage
and arterial supply
Local ischemia and inflammation of kidney
Impaired resorption, excretion, and fine tuning by tubules
Acute Tubular Necrosis (ATN) with granular casts
     ↓ Urine output
↓ Clearance of free water and solutes
↑ Intravascular volume
↑ Serum creatinine
↓ Medullary solute concentration, ischemia, ↓ response of collecting ducts to antidiuretic hormone
Lasts > 24hrs
Post-obstructive diuresis causes hypovolemia and electrolyte derangements
Authors: David Campbell, Matthew Hobart Reviewers: Raafi Ali, Luiza Radu Huneza Nadeem, Marissa Zhang, Julian Midgley* * MD at time of publication
Resolves < 24hrs in euvolemia
Physiologic post- obstructive diuresis
            ↑ Na+ and Cl- delivery to distal convoluted tubule is sensed by macula densa
Secretion of adenosine by macula densa
Adenosine constricts afferent arterioles
↓ GFR
↓ Renal clearance of drugs and waste products
       ↑ Venous hydrostatic pressure
↑ Volume in arterial system overwhelms pressure regulation mechanisms
Hypertension
   Fluid extravasation from veins and capillaries
   Generalized Edema
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Mar 25, 2024 on www.thecalgaryguide.com

Irritant Contact Dermatitis Pathogenesis and Clinical Findings

Irritant Contact Dermatitis: Pathogenesis and clinical findings
Authors: Zaini Sarwar, Mina Youakim Reviewers: Shahab Marzoughi, Ryan T. Lewinson, Yan Yu*, Laurie M. Parsons* * MD at time of publication
Repeated/chronic exposure causes damage to cell membranes
Skin barrier disruption
Chronic non-specific inflammatory response
Repetitive keratinocyte cytokine-mediated injury
Keratinocytes exhibit ↑ proliferation as a compensatory response
Rapid turnover of stratum corneum (outermost layer of the epidermis)
Hyperkeratotic skin is less amenable to skin stretching and pressure
Skin fissures (cracks in the skin)
   Irritant agents
(abrasives, cleaning solution, oxidative & reducing agents, dust, soils, water)
Acute exposure triggers inflammatory response
Keratinocytes undergo cytotoxic damage with ↑ neutrophil & cytokine release
Common occupational exposures (housekeeping, cleaning, catering, medical/dental, construction)
Risk factors
(atopy, fair skin, low temperature, low humidity)
     Stimulation of local nociceptors (free nerve endings extend into the mid epidermis)
         Perivascular (around the blood vessel) inflammation causes histamine release from mast cells
Damaged keratinocytes are destroyed via
apoptosis (programmed cell death)
Epidermal Necrosis (death of epidermal tissue)
Shedding of necrotic tissue
Ulceration (deep open wound on skin)
Burning
pain (uncomfortable stinging sensation)
Pruritus (itching)
           Histamine causes local blood vessel dilation and ↑ blood flow to the area of skin affected
Erythema (area appears red from ↑ blood flow)
Burning & Itching Spongiosis
Neutrophils Neutrophils
Histamine causes local blood vessel walls to have
↑ permeability, thereby ↑ leakage of fluid
Spongiosis
(↑ fluid between keratinocytes in the epidermis)
Fluid continues to build up from ongoing inflammation
Vesiculation (fluid collections in the epidermis)
Long-term skin scratching causes chronic irritation which eventually hardens the skin
Lichenification
(thick, hardened patches of skin)
↑ Overall keratin production
Hyperkeratosis (thickening of the outermost skin layer)
             Further fluid buildup bursts vesicles leaving behind erosions and dried crust on the epidermis
     Crust (scaling over the skin)
Lichenification
Erosions (open sore on skin)
  Ulcer
Epidermis
  Perivascular Inflammation
Hyperkeratosis
Dermis
Dermal-epidermal junction
 Legend:
 Pathophysiology
Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published Oct 19, 2016; updated Mar 30, 2024 on www.thecalgaryguide.com

