Uploaded by Максимилиан Огурцов

Osmosis Microbiology High-Yield Notes

advertisement
Medicalstudyzone.com
Table of contents
Adenoviridae 1
Aerobic rods 3
Bacillus anthracis 3
Bacillus cereus 5
C diphtheriae 6
Listeria monocytogenes 8
Anaerobic rods 9
Clostridium botulinum 9
Clostridium difficle 11
Clostridium perfringens 13
Clostridium tetani 15
Arenaviridae 18
Lymphocytic choriomeningitis 18
Bunyaviruses 20
Hantavirus 21
Calcaviruses 23
Norvovirus 23
Central Nervous System
infections 25
Acanthamoeba 26
Naegleria floweri 27
Toxoplasma gondii 28
Haemophilus ducreyi 45
Haemophilus influenza 47
Pasteurella multocida 49
Comma-shaped rods 51
Campylobacter jejuni 51
Helicobacter pylori 53
Vibrio cholerae 55
Coronaviruses 57
Coronavirus (sars) 58
Cutaneous fungal
infections 59
Malassezia 59
Diplococci: aerobic 62
Moraxella catarrhalis 63
Neisseria gonorrhoeae 64
Neisseria meningitidis 66
Ecotparasites 68
Pediculosis corporis, capitis
and pubis (lice) 68
Sarcoptes scabiei (scabies) 70
Enterococcus 73
Enterococcus 73
Cestodes(tapeworms) 31
Diphyllobothrium latum 32
Echinococcus granulosis 33
Filaments 75
Actinomyces israelii 76
Nocardia 78
Chlamydia 35
Chlamydia species (pneumonia) 35
Chlamydia trachomatis 36
Filoviruses 81
Ebola virus 81
Coccobacilli: aerobes 38
Bordetella pertussis 38
Francisella tularensis 40
Coccobacilli:
facultative anaerobes 42
Brucella 42
Flaviviruses 84
Dengue virus 85
West nile virus 86
Yellow fever virus 88
Zika virus 89
Gastrointestinal infections 92
Cryptosporidium 93
AfraTafreeh.com exclusive
Table of contents
Entamoeba histolytica
(amoebiasis) 95
Giardia lamblia 96
General infections 98
Abscesses 98
Sepsis 100
Gram variable 102
Gardnerella vaginallis 102
Hematologic infections 104
Babesia 105
Plasmodium
species (malaria) 107
Hepadnaviridae 110
Hepatitis b virus 110
Hepatitis d virus 114
Herpesviruses 115
Cytomegalovirus 115
Epstein-barr virus (ebv) 116
Herpes simplex virus 117
Human herpesvirus 6 (roseola) 119
Human herpesvirus
8 (kaposi’s sarcoma) 120
Varicella zoster virus 122
Leishmania 125
Mycoplasma 127
Mycoplasma pneumonia 127
Nematodes (roundworms) 129
Ancylostoma duodenale &
necator americanus 130
Angiostrongylus
(eosinophilic meningitis) 132
Anisakis 133
Ascaris lumbricoides 134
Enterobius vermicularis
(pinworm) 137
Guinea worm (dracunculiasis) 138
Loa loa (eye worm) 139
Onchocerca volvulus
(river blindness) 140
Strongyloides stercoralis 142
Toxocara canis
(visceral larva migrans) 144
Trichinella spiralis 145
Trichuris trichiura
(whipworm) 147
Wuchereria bancrofti
(lymphatic filariasis) 148
Non tuberculous
mycobacterium 151
Mycobacterium leprae 151
Opportunisitc
fungal infections 153
Aspergillus fumigatus 153
Candida 156
Cryptococcus neoformans 158
Mucormycosis 160
Pneumocystis jirovecii 162
Sporothrix schenckii 163
Orthomyxoviruses 165
Influenza virus 166
Papillomavirus 168
Human papillomavirus 168
Paramyxoviruses 171
Human parainfluenza
viruses (hpiv) 171
Measles virus 172
Mumps virus 174
Respiratory syncytial
virus (rsv) 176
Parvovirus 177
Picornaviruses 180
Coxsackievirus 181
Hepatitis a virus 182
Poliomyelitis 183
Table of contents
Rhinovirus 185
Polyomavirus 187
Bk virus (hemorrhagic
cystitis) 187
JC virus (PML) 188
Poxviridae 190
Molluscum contagiosum 190
Reoviruses 192
Rotavirus 192
Retroviruses 194
Human immunodeficiency
virus 194
Human T-lymphotropic
virus 202
Rhabdoviruses 204
Rabies virus 204
Rickettsial diseases 207
Anaplasma 208
Coxiella burnetti (q fever) 210
Ehrlichia 211
Rickettsia rickettsii 213
Rickettsia typhi 215
Rods 216
Bacteroides fragillis 216
Bartonella henselae 217
Enterobacter 219
Escherichia coli 220
Klebsiella pneuomniae 222
Legionella pneumophilia 223
Nontyphoidal salmonella 224
Proteus mirabilis 225
Pseudomonas aeruginosa 226
Serratia marcescens 228
Shigella 229
Yersinia marcescens 230
Spirochetes 233
Borrelia burgdorferi
(lyme disease) 233
Borrelia species
(relapsing fever) 235
Leptospira 237
Treponema pallidum
(syphilis) 238
Staphylococcus 241
Staphylococcus aureus 241
Staphylococcus
epidermidis 243
Staphylococcus
saprophyticus 244
Streptococcus 245
Streptococcus
agalactiae (group b strep) 245
Streptococcus
pneumoniae 246
Streptococcus
pyogenes (group a strep) 248
Streptococcus
viridans 249
Systemic mycoses 251
Blastomyces spp. 251
Coccidioides spp. 253
Histoplasma capsulatum 255
Paracoccidioides brasiliensis 256
Togavirus 258
Eastern equine
encephalitis virus (eeev) 258
Rubella virus 259
Western equine
encephalitis virus (wee) 261
Trematodes (flatworms) 263
Clonorchis sinesis 264
Paragonimus westermani 265
Schistosomes 266
AfraTafreeh.com exclusive
Table of contents
Trichomona 269
Trichomonas vaginalis 269
Trypanosoma 271
Trypanosoma brucei 271
Trypanosoma cruzi 273
Tuberculosis 276
Mycobacterium
tuberculosis 276
NOTES
NOTES
ADENOVIRIDAE
ADENOVIRUS
osms.it/adenovirus
PATHOLOGY & CAUSES
▪ Seven subgroups, 52 serotypes
▪ Directly invades human epithelial cells
(several organs) → multiple cytopathic
effects, usually within 2–7 days postinfection
▪ Responsible for variety of upper, lower
respiratory tract; gastrointestinal;
genitourinary infections
▪ Common clinical syndromes
▫ Upper respiratory disease,
pharyngoconjunctival fever, coryza,
pneumonia, gastroenteritis, hepatitis,
hemorrhagic cystitis, interstitial
nephritis, epidemic keratoconjunctivitis
Genetic material
▪ Double-stranded DNA virus
Taxonomy
▪ Adenoviridae family
Morphology
▪ Non-enveloped icosahedral nucleocapsid
▪ Unique
▫ Fiber-like projections (hemagglutinins)
protrude from capsid’s 12 vertices
Transmission
▪ Aerosol droplets
▪ Fecal-oral route
▪ Neonates
▫ Infected cervical secretion exposure
▪ Solid organ transplant
▪ Fomite contact
▫ Survives on environmental surfaces
for long periods; inactivated by heat,
formaldehyde, bleach
RISK FACTORS
▪ Close living space (e.g. military barracks,
daycare centers)
▪ Summer camps (especially with swimming
pools/lakes)
▪ Healthcare facilities
▪ Immunocompromised status (especially
cell-mediated immunity deficiency)
COMPLICATIONS
▪ Secondary bacterial infection;
meningoencephalitis; myocarditis;
disseminated intravascular
coagulation; myositis, rhabdomyolysis;
hypogammaglobulinemia
▪ Disseminated adenovirus infection
▫ Affects multiple organs,
immunocompromised individuals
especially; high mortality rate
SIGNS & SYMPTOMS
▪ Varies with clinical syndrome