Deep Partial Thickness Burns Pathogenesis and Clinical Findings

   Deep Partial Thickness Burns:
Pathogenesis and clinical findings
Radiation (Sunlight, x-ray, nuclear emission/explosion)
Ionizing radiation gets into contact with DNA
Damage to keratinocytes
Fire (Flash fire or direct contact with flame)
Contact (Hot solid objects)
Scalding (Hot liquid via immersion, spill or splash)
Chemical (Strong acid, alkali or irritant gas)
Electrical (Contact with exposed electrical wiring/appliances)
     Author and Illustrator: Amanda Eslinger Maharshi Gandhi Reviewers:
Alexander Arnold
Shahab Marzoughi
Duncan Nickerson*
* MD at time of publication
Direct transfer of heat energy
Coagulation necrosis (cell death due to ischemia) is induced
Deep Partial Thickness Burn
Injury to the epidermal layer and both the papillary and a portion of the reticular layer of the dermis
     Transfer of heat energy & direct injury to cellular membranes
           Epidermis
Papillary dermis Reticular dermis
Sub- cutaneous Tissue
↑ vascular permeability due to damage
Fluid leak results in edema between dermal & epidermal layer
Blisters (a bubble on the skin containing fluid)
Thin epidermal layer forming fluid-filled vesicle breaks open
Cutaneous capillary bed is destroyed
Blood flow to injured area is compromised
Pressing on the skin doesn’t easily force blood cells out of the area
Non-blanchable skin
Vasodilation in fascia (a thin casing of connective tissue) underlying subcutaneous tissue
Inflammatory mediators such as cytokines and prostaglandins activate fibroblasts
Collagen deposition and subsequently induration (thickening of skin)
Some healthy dermal appendages surrounded by islands of undamaged epithelial cells
Possible spontaneous re-epithelialization in 2-9 weeks
During re-epithelization of burns there can be excessive deposition of collagen and other extracellular matrix components
Thick raised scars in dermis due to excess collagen deposition
Somatosensory structures (nociceptors, thermoreceptors, and mechanoreceptors) are completely injured within the dermis
Analgesia of impacted area (the inability to feel pain)
               Ongoing Inflammation and dilated blood vessels
Red Induration (Thickened skin appearing red)
Compromised blood flow and more extensive tissue damage
White Induration (Thickened skin appearing white)
      Moist Wound
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Dec 2, 2013, updated Apr 20, 2024 on www.thecalgaryguide.com

Costochondritis

Costochondritis: Pathogenesis and clinical findings
Systemic infection originating in upper respiratory tract (commonly Staphylococcus aureus and Streptococcus bacterial infections)
Infection spreads through blood to ribcage and chest wall
    Age-related degeneration of cartilage within ribcage joints
Immune cells present in joints secrete proinflammatory cytokines
Cytokines sensitize nociceptors (pain receptors) in joints
Repetitive trauma or strain to chest wall muscles (e.g. chronic cough, chest wall injury, overexertion of chest wall muscles)
Excessive stretch and tearing of ribcage cartilage and ligaments
Autoimmune disease (e.g. rheumatoid arthritis)
Antigen presenting cells display self-antigens to CD4+ T cells
           Mechanical stimuli (e.g. Damaged cells release cytokines Pathogens activate immune cells at site of stretch) activate nociceptors that recruit immune cells inflammation
Costochondritis
 Benign inflammation of the ribcage (costal) cartilage, particularly at the costosternal junctions (connection between sternum and cartilage) and the costochondral junctions (connection between the cartilage and rib)
Authors:
Michelle J. Chen Reviewers:
Raafi Ali
Yan Yu*
Gerhard Kiefer*
* MD at time of publication
Cartilage, ligament, or muscle injury
Presence of foreign pathogen in costal cartilage
     Injury or pathogen activates inflammatory cascade in the ribcage cartilage
      Immune cells proliferate so that they exceed the available space in the cartilage matrix of the ribcage
Cartilage swelling compresses intercostal nerves
Compression acts as a mechanical stimulus to activate nociceptors
Sharp, localized pain reproducible upon palpation over ribcage
Immune cells secrete cytokines
Cytokines activate nociceptors so that they become more sensitive to mechanical stimuli
Muscle or ligament stretch from normal use activates sensitized nociceptors
Pain increases with inspiration or cough
Natural resolution of inflammation over time
     Macrophages phagocytose apoptotic cells (immune and cartilage cells) and cellular debris
Immune cells release factors that degrade proinflammatory cytokines
Fewer cytokines reduce immune cell recruitment into costal cartilage
Pain usually self-resolves
Immune cells release lipid molecules that bind to the same cells (autocrine signaling) to inhibit further production of inflammatory cytokines and chemokines
         Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Apr 29, 2024 on www.thecalgaryguide.com