▪ Upper respiratory
▫ Sore throat, nasal congestion,
rhinorrhea, dry cough, hoarseness,
inflamed tonsils, cervical
lymphadenopathy; wheezes/rhonchi on
auscultation
1
Chapter 54 Adenoviridae
▪ Ocular
▫ Conjunctivitis, preauricular,
lymphadenopathy
▪ Gastrointestinal
▫ Nausea, vomiting, diarrhea
▪ Systemic
▫ Fever, malaise, headache, myalgia
DIAGNOSIS
▪ History, physical examination
▫ Characteristic findings
▫ Diagnostic test choice based on clinical
presentation
DIAGNOSTIC IMAGING
Chest X-ray
▪ May show diffuse bilateral pulmonary
mononuclear cell infiltration, hyaline
membranes, necrosis
LAB RESULTS
Microbe identification
▪ Viral culture (e.g. nasopharyngeal swabs,
blood, urine, sputum)
▪ Viral assay (e.g. adenovirus-specific
enzyme immunoassay (EIA)/
immunofluorescence assay)
▪ Nucleotide amplification test (NAT)
▪ Polymerase chain reaction (PCR)
Serology
▪ Antigen/complement fixation assays
▪ Hemagglutination inhibition/neutralizing
antibodies
▪ Restriction endonuclease (RE) analysis
Tissue biopsy
▪ Eosinophilic inclusions (early stage)
▪ Basophilic inclusion surrounded by clear
intranuclear inclusion
Electron microscopy
▪ Icosahedral virions forming intranuclear
paracrystalline aggregates
TREATMENT
▪ Usually self-limiting disease
MEDICATIONS
Pharmacotherapy
▪ Immunocompromised/severe infection
individuals
▫ Antivirals, immunoglobulin (IVIG)
Prevention
▪ Infection control measures
▪ Live oral enteric-coated vaccine used for
military recruits only
2
NOTES
NOTES
AEROBIC RODS
MICROBE OVERVIEW
▪ Gram-positive, rod-shaped, aerobes/
facultative anaerobes
BACILLUS ANTHRACIS (ANTHRAX)
osms.it/bacillus-anthracis
PATHOLOGY & CAUSES
▪ Etiologic agent of anthrax; nonmotile,
nonhemolytic; potential biological weapon
▪ Endospore-forming (centrally located)
▫ Highly adaptable to extreme
environmental conditions
▪ Surrounded by protein capsule (composed
of poly-D-gamma-glutamic acid)
▫ Prevents phagocytosis
▪ Transmission (4 Is)
▫ Ingestion
▫ Inhalation
▫ skin Invasion
▫ direct Injection
▪ Anthrax toxin composed of three proteins
▫ Protective antigen (PA): essential for
binding, entry to cell
▫ Lethal factor (LF): alteration of
signaling pathways → cell death; ↑
3
Chapter 55 Aerobic Rods
proinflammatory cytokines production
→ inflammation
▫ Edema factor (EF): ↑ cyclic adenosine
monophosphate (cAMP) → disordered
water, electrolyte balance → edema
RISK FACTORS
▪ Occupational
▫ People who work with animals/
animal products (e.g. veterinarians,
livestock producers, butchers); possible
bioterrorism (e.g. military personnel);
laboratory professionals
▪ Injecting drug users (e.g. heroin
contaminated with anthrax spores)
COMPLICATIONS
▪ Hemorrhagic meningitis, mediastinitis;
pleural effusion; pneumonia; shock
SIGNS & SYMPTOMS
▪ Respiratory infection
▫ Prodromal phase: flu-like symptoms
(e.g. fever, malaise, myalgia),
hemoptysis, dyspnea, nausea, chest
pain
▫ Fulminant phase: severe dyspnea,
hypoxemia, cyanosis, shock, coma
▪ Gastrointestinal (GI) infection
▫ Severe abdominal pain; nausea;
vomiting; ascites; ulcerations → GI
hemorrhage
▪ Cutaneous infection
▫ Painless, pruritic papule → enlarges,
forms central black-colored necrotic
ulceration → black eschar
▫ Surrounding edema
▫ Regional lymphadenopathy,
lymphadenitis
Figure 55.1 A lesion on the neck caused by
Bacillus anthracis.
DIAGNOSIS
LAB RESULTS
▪ Identify microbe
▫ Gram stain, culture, direct fluorescent
antibody (DFA), polymerase chain
reaction (PCR)
OTHER DIAGNOSTICS
▪ History, physical examination
TREATMENT
MEDICATIONS
▪ Antibiotics
OTHER INTERVENTIONS
▪ Vaccine (people at high risk of exposure,
post-exposure prophylaxis)
4
AfraTafreeh.com exclusive
BACILLUS CEREUS
(FOOD POISONING)
osms.it/bacillus-cereus
PATHOLOGY & CAUSES
▪ Common foodborne pathogen; also
associated with contaminated medical
equipment (e.g. ventilators, dialysis
machines), opportunistic infections
▪ Endospore-forming (centrally located)
▫ Highly adaptable to extreme
environmental conditions
▪ Motile, catalase positive, beta-hemolytic
▪ Can be transient component of GI
microflora
Pathogenesis of food poisoning
▪ Production of enterotoxins
▫ Diarrheal toxin (thermolabile) → ↑ cAMP
→ disordered function of ion pumps
→ ↑ efflux of ions, water from infected
enterocytes → diarrhea
▫ Emetic toxin, cereulide (thermostable)
→ ↑ afferent vagus nerve stimulation →
nausea, vomiting
▪ Two types of food poisoning
▫ Diarrheal syndrome (meat, vegetables,
sauces) → toxicoinfection → ingestion
of bacteria, production of toxins in
digestive tract
▫ Emetic syndrome (rice) → alimentary
intoxication → direct ingestion of toxin
SIGNS & SYMPTOMS
▪ Abdominal cramps, diarrhea, nausea,
vomiting
DIAGNOSIS
LAB RESULTS
▪
▪
▪
▪
Stool/contaminated food sample
Gram stain
Culture (blood agar)
PCR
TREATMENT
OTHER INTERVENTIONS
▪ Intravenous (IV) fluid, electrolyte
replacement
RISK FACTORS
▪ Consumption of improperly cooked food
5
Chapter 55 Aerobic Rods
CORYNEBACTERIUM DIPHTHERIAE
(DIPHTHERIA)
osms.it/corynebacterium-diphtheriae
PATHOLOGY & CAUSES
▪ Infectious agent of diphtheria
▪ Rods with widening at polar regions
forming club-like shape
▪ Characteristic “Chinese-letter” arrangement
▪ Nonmotile, non-spore-forming
▪ Stain
▫ Albert’s/Ponder’s; metachromatic
granules (e.g. Babes–Ernst, volutin)
▪ Culture
▫ Löffler's medium
▪ Differentiation
▫ Hoyle’s tellurite agar
▪ Types of infection
▫ Respiratory (pharyngeal), cutaneous
RISK FACTORS
▪ Absent/incomplete immunization
▪ Immunocompromised individuals
▪ Citizens, migrants, returning travellers from
endemic areas (e.g. African, Asian, South
American countries)
COMPLICATIONS
▪ Myocarditis, nerve damage (e.g.
demyelinating polyneuropathy, paralysis),
renal failure, suffocation (due to
pseudomembrane aspiration)
Pathophysiology
▪ Diphtheria toxin → composed of two
subunits
▫ A: active, catalytic
▫ B: binding; composed of R (receptor), T
(translocation) domains
▪ Bacteria binds to host cell using R domain
→ endocytosis → acidification inside
endosome → T domain transfers to
endosomal membrane → translocation of A
subunit to cytosol
▪ Subunit A inactivates elongation factor EF2
→ inhibition of protein synthesis → cell
death
▪ Toxin expression regulated by level of iron
▫ ↑ Fe → ↓ production of toxin
▫ ↓ Fe → ↑ production of toxin
Figure 55.2 A pharyngeal pseudomembrane
in a child with diphtheria.
6
SIGNS & SYMPTOMS
▪ Respiratory infection
▫ Sore throat; malaise; low-grade fever;
dysphagia; thick, grey, isolated necrotic
tissue → pseudomembrane; massive
swelling of tonsils, cervical lymph nodes
→ “bull neck”; stridor
▪ Cutaneous infection
▫ Lesions, pain, rash, tenderness,
erythema, ulceration
DIAGNOSIS
LAB RESULTS
▪
▪
▪
▪
Gram stain
Culture
Elek test (differentiation of toxigenic strains)
PCR
OTHER DIAGNOSTICS
▪ History, physical examination
Figure 55.3 An ulcerating skin lesion on the
leg caused by cutaneous diphtheria.
TREATMENT
MEDICATIONS
▪ Diphtheria antitoxin
▪ Antibiotic
OTHER INTERVENTIONS
▪ Prophylaxis
▫ Young children: diphtheria-tetanusacellular pertussis (DTaP) vaccine
▫ Adolescents/adults: tetanus-diphtheria
(Td)/tetanus-diphtheria-pertussis (Tdap)
vaccine
7
Chapter 55 Aerobic Rods
LISTERIA MONOCYTOGENES
osms.it/listeria-monocytogenes
PATHOLOGY & CAUSES
▪ Facultative intracellular bacteria, anaerobe;
beta-hemolytic
▪ Capable of growing at refrigeration
temperatures (0–4°C/32–39.2°F)
▪ Motile
▫ ≤ 30°C/32°F (flagella); body temperature
(comet tail structures, polymerized host
cells actin)
▪ Foodborne pathogen, common cause of
bacterial neonatal meningitis
Pathophysiology
▪ Listeria enters host cell via zipper
mechanism → bacterial protein internalin
binds onto cell membrane protein cadherin
→ releases listeriolysin O → disruption of
vacuolar membrane → invasion of cytosol
→ actin assembling-inducing protein →
polymerisation of cytoskeleton → bacteria
gains motility → rapid movement through
cytosol, between cells
RISK FACTORS
▪ Mild febrile gastroenteritis
▫ Immunocompetent individuals, ingestion
of contaminated food (e.g. raw meat,
unpasteurized dairy, seafood)
▪ Cutaneous infection
▫ Direct inoculation of skin (e.g.
veterinarians, farmers handling infected
animals)
▪ Invasive listeriosis
▫ Immunocompromised individuals, age
(neonates, elderly), pregnancy
COMPLICATIONS
▪ Immunodeficient, elderly individuals
▫ Sepsis, meningitis, encephalitis,
pneumonia, corneal ulcer
▪ Pregnant individuals
▫ Neonatal meningitis, granulomatosis
infantiseptica, miscarriage,
stillbirth, premature delivery with
chorioamnionitis
SIGNS & SYMPTOMS
▪ Previously healthy individuals
▫ Fever, headache, diarrhea, vomiting,
nausea, pustular skin lesions
▪ Individuals with weakened immune system
▫ Stiff neck, confusion, convulsions, loss of
balance, cranial nerve palsies
▪ Pregnant individuals
▫ Nonspecific flu-like illness
▪ Newborns
▫ Low birth weight, irritability, fever,
poor feeding, circulatory/respiratory
insufficiency, pyogranulomatous lesions
DIAGNOSIS
LAB RESULTS
▪ Culture
▫ Blood, cerebrospinal fluid (CSF); cervix,
amniotic fluid in pregnant individuals;
meconium, gastric aspirate, infected
tissues (e.g. skin granulomatous lesions)
in newborns
OTHER DIAGNOSTICS
▪ History, physical examination
TREATMENT
MEDICATIONS
▪ Antibiotics (e.g. ampicillin, penicillin G,
gentamicin)
8
NOTES
NOTES
ANAEROBIC RODS
MICROBE OVERVIEW
▪ Rod-shaped (bacilli) bacteria, grampositive, strict anaerobes
CLOSTRIDIUM BOTULINUM
(BOTULISM)
osms.it/clostridium-botulinum
PATHOLOGY & CAUSES
▪ Ubiquitous presence (esp. soil, water);
spore-forming; catalase negative,
superoxide dismutase positive, subterminal
endospore
▪ Fermentation
▫ Carbohydrates
▪ Obligate anaerobe, can tolerate small
amounts of oxygen due to superoxide
dismutase
▪ Potential bioterrorism weapon
▪ Absorption of toxin into bloodstream →
spreads to nervous system → binds to
presynaptic receptors → endocytosis →
cleavage of SNAP-25 protein → lack of
acetylcholine with impaired transduction
Virulence factors
▪ Seven distinct types of neurotoxins (A–G)
▫ Type A: most potent
9
Chapter 56 Anaerobic Rods
▪ Heat-resistant
▫ Toxins enduring temperature of
100ºC/212°F for several hours
▪ H-antigen from flagelle
Culture
▪ Chopped meat, glucose, starch medium
for isolation; egg yolk agar incubation in
anaerobic conditions
Disease
▪ Causes disease characterized by muscle
weakness, nervous system impairment
▪ Infant botulism
▫ Spores ingestion → germination in
gastrointestinal (GI) tract → toxin
produced in vivo
▪ Foodborne
▫ Ingestion of botulinum toxincontaminated food
▫ Average incubation period is 12–36
hours
▪ Wound botulism
▫ Wound infection with spores → toxin
produced in vivo
▫ Average incubation period is 10 days
▪ Adult intestinal toxemia botulism
▫ Colonization of intestines with toxins
production
▪ Iatrogenic botulism
▫ Complication of therapeutic use of
botulinum neurotoxins
▪ Enteric infectious botulism
▫ C. botulinum colonizes adult GI tract
RISK FACTORS
▪ Infant botulism
▫ Honey consumption in first year of
life; ingestion of dust/soil containing C.
botulinum spores
▪ Foodborne botulism
▫ Home-canned, improperly preserved
food; smoked fish
▪ Wound botulism
▫ IV/subcutaneous drug usage; crush
injuries
▪ Enteric infectious botulism
▫ Achlorhydria/other GI diseases →
colonization
COMPLICATIONS
▪ Sudden infant death syndrome (SIDS),
seizures, ileus, death
SIGNS & SYMPTOMS
▪ General symptoms precede muscle
weakness
▫ Abdominal pain, nausea, vomiting, lack
of fever (wound botulism only type with
fever)
▪ Cranial nerve impairment
▫ Dilated pupil, ptosis; double vision
(due to disconjugate eye movement);
ophthalmoplegia; dry mouth, difficulty
swallowing; loss of facial expressions
▪ Progressive symmetrical muscle weakness
descending from head
▫ Hypotonia, hyporeflexia; floppiness in
infants
▫ Respiratory muscles: breathing
difficulties
▪ Hyperactivation of autonomic system
▫ ↓ salivation, lacrimation, orthostatic
hypotension, obstipation, urine retention
DIAGNOSIS
LAB RESULTS
▪ Toxin detection/bacteria isolation
Enzyme-linked immunosorbent assay (ELISA), mass spectroscopy, polymerase chain
reaction (PCR)
▪ Isolation of C. botulinum from feces,
vomitus, food
OTHER DIAGNOSTICS
▪ History, physical examination
Electromyogram (EMG)
▪ Short-lasting motor unit potentials with
small amplitude
10
TREATMENT
MEDICATIONS
▪ IV botulinum immunoglobulin/heptavalent
botulinum antitoxin
▫ Manage infection
▪ Antibiotics
▫ Manage secondary infection
▪ Cholinesterase inhibitors
OTHER INTERVENTIONS
▪ Manage infection
▫ Debridement, irrigation of wound
▫ Colon cleansing (enema, cathartics)
▪ Nasogastric tube (feeding), Foley catheter
(urinary retention), intubation/mechanical
ventilation
Prevention
▪ Pentavalent-botulinum-toxoid (PBT)
vaccine
▫ Five dose vaccination with booster dose
once per year
CLOSTRIDIUM DIFFICILE
(PSEUDOMEMBRANOUS COLITIS)
osms.it/clostridium-difficile
PATHOLOGY & CAUSES
▪ Ubiquitously present, can be part of
“normal” flora; subterminal endospore;