Acute Otitis Media Complications

Acute Otitis Media: Complications Prolonged mucus buildup or swelling in
Eustachian tube due to colds/allergies obstructs the Eustachian tube
Fluid unable to drain through tube and accumulates in middle ear
Acute Otitis Media
Infection and inflammation of the middle ear
Mastoid air cells are in physical contact with distal middle ear
Pathogens move into mastoid air spaces
Inflammation & infection in air spaces
Mastoiditis
Bacterial and immune cell debris (pus) accumulates in mastoid air spaces
Untreated middle ear infection allows further bacterial proliferation
           Persistent effusion
causes ion channel changes in inner ear
Composition of endolymph and perilymph in inner ear changes
Vestibular/Labyrinth dysfunction
Feelings of Vertigo imbalance
Purulent discharge from middle ear through perforation
Otorrhea (ear discharge)
↑ Pressure in middle ear
Pressure stretches tympanic membrane
Cytokines reach hypothalamus through the bloodstream
Hypothalamus responds to stimulation and ↑ thermoregulatory set-point
High-grade fever
Infection spreads into bloodstream
Sepsis
Cytokines alter metabolism pathways of neurotransmitters in the brain
Cerebral cortex dysfunction
Infection spreads to cranium
Intracranial complications (e.g., meningitis, brain abscess, thrombus)
Author: Jody Platt Stephanie de Waal Reviewers: Yan Yu Elizabeth De Klerk William Kim Annie Pham Michelle J. Chen Danielle Nelson* * MD at time of publication
Helper T cells and macrophages release inflammatory cytokines into the bloodstream
                    Perforation of tympanic membrane
↓ Conduction of sound waves
Conductive hearing loss
              Pressure in middle ear diffuses out of perforation
↓ Tympanic membrane stretching
Otalgia (ear pain) fades
Accumulated debris compresses cranial nerve VII
Facial nerve palsy
Mastoid abscess
    Febrile seizures
    Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Feb 28, 2013, updated Apr 29, 2024 on www.thecalgaryguide.com

Sickle Cell Disease Pathogenesis Clinical Findings and Complications

Sickle Cell Disease: Pathogenesis, Clinical Findings, and Complications
DNA point mutation in chromosome 11 causes a substitution of glutamate to valine for the sixth amino acid of the β-globin chain
Authors: Yang (Steven) Liu Priyanka Grewal Reviewers: Alexander Arnold Luiza Radu JoyAnne Krupa Yan Yu* Lynn Savoie* * MD at time of publication
  Hemoglobin S (HbS) variant formed instead of normal Hemoglobin A (HbA)
  Hb electrophoresis shows approximately 45% HbS, 52% HbA (ααββ), 2% HbA
Hb electrophoresis shows approximately 90% HbS, 8% HbF, & 2% HbA .
No HbA present
  (ααδδ), & 1% HbF (ααγγ;2 fetal hemoglobin)
Heterozygous: point mutation in one of the two chromosomes (Hb AS)
Sickle cell trait
(asymptomatic unless severely hypoxic)
Homozygous: point mutation in both chromosomes (Hb SS)
Sickle cell disease
An inherited blood disorder characterized by defective hemoglobin that leads to red blood cells sickling
2
      Dehydration Hypoxemia
Acidosis
↓ Volume of RBC cytoplasm
↓ O2 Saturation of Hb
O morereadily 2
released from Hb in low pH environment
↑ Concentration of deoxygenated HbSinred blood cells (RBCs)
Hydrophilic glutamate→ hydrophobic valine substitution makes HbS less soluble in the cytoplasm & more prone to polymerization & precipitation in its deoxygenated state
↑ Concentration of deoxygenated Hb in RBC leads to ↑ polymerization rate Polymerized & precipitated HbS forms long needle-like fibers
        RBC shape becomes sickled
Sickle cells on peripheral blood smear
     Vaso-occlusion
(sickled RBCs lodge in small vessels, blocking bloodflow to organs & tissues)
Blockage of venous outflow
Occlusion of vessels in lungs ↑ pulmonary blood pressure
Fluid extravasates into interstitial tissue leading to pulmonary edema
Acute chest syndrome (chest pain, hypoxemia (↓blood oxygen), etc.)**
to the penis:
to the spleen:
Priapism (persistent, painful erection)
Splenic Sequestration (blood pools in spleenà splenomegaly & hypotension)
Extravascular hemolysis (macrophages in the spleen phagocytose sickled RBCs)
Normocytic anemia
↑Marrow erythropoiesis (RBC production) to compensate for hemolysis
                  Infarction of bone
Pain crises
If occurring in hands
Dactylitis (inflammation of digits)
Blockage of arteries ↓ oxygenation of organs & tissues
Vaso-occlusion of the splenic artery
Splenic infarction (↓ blood supply leads to tissue death)
RBC inclusions (structures found in RBCs) not removed by spleen
Howell-Jolly bodies (RBC DNA remnants) on blood smear
Vaso-occlusion of other arteries (cranial, renal, etc)
Stroke, renal failure
↑ RBC breakdown
↑ Unconjugated bilirubin released from RBC breakdown
RBC precursors (reticulocytes) are released into the blood stream
Reticulocytosis (increased number of immature red blood cells) on peripheral blood smear
Patient’s RBC level becomes dependent on increased marrow activity
Bone marrow infarction or viral infections (i.e. Parvovirus B19) suppresses bone marrow activity
Aplastic crises (profound anemia)
       ** See corresponding Calgary Guide slide(s)
↑ Serum level of unconjugated bilirubin
Some of the circulating unconjugated bilirubin deposits in the skin
Jaundice (yellowing of skin)
↑ Conjugation of bilirubin in liver
↑Amounts of conjugated bilirubin released into bile
Gallstone formation
Cholelithiasis (presence of gallstones in the gallbladder)
         Spleen releases invasive encapsulated bacteria (eg. Haemophilus influenzae, Streptococcus pneumoniae, Neisseria meningitidis) into circulation, which causes infections
         Meningitis
Sepsis
  Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Sept 16, 2013, updated Apr 29, 2024 on www.thecalgaryguide.com

Wiskott-Aldrich Syndrome

Wiskott-Aldrich Syndrome (WAS): Pathogenesis and clinical findings X-linked recessive inheritance (mostly males impacted) Spontaneous DNA mutation
Genetic mutations in Wiskott-Aldrich Syndrome Protein (WASP) impair actin cytoskeleton remodeling in hematopoietic cells (immature blood cells)
Authors: Mina Mina Reviewers: Hana Osman Mao Ding Maharshi Gandhi Shahab Marzoughi Louis-Philippe Girard* * MD at time of publication
          Megakaryocytes (platelet precursor cells) depend on the cytoskeleton changing shape to form pseudopodia (arm-like projections) which bud off forming cellular fragments (platelets)
Abnormal cytoskeleton reorganization leads to ineffective thrombocytopoiesis (production of platelets)
Microthrombocytopenia (↓ platelet size and quantity)
Issues with formation of a platelet plug due to abnormal platelets (dysfunction of primary hemostasis)
Reduced ability for platelet adhesion, activation, and/or aggregation
Natural killer cells depend on the cytoskeleton reorganization to form immunological synapses (communication) with body cells in order to have effective surveillance
T cells depend on the cytoskeleton remodeling to form pseudopods which allow them to synapse with other cells when a pathogen is encountered
    Defective immune synapse formation
↓ Cancer immunosurveillance
↑ Risk of malignancy
Helper T cells cannot activate B cells which generate antibodies (immunoglobulins) to destroy pathogens
Regulatory T cells can not sufficiently downregulate effector cells which typically limit the immune response and prevent autoimmune conditions
↑ Risk of autoimmune diseases
Recurrent opportunistic infections
       Immune dysregulation
Impaired immune response
        ↓ Number and function of T cells
Immune responses contribute to Type 2 immune responses
Atopic dermatitis (AD; chronic inflammation that causes itchy skin)
↑ Immunoglobulin (Ig) A
      Epistaxis (nosebleed)
Red blood cells leak from capillaries
Menorrhagia (heavy menstrual bleeding)
  Petechiae (small, flat, red spots that appear on the skin)
Inflammation in AD triggers release of interleukins (modulatory proteins during inflammatory and immune responses) leading to Immunoglobulin (Ig) class switching
 ↑ IgE levels
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Apr 20, 2024 on www.thecalgaryguide.com

Scabies pathogenesis and clinical findings

 Scabies: Pathogenesis and clinical findings
Direct person-person transmission of Sarcoptes Scabiei var. hominis to new host
Fertilized females secrete proteolytic enzymes that allow them to burrow through the stratum corneum (2 mm/day)
Authors: Heena Singh Amanda Eslinger Nirav Saini Reviewers: Shahab Marzoughi Danny Guo Yan Yu Richard Haber* * MD at time of publication
Mechanical irritation and immunological reaction
Mast cell activation and histamine release
Itch
(> 10 Mites)
     Stratum corneum
Stratum lucidum Stratum granulosum
Stratum spinosum Stratum basale
Females lay 2-3 ova/day which hatch in the stratum corneum in pockets after 3 days
The larvae molt and mature for 2 weeks and mate within the pockets of the stratum corneum
Following mating, female mites begin burrowing
Cycle is propagated as new female mites create more burrows in stratum corneum
Superficial, Linear, Tortuous (i.e., Twisted) +/- Scaly Burrows
         ↑ Exposure to mite antigens such as Sar s 14.3 and Sar s 14.2
Type 2 helper T-cells produce proteins IL-4, IL-5, IL-13
↑ Serum Immunoglobulin G & E ↑ Eosinophil Count in Peripheral Blood Inappropriate T helper-type immune response in skin (mechanism unknown)
Antigens forming from mite feces, ova, or decomposing bodies
Type IV hypersensitivity reaction (delayed immune response 2-4 weeks following initial contact)
            Skin barrier dysfunction from inflammation and histamine release
Pre-existing immunosuppression Chronic inflammatory response Allergic reaction to ↑ mite activity at night
     Uncontrollable Itch (Worse At Night) + Excoriations (Scratch Marks)
  Urticarial (Hive-Like) Crusted Papules Eczematous Plaques
Skin breakdown Opening for bacteria (commonly S. aureus) 2° Bacterial infection Pustules
       Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Oct 17, 2013; updated Jun 9, 2024 on www.thecalgaryguide.com