motile
▪ Intestinal microbiota disturbance →
infection, colonization of gut → produces
A, B toxins → binds to receptors on
intestinal wall → internalization → fusion
with lysosome → toxins exit endosome →
damage of cytoskeleton → cell apoptosis →
inflammatory response with accumulation
of inflammatory cells, fibrin, dead cells
→ formation of membrane-like structure
(pseudomembrane)
Virulence factors
▪ Enterotoxin A (TcdA), cytotoxin B (TcdB),
H-antigen from flagelle
Culture
▪ Agar with cycloserine, cefoxitin, fructose
▪ C. difficile-associated colitis combined with
formation of pseudomembranous plaques
Transmission
▪ Fecal-oral route by ingestion of spores
RISK FACTORS
▪ Antibiotic exposure
▫ Disturbance of intestinal microbiota
▪ Previous hospitalization
▫ ↑ risk for infection
▪ Children with neutropenia
▫ More prone to common bacterial
infections requiring antibiotics →
disturbance of intestinal microbiota
▪ Gastric acid suppression, > 65 years,
comorbidities
COMPLICATIONS
▪ Multiple relapses, dehydration (due to
excessive diarrhea)
GI complications
▪ Toxic megacolon (due to inflammation
damaging muscularis propria, underlying
neurons); ileus; colon perforation;
intussusception; pneumatosis; ascites;
sepsis
Extraintestinal complications
▪ Splenic abscess; osteomyelitis
11
Chapter 56 Anaerobic Rods
SIGNS & SYMPTOMS
▪ Watery diarrhea (most common) with
mucus/blood
▪ Abdominal distension, cramps
▪ Malaise
▪ Fever
DIAGNOSIS
DIAGNOSTIC IMAGING
Sigmoidoscopy/colonoscopy
▪ Visualization of plaques
LAB RESULTS
SURGERY
▪ Colectomy
▫ In persons with acute abdomen,
refractory colitis, fulminant colitis
OTHER INTERVENTIONS
▪ End previous antibiotic use
▪ Second relapse: pulse therapy
▪ Recurrent: biological therapies
▫ Fecal microbiota transplant:
transplantation of microbiota from
healthy individual
▪ Electrolytes, fluids replacement; appropriate
nutrition
▪ Prevention of hospital-acquired infection
▫ Infection control protocol, antibiotic
stewardship
WBCs
▪ ↑ white blood cells
Stool analysis
▪ Presence of blood/mucus
▪ Stool culture
▪ Enzyme immunoassay test
▫ Detection of glutamate dehydrogenase
antigen
▪ Toxin detection
▫ Enzyme immunoassay, real-time PCR
▪ Cell culture cytotoxicity assay
▪ Biomarkers
▫ Differentiation between colonization,
actual disease; ↑ fecal cytokines, CXCL5,
phosphorylated p38
Figure 56.1 The gross pathological
appearance of the colonic mucosa in
psuedomembranous colitis.
TREATMENT
MEDICATIONS
▪ Mild disease: oral metronidazole
▪ Severe disease: oral vancomycin
▪ Complications: combination of oral
vancomycin, IV metronidazole
▪ Second relapse: oral vancomycin tapered
▪ Recurrent: probiotics
12
CLOSTRIDIUM PERFRINGENS
osms.it/clostridium-perfringens
PATHOLOGY & CAUSES
▪ Ubiquitous in nature, also part of human
microbiota; subterminal endospore;
nonmotile
Virulence factors
▪ Divided into subtypes A–E
▪ Produces 12 toxins
▪ Alpha
▫ Enzyme lecithinase splits lecithin → ↑
vascular permeability → cell destruction
▫ Responsible for gas gangrene,
hemolysis
▪ Beta
▫ Formation of selective pores → ↑
permeability with cell destruction
▫ Responsible for enteritis necroticans;
deactivated by trypsin
▪ Epsilon
▫ Pores form on cells → destruction
▪ Iota
▫ AB toxin: enzyme (A), binding (B)
domain
▫ Destruction of cells through affection of
cytoskeleton
Culture
▪ Growth on tryptose sulfite cycloserine agar
Clostridial gas gangrene
▪ Saprophytic anaerobic bacteria → clostridial
gas gangrene due to tissue infection/food
poisoning through ingestion
▪ Disease characterized by necrosis,
gangrene due to infection of skin, deep
tissues
▪ Bacteria produces gas → characteristic
crepitation sound during palpitation
▪ Divided into
▫ Traumatic: wound → spore inoculation
→ bacteria growth in appropriate
anaerobic conditions → production
of toxins → destruction of fibroblasts,
blood cells, muscle cells → necrosis of
muscles, subcutaneous fat with blood
vessels thrombosis
▫ Postoperative: after interventions on
intestinal system
▫ Spontaneous: due to immune system
weakness/intestinal diseases
Food poisoning
▪ Begins 6–24 hours after ingestion
▪ Infection of food with spores → food
standing temperature 30–50ºC/86–122°F
→ development of vegetative form
→ ingestion of infected food → toxin
production → ↓ glucose absorption, ↑
water, sodium, chloride secretion →
damage of intestinal epithelium
Enteritis necroticans
▪ Inflammation of jejunum, ileum; type of food
poisoning in persons who lack trypsin
RISK FACTORS
▪ Gas gangrene: begins 1–4 days after
infection
▫ Traumatic: injury during war/natural
disaster; car crash
▫ Postoperative: GI/biliary surgery; septic
abortion
▫ Spontaneous: colorectal
adenocarcinoma; neutropenic states
(e.g. AIDS, chemotherapy); intestinal
diseases
▪ Food poisoning: improper food-handling
▪ Enteritis necroticans: improper nutrition
(e.g. too many sweet potatoes); trypsin
inhibitors → ascariasis
COMPLICATIONS
▪ Disseminated intravascular coagulation;
hemodynamic shock, hypotension, renal
failure; systemic hemolysis; peritonitis;
sepsis; death
13
Chapter 56 Anaerobic Rods
DIAGNOSIS
SIGNS & SYMPTOMS
Gas gangrene
▪ Two types
▫ Cellulitis: infection of necrotic skin;
crepitation sounds
▫ Clostridial myonecrosis: infection,
destruction of muscles, adjacent tissue
▪ Paleness, serosanguineous exudate, pain,
swelling, tenderness at injury site → color
changes to bronze → purple bullae with
green/black discoloration due to necrosis
▪ Characteristic sweet odor
▪ ↑ heart rate with low-grade fever
Food poisoning
▪ If only toxins ingested
▫ Asymptomatic/diarrhea
▪ If vegetative form ingested
▫ Watery diarrhea without blood/mucus;
abdominal cramps, last > 24 hours
Enteritis necroticans
▪ Abdominal cramps; diarrhea; vomiting;
meteorism (excessive gas production in
intestines); blood in stool; fever
DIAGNOSTIC IMAGING
X-ray
▪ Gas gangrene: detection of gas in softtissue
▪ Enteritis necroticans: small bowel dilatation
with presence of gas
LAB RESULTS
▪ Gas gangrene: molecular methods for
detection of alpha toxins; ELISA, PCR
▪ Food poisoning: detection of toxins in
feces; PCR
Biopsy
▪ Gas gangrene: necrosis of myocytes,
connective tissue with small number of
neutrophils
OTHER DIAGNOSTICS
▪ Gas gangrene: physical examination;
characteristic discoloration, odor, crepitation
sound
TREATMENT
MEDICATIONS
▪ Gas gangrene: antimicrobial therapy
(combination of penicillin G, clindamycin)
▪ Enteritis necroticans: penicillin G/
metronidazole; destruction of bacteria
SURGERY
Figure 56.2 An individual with gas gangrene
of the right leg. The causative agent is
Clostridium perfringens.