Posterior Cruciate Ligament PCL Injury Pathogenesis and Clinical Findings

Posterior Cruciate Ligament (PCL) Injury: Pathogenesis and clinical findings
Authors: Luc Wittig Stephanie de Waal Michelle J. Chen Reviewers: Reza Ojaghi Usama Malik Sunawer Aujla Nojan Mannani Mankirat Bhogal Dr. R. Buckley* Dr. Gerhard Kiefer* * MD at time of publication
  Sport-related trauma
Dashboard knee injury from a motor vehicle collision
Tibia contacts dashboard at high velocity with knee in flexed position
Tibia is forced posteriorly, relative to knee joint The PCL undergoes sudden & forceful strain
     Hyperextension injury (knee joint moves past its normal extension limit)
Extreme tibia movement anteriorly past knee joint overstretches PCL
Hard fall on flexed knee
      Posterior Cruciate Ligament Injury
Strain & overstretching of the PCL, which connects the anterior distal femur to the posterior proximal tibia to prevent posterior translocation of the tibia relative to the femur, can result in a partial or complete tear. The tear can be isolated (rare) or be one of multiple ligaments that are torn (multi-ligamentous tear).
     Mild injurious force causes a partial tear
Intact portion of PCL prevents extreme tibial translocation posterior to the femur. Torn portion still allows 1-5 mm of posterior tibial translocation (Grade 1)
Positive posterior drawer test
Knee injury tests
• Posterior drawer test – Push against leg below
the knee to test for posterior tibial translocation
relative to femur
• Lachman test – Pull leg below the knee to test
for anterior tibial translocation relative to femur • McMurray test – Tests for medial and lateral
meniscus tears
• Varus & valgus stress test – Tests for medial and
Strong injurious force causes a complete, isolated tear
PCL is unable to prevent posterior translocation of the tibia relative to the femur, allowing 6-10 mm of posterior tibial translocation (Grade 2)
Positive posterior Negative Lachman, McMurray, drawer test and varus & valgus stress test
Very strong injurious force causes a multi- ligamentous tear and damage to the knee joint capsule (capsuloligamentous injury)
Absence of stability from PCL combined with absence of stability from other knee ligaments allows for > 10 mm of posterior tibial translocation (Grade 3)
Positive posterior Positive Lachman OR McMurray drawer test OR varus & valgus stress test
                PCL unable to stabilize knee by preventing posterior tibial translocation (chronic PCL insufficiency)
    Body uses quadriceps tendon for knee stabilization to compensate for PCL insufficiency
Inflammation from injury contributes to progressive degenerative articular changes
  lateral collateral ligament tears
Post-traumatic patellofemoral pain
Osteoarthritis
 Legend:
 Pathophysiology
Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Sept 20, 2018; updated Jun 9, 2024 on www.thecalgaryguide.com

Migraines and auras pathogenesis and clinical findings

Migraines and Auras: Pathogenesis and clinical findings Genetic mutations at certain loci (e.g., Familial hemiplegic
 migraine mutation in gene encoding P/Q-type Ca2+ channels)
    Personal triggers (e.g., lack of food, emotional stress)
“Cortical Spreading Depolarization” followed by “Cortical Spreading Depression” across one cortical hemisphere
Cortical spreading depression
creates “auras” via unknown mechanism(s)
Expanding scotoma (area of visual blurriness)
Spreading paraesthesias (numbness that travels across body)
Dysphasic language (trouble finding words)
Brainstem symptoms (e.g., vertigo, tinnitus)
Motor symptoms (e.g., hemiplegia)
 Triggering of a depolarization wave in a unilateral region of the cortex with associated ↑ blood flow to that region
Neurons and glia release K+ into extracellular space that spreads depolarization wave to nearby cortical areas
Ion gradient imbalances cause Prolonged vasoconstriction neurons in original cortical area to and ↓ blood flow
swell and become inhibited
              Activation of the hypothalamus (responsible for maintaining homeostasis)
Prodromal symptoms (↑ thirst, hunger, yawning, ↓ cognitive function)
Author:
Yan Yu
Braxton Phillips
Reviewers:
Shahab Marzoughi
Owen Stechishin
Dustin Anderson
Scott Jarvis*
Sina Marzoughi*
* MD at time of publication
Release of hypothalamic neurotransmitters (e.g., orexins, neuropeptide Y) at the trigeminalcervical complex (TCC) of the brainstem and cervical spinal cord
Depolarizing wave passes over pseudounipolar neurons (neurons with two axons) of the trigeminal ganglion which synapse in brainstem & dura matter
       These neurotransmitters reduce the activation threshold of spinal trigeminal nucleus neurons in the TCC
Neuropeptides (e.g., calcitonin gene-related peptide, pituitary adenylate cyclase-activating polypeptide)
are released at the TTC, triggering local inflammation (termed neurogenic inflammation)
Ongoing neurogenic inflammation activates secondary nociceptive neurons in the TTC
Central sensitization (↓ response threshold of secondary nociceptive neurons in the TTC)
Brain perceives referred pain from the face as the TTC also receives convergent nociceptive input from the face
Facial allodynia (pain with normally non-painful stimuli)
Serotonin & histamine are released on dural blood vessels triggering neurogenic inflammation in the dura matter
Ongoing neurogenic inflammation activates primary nociceptive neurons in the dura matter
Peripheral sensitization (↓ response threshold of primary nociceptive neurons around the dural blood vessels_
Unknown mechanisms no longer believed to be related to dural blood vessel dilation
Unilateral throbbing headache
          Legend:
 Pathophysiology
 Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published Nov 22, 2012, updated Jul 1, 2024 on www.thecalgaryguide.com

Legg Calve Perthes Disease

Legg-Calvé-Perthes Disease: Signs and Symptoms
    Idiopathic abnormalities in the
common pathway of the blood clotting cascade ↑ blood thickness
Initial stage (6 months)
Early interruption of blood supply to femoral head
↓ Venous outflow from intraosseous vasculature ↑ pressure within bone
Fragmentation stage (8 months)
Body replaces resorbed necrotic bone with structurally weaker bone
Backup of blood within bone vasculature impedes arterial inflow
Temporary interruption of blood supply to proximal femoral epiphysis primarily during childhood
Residual stage
Differential growth of proximal femoral epiphysis
Overgrowth of greater trochanter
        Healing stage (4 years)
Body gradually replaces necrotic bone with stronger, more dense bone
                      Lack of O2 & nutrients disrupts normal bone growth
Bone tissue begins dying from loss of O2 & nutrients (osteonecrosis)
Osteonecrosis is associated with inflammation of synovial tissue (synovitis) at the femoral head
Metaphyseal cyst (area of bone resorption) grows on proximal femoral neck metaphysis
Growth plate abnormalities
Femoral bone head deformity
Limited hip internal rotation & abduction
Antalgic gait
Femoral epiphysis collapses laterally and extrudes from acetabulum
Femoral head now composed of dense and less dense bone
Scattered radiolucency & radiodensity on plain radiographs
Femoral head deformity becomes set in place and limits hip range of motion
Femoral bone head deformity
Limb length discrepancy
Normal bone density on plain radiographs
Abnormal articulo- trochanteric distance (vertical distance between highest point of greater trochanter & highest point of femoral head)
         Remodeling of femoral head & acetabulum seen on plain radiographs
          Growth plate irregularity
↓ Range of motion
Widened medial joint space
Antalgic gait
Femoral head is displaced laterally
Fragmented epiphysis on plain radiographs
Groin pain & referred pain to anteromedial thigh or knee region
Authors: Janelle Wai Michelle J. Chen Reviewers: Patrick Pankow Nojan Mannani Kirat Bhogal Dr. Gerhard Kiefer* * MD at time of publication
  Findings on magnetic resonance imaging (MRI) & plain radiographs
  Legend:
 Pathophysiology
Mechanism
 Sign/Symptom/Lab Finding
 Complications
 Published July 19, 2024 on www.thecalgaryguide.com