▪ Gas gangrene: removal of dead tissue;
hyperbaric oxygen (augments neutrophil,
↓ clostridial exotoxin, spore production);
amputation
▪ Enteritis necroticans: resection of necrotic
intestinal parts
OTHER INTERVENTIONS
▪ Food poisoning: oral/IV rehydration
14
AfraTafreeh.com exclusive
CLOSTRIDIUM TETANI (TETANUS)
osms.it/clostridium-tetani
PATHOLOGY & CAUSES
▪ Ubiquitously present in soil; resistant to
chemical/heat disinfection; terminally
present endospore; motile
▪ Anaerobic rod causes nervous system
disorder (i.e. tetanus)
▪ Injury → infection with spores →
development of vegetative form in
appropriate anaerobic conditions →
production of exotoxins (tetanospasmin) →
blood/lymphatic transmission → spread to
neurons through neuromuscular junction
→ retrograde transport to spinal cord
→ endocytosis into inhibitory Renshaw
cell interneurons → exotoxins protease
activity cleaves SNARE proteins → block of
glycine, gamma-aminobutyric acid (GABA)
neurotransmitter-filled vesicles release at
neuromuscular junction → overactivation of
muscles → muscle spasms
Virulence factors
▪ Two toxins: tetanolysin (relatively unknown
function); tetanospasmin (responsible for
clinical presentation of tetanus)
▪ H-antigen from flagelle
Culture
▪ Oxygen-reduced blood agar with anaerobic
incubation
TYPES
▪ Four types of tetanus
▫ Generalized: affects whole musculature,
from head downwards
▫ Localized: affects only area near injury
▫ Cephalic: cranial nerves affected due to
head/neck injury
▫ Neonatal: tetanus occurring in neonates
via infection of umbilical stump
RISK FACTORS
▪ Deep puncture wounds with/without
splinters, crush injuries, middle ear
infections, frostbites, burns
▪ IV/subcutaneous drug abuse
▪ Infected diabetic wounds
▪ Septic abortion
▪ Umbilical stump infection via contaminated
instruments, hands, cultural practices (e.g.
application of cow dung, ghee)
▪ Lack of vaccination/immunization
COMPLICATIONS
▪ Cardiac arrest
▫ Due to ↑ catecholamine levels,
brainstem damage
▪ Palsies of phrenic, laryngeal nerves due to
toxic effect
▪ Respiratory muscles
▫ Apnea
▪ Vertebral fractures/intramuscular bleeding
(due to opisthotonus)
SIGNS & SYMPTOMS
Generalized
▪ Masseter muscle spasm →
trismus/“lockjaw” → severe generalized
muscle contractions
▫ Risus sardonicus: characteristic
sarcastic-like facial expression due to
facial muscles contraction
▫ Opisthotonus: arched back due to
contraction of back muscles
▪ Abdominal respiratory muscles,
diaphragm: breathing arrest
Localized
▪ Weakness, pain of muscles/extremites in
proximity of injury
▪ Stiffness of affected muscles occurring in
following days, lasting up to few months
▪ May progress into generalized form
15
Chapter 56 Anaerobic Rods
Cephalic
▪ Palsy of facial nerve
▪ Difficulty swallowing
▪ Weakness of extraocular muscles
Neonatal
▪ Sucking difficulty in first days of life
▪ Generalization, opisthotonus may occur
Sympathetic overactivity
▪ ↑ heart rate, arrhythmias
▪ Agitation, diaphoresis
▪ ↑/↓ blood pressure
▪ Fever
▫ Due to overactivity/superinfection
TREATMENT
MEDICATIONS
▪ Human tetanus immunoglobulin (TIG)/
intravenous immunoglobulins (IVIG)
▫ Neutralization of toxin
▪ Antibiotics
▫ Penicillin, cephalosporins, tetracyclines,
metronidazole, vancomycin
▪ Benzodiazepines
▫ Muscle relaxation, sedation
▪ Muscle relaxants
▫ Pancuronium/vecuronium
▪ Labetalol
▫ Autonomic hyperactivity management
OTHER INTERVENTIONS
Figure 56.3 This body posture is known
as opisthotonus and is caused by the
Clostridium tetani toxin.
▪ Active immunization with tetanus/
diphtheria toxoid (Td)
▪ Respiratory support if needed
▪ Prevention
▫ Primary: vaccination at two months of
life, with booster doses at 4, 6, 12–18
months, 4–6, 11–12 years (booster
doses every ten years later); passive
immunization in immunocompromised
▫ Secondary: vaccination, antitoxin after
injury
▫ Tertiary: vaccination, antitoxin after
tetanus presentation
DIAGNOSIS
OTHER DIAGNOSTICS
▪ History, physical investigation
▪ Spatula test
▫ Touching oropharynx with spatula
causes reflex biting due to masseter
spasm
EMG
▪ Loss/shortening of silent interval between
action potentials with continuous discharge
from motor units
16
17
NOTES
NOTES
ARENAVIRIDAE
MICROBE OVERVIEW
Taxonomy
▪ Zoonotic virus family
▫ Associated with hemorrhagic fevers,
meningoencephalitis
▪ Comprises > 20 viruses
▪ Groups
▫ Old World viruses (e.g. lymphocytic
choriomeningitis virus, Lassa, Lujo)
▫ New World viruses (e.g. Junin, Machupo)
Morphology
▪ Single-stranded RNA virus
▪ Ribosomes produce sand-like granular
appearance under electron microscopy
▫ Latin: arena = sand
▪ Shape varies: pleomorphic–spherical
▪ Enveloped in lipid membrane with
glycoprotein spikes
▫ Lipids in membrane → inactivation
susceptibility by organic solvents/
detergents
▫ Also inactivated by temperatures >
55°C/131°F, ↑ ↓ pH, UV light, gamma
irradiation
Transmission
▪ Reservoir
▫ Rodents shed virus in urine, feces,
saliva, nasal secretion
▪ Transmission
▫ Ingestion, direct contact, aerosolizedparticle inhalation → entry into cell by
GP1 glycoprotein attachment to cellular
receptors → systemic dissemination
LYMPHOCYTIC CHORIOMENINGITIS
VIRUS (LCMV)
osms.it/LCMV
PATHOLOGY & CAUSES
▪ Arenavirus
▫ Causes febrile illness with central
nervous system (CNS) involvement,
congenital infection
▪ Reservoir
▫ Mice (M. musculus/M. domesticus);
occasional human disease outbreak
related to pet hamster/guinea pig
▪ Transmission
▫ Ingestion, aerosolized-particle
inhalation, transplacentally (infected
mother → fetus)
▪ Post-transmission → 7–14 days incubation
→ replication in lungs → hematogenous
dissemination → strong neurotropism
(infects meninges, choroid plexus,
ventricular ependyma)
RISK FACTORS
▪ Infected rodent contact; dwelling/working in
rodent habitats
▪ ↑ risk in substandard housing (e.g.
mobile homes, inner-city housing), barns/
18
outbuildings
▪ Peak incidence: autumn, winter
COMPLICATIONS
▪ Encephalitis, aseptic meningitis
▪ Transplacental infection: spontaneous
abortion, neonatal neurologic deficits,
chorioretinitis, seizures, periventricular
calcifications, hydrocephalus, microcephaly
▪ Rarely: orchitis, parotitis, myocarditis
SIGNS & SYMPTOMS
▪ May be asymptomatic/mild flu-like
symptoms
▪ Prodrome: headache (retro-orbital), nausea,
vomiting
▪ Meningitis: fever, rigors, malaise, myalgia,
arthralgia anorexia, photophobia
▪ Encephalitis: focal seizures, cranial nerve
palsies, papilledema
DIAGNOSIS
LAB RESULTS
▪ Cerebrospinal fluid (CSF)
▫ ↑ lymphocytes, ↑ protein, ↓ glucose,
negative Gram stain
Microbe identification
▪ Viral culture, polymerase chain reaction
(PCR)
Serology
▪ ↑ antibody titer (IgM, IgG)
TREATMENT
OTHER INTERVENTIONS
Prevention
▪ Rodent control
19
NOTES
NOTES
BUNYAVIRUSES
MICROBE OVERVIEW
▪ Bunyaviruses: RNA virus order
▪ AKA Bunyavirales
Genetic material
▪ Negative-sense, single-stranded RNA
▪ Tripartite genome
▫ Large (L) segment (encodes RNApolymerase); medium (M) segment
(encodes surface glycoproteins); small
(S) segment (encodes nucleocapsid
proteins)
Morphology
▪ Enveloped (outer lipid membrane)
▪ Spherical
▪ Size: 90–100nm
Replication
▪ Host cytoplasm replication
Transmission
▪ Vector-borne, person-to-person, contact
with reservoir (species-dependent)
▪ Reservoirs: arthropods, mammals
(especially rodents
Figure 58.1 Families of the Bunyavirus order and associated clinical syndromes.
20
HANTAVIRUS
osms.it/hantavirus
PATHOLOGY & CAUSES
▪ Single-stranded RNA virus genus →
causes severe renal/pulmonary disease
▪ AKA Orthohantavirus
▪ Hantaviridae family
▪ Reservoir
▫ Rodents (mice, voles, shrews, rats)
▪ Incubation period
▫ 9–33 days
▪ Tissue tropism
▫ Lymphoid organ, heart, lung, kidney
vascular endothelium
▪ Hantavirus inhalation → lung phagocytosis
→ transport to lymph nodes → hantavirus
enters epithelial cells using beta-3 integrins
→ ↑ vascular permeability → dissemination
▪ Immune response
▫ CD4+/CD8+ cytotoxic T cells, dendritic
cells
▪ High-prevalence regions
▫ China, Russia, Europe
Associated clinical syndromes
▪ Hemorrhagic fever + renal syndrome (“Old
World” AKA Asia, Europe)
▪ ↑ vascular permeability → ↓ blood pressure
→ kidney endothelial dysfunction
▪ Hantavirus cardiopulmonary/pulmonary
syndrome (“New World” AKA North, South
America)
▪ ↑ vascular permeability → non-cardiogenic
pulmonary edema
CAUSES
▪ Rodents (urine/feces/saliva particle
inhalation); person-to-person (rare)
RISK FACTORS
▪
▪
▪
▪
Rural indoor spaces (e.g. barns)
Wild rodent exposure
Smoking → Puumala virus infection risk
↑ risk among biological males
COMPLICATIONS
▪ Cardiogenic shock, pulmonary edema,
arrhythmia, renal insufficiency, acute kidney
injury (AKI), coagulopathy, disseminated
intravascular dissemination (DIC),
hemorrhage; high mortality rate
21
Chapter 58 Bunyaviruses
SIGNS & SYMPTOMS
Urinalysis
▪ Renal syndrome: proteinuria, hematuria
Pulmonary syndrome
▪ Systemic
▫ Fever, chills, myalgia, headaches,
weakness
▪ Nausea, vomiting, diarrhea, abdominal pain,
cough, oliguria
▪ Sometimes conjunctivitis, flushing,
petechiae
Tissue biopsy
▪ Kidneys: edema, perirenal hemorrhage,
tubular destruction
▪ Lungs: edema, mononuclear cell infiltrates,
tracheal/pleural fluid, hyaline deposits
▪ Lymphoid organs: mononuclear cell
infiltrates
Hemorrhagic fever + renal syndrome
▪ Fever, hypotension, malaise, headache,
diffuse hemorrhage (e.g. petechiae, melena,
ecchymoses), abdominal/loin pain, nausea/
vomiting, oliguria
TREATMENT
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Lungs: bilateral pulmonary infiltrates,
pleural effusion
Abdominal ultrasound
▪ Kidneys: ↑ size, perirenal fluid
LAB RESULTS
MEDICATIONS
▪ Renal syndrome antivirals
▫ Nucleoside analogue
OTHER INTERVENTIONS
▪ ICU monitoring, mechanical ventilation
▪ Severe cases
▫ Extracorporeal membrane oxygenation
(ECMO), dialysis, platelet transfusion
Prevention
▪ Potential rodent nesting site removal (e.g.
debris, garbage)
▪ Closed spaces ventilation
▪ Avoid rodent contact
▪ Reverse-transcriptase polymerase chain
reaction (RT-PCR)
Tests
▪ Diagnostic triad: thrombocytopenia,
↑ immunoblasts (> 10%), left-shifted
granulocytic series
▪ ↑ lactate dehydrogenase (LDH), serum
lactate
▪ ↑ liver enzymes
▪ ↑ C-reactive protein
▪ Severe
▫ ↑ hemoglobin, hematocrit; ↓ albumin;
altered coagulation tests
▪ Renal syndrome
▫ ↑ serum creatinine, ↓ glomerular filtration
rate (GFR)
Serologic tests
▪ IgM/IgG detection through ELISA,
strip immunoblot, Western blot,
immunofluorescence
22
NOTES
NOTES
CALICVIRUSES
MICROBE OVERVIEW
Genetic material
▪ Single-stranded RNA viruses
Morphology
▪ Small, icosahedral, non-enveloped
Taxonomy
▪ Genera: Norovirus, Sapovirus, Lagovirus,
Nebovirus, Vesivirus
Transmission
▪ Ingestion → small intestine → villi infection
→ malabsorption → osmotic diarrhea
NOROVIRUS
osms.it/norovirus
PATHOLOGY & CAUSES
▪ Virus known as viral gastroenteritis agent
▫ AKA Norwalk virus
▪ Classified into seven genogroups
▫ I, II, IV → cause human disease
Transmission
▪ Fecal-oral
▫ Infected individual: virus in stool < four
weeks
▫ Immunocompetent individual: peak
concentration 2–5 days post-infection
▪ Direct person-to-person
▪ Contaminated food, water, fomite
▫ Contaminated water in leafy vegetables,
oysters, raspberries
▫ Prepared food → contaminated at point
of service (e.g. salads, sandwiches)
▪ Droplet spread from vomitus
Pathogenesis
▪ Ingestion → small intestine villi infection →
disruption of epithelium, anion transport
system → malabsorption of D-xylose, fat →
osmotic diarrhea
Small intestine histopathology
▪ Villous shortening, epithelial vacuolization,
crypt hypertrophy, microvilli brush
border loss, intercellular space widening,
lymphocytic proliferation of lamina propria
RISK FACTORS
▪ Blood group antigens
▫ Viral binding preference for A, B
antigens genogroup-determinant
▪ Close-quarter residence (e.g. nursing
homes, cruise ships)
COMPLICATIONS
▪ Severe dehydration, malnutrition, persistent
disease (immunosuppressed individuals)
SIGNS & SYMPTOMS
▪ 12–48 hour incubation period
▪ Acute onset vomiting
▫ Nonbloody, nonbilious
▪ Watery diarrhea
▫ Nonbloody
▪ Fever, abdominal pain, myalgia, malaise
23
Chapter 59 Calicviruses
DIAGNOSIS
▪ History
▫ Pathogen contact, contaminated food
source
LAB RESULTS
▪ Reverse transcriptase polymerase chain
reaction (RT-PCR)
▫ Stool
OTHER INTERVENTIONS
▪ Fluid maintenance, repletion
▫ From oral intake → IV fluids
▪ Electrolyte balance
Prevention
▪ Hand hygiene
▪ Environmental fecal contact
decontamination
▪ Limited pathogen contact
TREATMENT
▪ No cure
MEDICATIONS
▪ Antimotility agents
▪ Antiemetics
▫ Reserved for individuals unable to take
oral hydration
24
AfraTafreeh.com exclusive
NOTES
NOTES
CENTRAL NERVOUS SYSTEM
INFECTIONS
MICROBE OVERVIEW
PATHOLOGY & CAUSES
▪ Rare infections of central nervous system
(CNS) by ameba, parasites
RISK FACTORS
▪ Immunosuppression (Acanthamoeba,
Toxoplasmosis gondii), immersion in
infested water (Naegleria fowleri)
SIGNS & SYMPTOMS
▪ Fever, headache, seizures, focal
neurological signs, mental status change
DIAGNOSIS
LAB RESULTS
▪ Presence of infectious agent via
microscopy, culture, polymerase chain
reaction (PCR), presence of specific
antibodies
Granulomatous amebic encephalitis
▫ Brain biopsy: trophozoites in
perivascular space and thick walled
cysts, PCR/DNA probes may show
Acanthamoeba
Primary amoebic meningoencephalitis
▪ Lumbar puncture
▫ CSF microscopy: motile amebae/
fluorescent antibody staining
▫ CSF PCR: Naegleria fowleri DNA
▫ CSF culture: Naegleria fowleri can be
grown on nonnutrient agar coated with