Acute Otitis Media Pathogenesis and Clinical Findings in Children

Acute Otitis Media in Children: Pathogenesis and clinical findings
      Congenital conditions (e.g. Down Syndrome, Pierre Robin syndrome)
Exposure to tobacco smoke
Impaired macrophage function in nasopharynx
Lack of immunizations
Lack of breastfeeding
Infant does not receive antibodies from breast milk
Overcrowding
Age 6 – 16 months
        Immune system deficiencies
Close proximity of kids & ↓ sanitation (e.g. daycare)
↑ Infection risk Upper Respiratory Tract Infection (i.e. bacterial (Streptococcus pneumoniae, Haemophilus influenzae, Moraxella catarrhalis), viral)
Immature Eustachian tube anatomy which facilitates pathogen transmission to middle ear
Author: Jody Platt Stephanie de Waal Reviewers: Yan Yu Elizabeth De Klerk William Kim Annie Pham Michelle J. Chen Danielle Nelson* * MD at time of publication
Abnormal anatomical structure (e.g. cleft palate)
↑ Nasopharyngeal streptococcus pneumoniae
Lack of immunity to pathogens
↑ Colonization of nasopharynx with bacterial pathogens
       Inflammation & edema of respiratory mucosa including the nose, nasopharynx, and Eustachian tube
Obstruction of the Eustachian tube
Air from middle ear resorbs into circulation which creates a low-pressure environment
Negative pressure gradient pulls viral/bacterial pathogens into middle ear
Degenerating white blood cells, tissue debris, and microorganisms accumulate & develops into purulent effusion
      Inflammation & infection of middle ear
Complications of acute otitis media**
       ↑ Pressure in middle ear
Stretching of tympanic membrane
Helper T cells & macrophages release cytokines into the bloodstream
Cytokines trigger hypothalamus to ↑ thermoregulatory set-point
Effusion behind tympanic membrane
Effusion obstructs visualization of ossicles
Neutrophils infiltrate middle ear & phagocytose pathogens
Pus accumulates behind the tympanic membrane
Blood vessels of the tympanic membrane vasodilate
             Bulging tympanic membrane
Otalgia (ear pain)
Discomfort disrupts daily activities in young children
Fever
Irritable Poor feeding
Tympanic membrane erythema
Painful blisters on tympanic membrane (e.g. Bullous myringitis)
     Can persist for up to 3 months after infection resolves
Loss of tympanic membrane landmarks (i.e. handle of malleus, light reflex)
** See corresponding Calgary Guide slide
  Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published Feb 28, 2013; updated Aug 25, 2024 on www.thecalgaryguide.com

Pediatric Pneumonia Pathogenesis and Clinical Findings

Pediatric Pneumonia: Pathogenesis and clinical findings
Authors: Jasmine Nguyen Nicola Adderley Reviewers: Midas (Kening) Kang Usama Malik Annie Pham Eric Leung* Jean Mah* * MD at time of publication
  Immunological: unvaccinated, primary immunocompromise, pre-existing illness (e.g. HIV, measles), malnutrition
Environmental: smoke, air pollution, mold, crowded housing
Recent hospitalization or antibiotic-use
Physiological: neonates, low-birth weight, underlying lung disease
  These factors make the host more susceptible to infection
Infection and proliferation of pathogen in lower respiratory tract/parenchyma
Pediatric pneumonia:
Inflammatory response to infection/proliferation of microbial pathogens at the alveolar level
Exposure to pathogen via inhalation, hematogenous, direct exposure, or aspiration
           Epithelial cells in respiratory tract release cytokines that recruit neutrophils & plasma proteins to infection site, initiating a local inflammatory response
Cytokines released into the bloodstream (e.g. TNF, IL-1) initiate a systemic inflammatory response
   ↑ Vascular permeability
Accumulation of exudate, cellular debris, serous fluid, fibrin, or bacteria in the airway spaces
↑ Respiratory drive
Tachypnea
↑ Excitability of the peripheral somatosensory system
Circulating cytokines induce prostaglandin synthesis
          Airway irritation as cilia are unable to efficiently clear fluid buildup
Crackles, ↓ breath sounds
Fluid, protein, or inflammatory cells leak into pleural space
Pleural effusion
Pulmonary edema
Fluid buildup in interstitial spaces ↑ gas diffusion distance
Bacteria enter the bloodstream (if bacterial pneumonia)
Sepsis
Fluid buildup in the alveoli ↓ available surface area for gas diffusion
↓ Efficiency of gas exchange
Intra- and extracranial arteries dilate
Headache
↑ Thermo-regulatory set-point of the hypothalamus
Fever
             Myalgia
Hypoxemia
Malaise
    Cough
    Fluid accumulation in the pleural space prevents full lung expansion
↑ Work of breathing (tracheal tug, paradoxical abdominal breathing, subcostal/suprasternal indrawing)
    Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
 Published May 28, 2018; updated Aug 25, 2024 on www.thecalgaryguide.com
  