Escherichia coli
Toxoplasmosis
▪ PCR (blood, CSF): Toxoplasma gondii DNA
(inactive cysts may evade detection)
▪ Antibody titres
▫ IgG: evidence of current/previous
infection
▫ IgM: occur in weeks after initial infection
▫ Antibody avidity testing: affinity for
antigen increases with duration of
infection
▪ Sabin–Feldman dye test: high titers →
acute infection
▪ Tissue biopsy: tachyzoites in tissues/
smears
TREATMENT
MEDICATIONS
▪ Antifungal, antiparasitic agents
25
ACANTHAMOEBA
osmosis.org/learn/acanthamoeba
PATHOLOGY & CAUSES
Genus of amebae
▪ Single-celled eukaryotes
▪ Environmentally ubiquitous organisms
▫ Acanthamoeba spp. isolated from soil,
air, fresh water, sewage, seawater,
chlorinated swimming pools, domestic
tap water, bottled water, hospitals, airconditioning units, contact lens cases
▪ Life stages
▫ Metabolically active trophozoite
▫ Dormant stress resistant cyst
▪ Generally free living bacterivores, can cause
human infection (acanthamebiasis)
Granulomatous amoebic encephalitis
▪ Infection associated with
immunosuppression (e.g. diabetes,
HIV/AIDS, hematological malignancy,
malnutrition, hepatic cirrhosis, chronic renal
failure, systemic lupus, chemotherapy)
▪ Parasite enters body through cuts in skin/
inhalation → hematogenous spread to
CNS → invasion of connective tissue
→ inflammatory response → neuronal
damage
Endosymbiosis, secondary infection
▪ Several human pathogens infect, replicate
within Acanthamoeba
▫ Legionella pneumophila, Pseudomonas
aeruginosa, some strains of E. coli,
Staphylococcus aureus
▪ Replication inside Acanthamoeba →
enhanced growth in human macrophages,
increased antibiotic resistance → more
virulent, fulminant infections
SIGNS & SYMPTOMS
confusion), sepsis → progressive
worsening over weeks/months → death
DIAGNOSIS
DIAGNOSTIC IMAGING
Brain CT/MRI
▪ Meningeal exudate, pseudotumoral lesions,
multiple space-occupying lesions with ring
enhancement
LAB RESULTS
Lumbar puncture
▪ Often contraindicated due to risk of
herniation associated with mass lesions
▪ Analytical findings generally nonspecific
▫ Intermediate elevations in white blood
cell count, elevated protein, decreased
glucose levels
▪ Giemsa staining, microscopy
▫ Trophozoites
Tissue biopsy
▪ Brain biopsy
▫ Trophozoites in perivascular space,
thick-walled cysts on light microscopy;
PCR/DNA probes may reveal
Acanthamoeba
▫ Immunocompetent host: granulomatous
lesions
▫ Severely immunosuppressed host:
insufficient CD+ve T-cells to mount
granulomatous response → perivascular
cuffing with amoebae in necrotic tissue
▪ If other organs involved (e.g. skin,
conjunctiva, lungs)
▫ Trophozoites
▪ Fever, headache, seizures, focal
neurological signs (e.g. cranial nerve
palsies), mental status change (e.g.
26
Chapter 64 Central Nervous System Infections
TREATMENT
MEDICATIONS
▪ Current treatment regimes uncertain (based
on in vitro studies, case reports)
▫ Antifungal, antiparasitic agents in
combination
▫ Empiric antifungal regime: miltefosine,
fluconazole, pentamidine isethionate
+/- trimethoprim-sulfamethoxazole,
metronidazole, macrolide antibiotic
SURGERY
▪ Single cerebral lesions
▫ Surgical resection
NAEGLERIA FOWLERI (PRIMARY
AMEBIC MENINGOENCEPHALITIS)
osmosis.org/learn/naegleria_fowleri
PATHOLOGY & CAUSES
▪ Thermophilic, free-living ameba, found in
bodies of warm (stagnant), freshwater
TYPES
▪ Life cycle, three forms
1. Cyst
▫ Immotile, dormant, survival phase
▫ Smooth, single-layered cell wall with
single nucleus, naturally resistant to
environmental factors
▫ Formation of cysts induced by
unfavorable conditions such as food
shortage, overcrowding, desiccation,
accumulation of waste products, cold
temperatures (< 10° celsius)
2.Trophozoite (ameboid)
▫ Feeding, reproductive, infective phase
▫ Transformation into trophozoites occurs
around 25° celsius
▫ Reproduction occurs via binary fission
(single cell divides into two offspring),
optimal temperature 42° celsius
3.Biflagellate (two flagella)
▫ Mobile, infective phase
▫ Pear-shaped body with two flagella
▫ Flagellate phase occurs when ameba
encounters change in fluid ionic
concentration → allows movement to
suitable environment
▪ In human tissues Naegleria fowleri exists
as ameboid trophozoite; flagellate form
may be found in CSF/during initial nasal
insufflation
Primary amoebic meningoencephalitis
▪ AKA naegleriasis
▪ Rare infection, fatality rate > 95%
▪ Mechanism of entry
▫ Insufflated into sinuses during waterbased activities → attaches to olfactory
epithelium → follows olfactory axon
through cribiform plate → migration to
olfactory bulbs → spread throughout
brain → diffuse meningoencephalitis
▪ In tissues, Naegleria fowleri feeds via two
mechanisms; feeding on neurological tissue
→ necrosis, bleeding
▫ Phagocytosis of red, white blood cells
▫ Piecemeal consumption of astrocytes,
neurons via amoebostome (actin-rich
sucking apparatus extended from cell
surface)
27
SIGNS & SYMPTOMS
▪ Symptoms appear 1–9 days after nasal
exposure → death likely follows within two
weeks
▪ Change in sensation of taste, smell;
headache, fever, nausea, stiff neck,
seizures, coma
DIAGNOSIS
DIAGNOSTIC IMAGING
Brain imaging
▪ Initially unchanged
▫ Reveals associated complications
Leptomeningeal enhancement, diffuse
subarachnoid hemorrhage, oedema,
hydrocephalus, multiple cerebral infarcts
LAB RESULTS
▪ Lumbar puncture
▫ CSF microscopy: motile amebae/
fluorescent antibody staining
▫ CSF PCR: Naegleria fowleri DNA
▫ CSF culture: Naegleria fowleri can be
grown on nonnutrient agar coated
with E. coli → drop of CSF of infected
individual added, incubated at 37°
celsius; clearing of E. coli in thin tracks
indicative of trophozoite feeding → likely
infection
TREATMENT
MEDICATIONS:
▪ Amphotericin B +/- fluconazole
▪ Miltefosine
TOXOPLASMA GONDII
(TOXOPLASMOSIS)
osmosis.org/learn/toxoplasma_gondii
PATHOLOGY & CAUSES
▪ Obligate intracellular parasite capable of
infecting nearly all warm-blooded animals
▫ Only definitive hosts: biological family
Felidae (e.g. house cats)
▪ 30–50% of global population exposed, may
be chronically infected
Life cycle
▪ Sexual reproduction
▫ Consumes infected animal meal (e.g.