Pediatric Pneumonia: Pathogenesis and clinical findings
Authors: Jasmine Nguyen Nicola Adderley Reviewers: Midas (Kening) Kang Usama Malik Annie Pham Eric Leung* * MD at time of publication
   Immunological: unvaccinated, primary immunocompromise, pre-existing illness (e.g. HIV, measles), malnutrition
Environmental: smoke, air pollution, mold, crowded housing
Recent Hospitalization: length of stay, recent antibiotics, mechanical ventilation
Physiological: neonates, low-birth weight, underlying lung disease (ciliary dysfunction, asthma, cystic fibrosis, bronchiectasis)
Host is more susceptible to infection
Exposure to pathogen:
inhalation, hematogenous, direct, aspiration
       Infection and proliferation of pathogen in lower respiratory tract/parenchyma
Pediatric pneumonia:
Inflammatory response to infection/proliferation of microbial pathogens at the alveolar level
Notes:
• Additional findings in pediatric pneumonia may include increased
irritability, nausea/vomiting, diarrhea,
otitis, and headache
• Viral pathogens most common in
children <2yrs; bacterial pathogens most common in children >2yrs
      Local inflammatory response: epithelial cells release cytokines in response to infection, which recruit neutrophils and plasma proteins to site of infection
↑ Vascular permeability causes accumulation of plasma exudate, cellular debris, serous fluid, fibrin, or bacteria in the airway spaces
Systemic inflammatory response:
Cytokine release (eg. TNF, IL-1)
↑ respiratory drive
          Airway irritation as cilia are unable to efficiently clear fluid buildup
Crackles, ↓ breath sounds
Fluid, protein, or inflammatory
cells leak into pleural space
Pleural effusion
Pulmonary edema
Fluid buildup in interstitial spaces increases gas diffusion distance
Fluid buildup in the alveoli decreases
available surface area for gas diffusion
↓ efficiency of gas exchange
Bacteria invade into the bloodstream (if bacterial pneumonia)
Sepsis
Hypoxemia
Circulating cytokines induce prostaglandin synthesis, which raise the thermoregulatory set-point of the hypothalamus
paradoxical abdominal breathing, subcostal/suprasternal indrawing)
            Fever
    Cough
Fluid accumulation in the pleural space prevents full
lung expansion, resulting in ↓ lung volumes
Tachypnea
↑ Work of breathing (tracheal tug,
      Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
Published Month Day, Year on www.thecalgaryguide.com
   
Pediatric Pneumonia: Pathogenesis and clinical findings
Immunological: immunization status, immune compromise
Environmental: second-hand smoke, air pollution
Hospitalization: length of stay, recent antibiotics, mechanical ventilation
Neonates, immunocompromise, underlying lung disease (ciliary dysfunction, Cystic Fibrosis, bronchiectasis)
Authors: Nicola Adderley Reviewers: Midas (Kening) Kang Usama Malik Eric Leung* * MD at time of publication
Additional findings in pediatric pneumonia may include nausea, otitis, headache
Viral pathogens most common in children <2yrs; bacterial pathogens most common in children >2yrs
Interstitial pattern: suspect Mycoplasma pneumoniae, Influenza A + B, Parainfluenza Lobar pattern: suspect S. pneumonia, H. influenzae, Moraxella, S. aureus
Systemic inflammatory response:
Cytokine release (eg. TNF, IL-1)
  Exposure to pathogen: inhalation, hematogenous, direct, aspiration
Susceptible host and/or virulent pathogen
Infection and proliferation of pathogen in lower respiratory tract/parenchyma
Pediatric pneumonia:
Inflammatory response to proliferation of microbial pathogens at the alveolar level
Notes:
     • •
• •
        Local inflammatory response: neutrophils recruited to site of infection (LOBAR or INTERSTITIAL PATTERN, depending on pathogen) by epithelial cytokine release
      Irritation of contiguous structures and/or referred pain (mechanism unclear)
Acute abdominal pain
Cough
Accumulation of plasma exudate (from capillary leakage at sites of inflammation), cell-debris, serous fluid, bacteria, fibrin
↑ respiratory drive
Disruption of hypothalamic thermoregulation
Fever/chills
         Irritation of airways and failure of ciliary clearance to keep up with fluid buildup
Crackles, ↓ breath sounds
Fluid buildup in spaces between
alveoli (INTERSTITIAL PATTERN)
Interstitial opacity on CXR
Fluid buildup in alveoli (LOBAR PATTERN)
↓ efficiency of gas exchange (↑ diffusion distance in INTERSTITIAL, ↓ surface area in LOBAR)
Hypoxemia
       Tachypnea
          Lobar consolidation on CXR
Respiratory accessory muscle use (chest indrawing, paradoxical breathing, muscle retractions)
     Legend:
 Pathophysiology
 Mechanism
Sign/Symptom/Lab Finding
 Complications
Published May 28, 2018 on www.thecalgaryguide.com
   gin