mouse) → parasite survives transit
through stomach → infects small
intestinal epithelial cells → parasites
undergo sexual development,
reproduction → millions of thick-walled,
zygote-containing, oocytes produced
▪ Felid shedding
▫ Infected epithelial cells rupture →
release oocytes into intestinal lumen
→ shedding in feces → spread via soil,
water, food
▫ Oocysts highly resilient; can survive,
remain infective for months in cold, dry
climates
▪ Infection of intermediate host
▫ Ingestion of oocysts by warm blooded
animals (e.g. humans) → oocyst wall
dissolved by proteolytic enzymes
in stomach, small intestine → frees
sporozoites from within oocyst →
parasites invade intestinal epithelium,
surrounding cells → differentiation into
tachyzoites (motile, quickly-multiplying
phase)
▪ Asexual reproduction in intermediate host
▫ Tachyzoites replicated inside specialized
vacuoles until host cell dies, ruptures
→ release, hematogenous spread of
tachyzoites to all tissues
28
Chapter 64 Central Nervous System Infections
▪ Formation of tissue cysts
▫ Host immune response → tachyzoite
conversion → bradyzoites (semidormant, slowly dividing stage) → inside
host cells known as tissue cysts → can
form in any organ; predominantly brain,
eyes, striated muscle (including cardiac
muscle)
▫ Consumption of tissue cysts in meat
from infected animal
▫ Primary means of infection (e.g. pork,
lamb)
▫ Tissue cysts maintained in host tissue
for remainder of life via periodic cyst
rupture, re-encysting
RISK FACTORS
▪ Consumption of raw/undercooked meat;
ingestion of contaminated water, soil/
vegetables; previous blood transfusion/
organ transplant; transplacental
transmission
COMPLICATIONS
▪ Toxoplasmic chorioretinitis
▫ AKA ocular toxoplasmosis
▫ Common cause of posterior segment
infection
▫ Majority of cases acquired; also strongly
associated with congenital infection
SIGNS & SYMPTOMS
▪ Initial infection (immunocompetent host)
▫ Mild flu-like symptoms (e.g. swollen
lymph nodes, headache, fever, fatigue,
muscle aches, pains)
▪ Congenital infection
▫ Chorioretinitis (unilateral decrease in
visual acuity), hydrocephalus, seizures,
lymphadenopathy, hepatosplenomegaly
▪ Chronic/latent infection
▫ Asymptomatic in healthy hosts
▪ Immunocompromised host
▫ Active infection (toxoplasmosis)
▫ Headache, confusion, poor coordination,
seizures, cough, dyspnea
▫ Reactivation of latent infection:
worsening of immunosuppression
due to progression of underlying
disease (e.g. HIV/AIDS, iatrogenic
immunosuppression) → loss of immune
balance → progression to active
infection
DIAGNOSIS
DIAGNOSTIC IMAGING
CT scan with contrast
▪ Multiple 1–3 cm hypodense regions with
nodular/ring enhancement predominantly in
basal ganglia, corticomedullary junction
T2 weighted MRI
▪ Iso/hyper-intense lesions surrounded by
perilesional edema
Fundoscopy
▪ Toxoplasmic chorioretinitis
▫ Unifocal area of acute-onset
inflammation adjacent to old
chorioretinal scar
Figure 8.1 A histological section of
the cerebrum demonstrating cerebral
toxoplasmosis. There are bradyzoites present
and a mixed inflammatory infiltrate which
includes eosinophils.
LAB RESULTS
PCR (blood, CSF)
▪ Toxoplasma gondii DNA (inactive cysts
may evade detection)
Antibody titres
▪ IgG (persist for life)
29
▫ Evidence of current/previous infection
▪ IgM (acute infection)
▫ Occur in weeks after initial infection,
remain detectable for months
▫ Antibody avidity testing may clarify
nature of infection; early toxoplasmaspecific IgG has low affinity for
toxoplasma antigen; affinity increases
with duration of infection
▪ Sabin–Feldman dye test
▫ Requires specialised laboratories (live
Toxoplasma gondii required); high titers
→ acute infection
▫ Patient serum treated with Toxoplasma
trophozoites + complement, incubated
→ methylene blue added (membrane
stain) → if anti-toxoplasma antibodies
present, complement facilitates lysis of
parasite membrane → no staining of
lysed membrane
▫ No antibodies in serum → intact
membranes → membrane stained blue
under microscopy
▪ Tissue (brain/lymph node/muscle) biopsy
▫ Tachyzoites (acute infection) may be
demonstrated in tissues/smears
TREATMENT
MEDICATIONS
▪ Prevention
▫ Trimethoprim/sulfamethoxazole
▪ Acute infection
▫ Antimalarials: pyrimethamine
▫ Antibiotics: sulfadiazine with
pyrimethamine, clindamycin, spiramycin
▪ Latent infection
▫ Cysts not sufficiently penetrated by
traditional therapy
▫ Atovaquone (antimalarial) +/clindamycin (lincomycin antibiotic)
▪ Toxoplasmic chorioretinitis
▫ Sight-threatening lesions
▫ Triple therapy: pyrimethamine,
sulfadiazine, folinic acid
▫ Mono-antibiotic therapy: trimethoprimsulfamethoxazole, clindamycin,
spiramycin
Figure 8.2 An MRI scan of the head in
the axial plane demonstrating cerebral
toxoplasmosis. There are numerous
peripherally enhancing nodules in the basal
ganglia.
30
NOTES
NOTES
CESTODES (TAPEWORMS)
GENERALLY, WHAT ARE THEY?
PATHOLOGY & CAUSES
▪ Human gastrointestinal tract parasites;
AKA tapeworms
▫ Adult tapeworms live in intestines
▫ Larvae live in different tissue (brain, liver,
eye, etc.)
▪ Tripartite body
▫ Head/scolex (contain suckers, hooks/
attachment organs)
▫ Thin neck
▫ Trunk (made of numerous proglottids)
▪ Hermaphroditic
▫ Each proglottid has male, female organs
▪ Transmission
▫ Egg/larvae-contaminated water/food
ingestion
RISK FACTORS
▪
▪
▪
▪
▪
Poor hygiene
Low socioeconomic status
Raw/undercooked fish/meat
Livestock exposure
Living/travelling in endemic areas
SIGNS & SYMPTOMS
▪ Tapeworm species-dependent
▪ Can be asymptomatic, abdominal pain.
nausea/vomiting, weight loss
DIAGNOSIS
DIAGNOSTIC IMAGING
MRI, CT scan, ultrasound
▪ Cyst presence
LAB RESULTS
▪ Microscopy
▫ Identify eggs/proglottids in stool
▪ Complete blood count (CBC), serology
TREATMENT
▪ Tapeworm species-dependent
MEDICATIONS
▪ Anthelmintics
COMPLICATIONS
▪
▪
▪
▪
Cysticercosis (Taenia)
Cyst rupture
Intestinal obstruction
Malabsorption → vitamin B12 deficiency →
megaloblastic anemia
31
Chapter 61 Cestodes (Tapeworms)
DIPHYLLOBOTHRIUM LATUM
osms.it/diphyllobothrium-latum
PATHOLOGY & CAUSES
▪ AKA fish tapeworm
▪ Longest human-infecting tapeworm
(4–15m/13–49ft)
▪ Causes diphyllobothriasis in humans
▪ Proglottids
▫ Width > length
▪ Competes for vitamin B12 → vitamin B12
deficiency
CAUSES
▪ Raw/undercooked fish → larvae ingestion
COMPLICATIONS
▪ Tapeworms → mechanical intestinal
obstruction
▪ Malabsorption → weight loss
▪ Vitamin B12 deficiency → megaloblastic
anemia
SIGNS & SYMPTOMS
▪ Vitamin B12 deficiency
▫ Impaired oxygen delivery:
fatigue, activity intolerance, pallor,
compensatory mechanisms (↑ heart
rate, bounding pulse)
▫ Neuronal demyelination: numbness,
tingling, weakness
▪ Weight loss
▪ Abdominal pain
DIAGNOSIS
LAB RESULTS
▪ Megaloblastic anemia; e.g. increased mean
corpuscular volume (MCV)
▪ Microscopy
▫ Identify eggs/proglottids in stool
▪ ↓ serum vitamin B12
TREATMENT
MEDICATIONS
▪ Anthelmintics
32
ECHINOCOCCUS GRANULOSUS
(HYDATID DISEASE)
osms.it/echinococcus-granulosus
PATHOLOGY & CAUSES
▪ Parasitic infection caused by E. granulosus
▫ AKA echinococcosis
▪ Produce protoscoleces
▫ Juvenile scolex invaginated in cysts
▫ Tapeworm maturation in definitive
host’s intestine
▪ Humans (incidental hosts); herbivores
(intermediate hosts); canids (definitive
hosts)
CAUSES
▪ Viable parasite egg-containing food
consumption
RISK FACTORS
▪ Parasite/egg-contaminated food/water
ingestion
▪ Close contact with infected animals
Figure 61.2 The gross pathology of hydatid
cysts excised from the lung.
Figure 61.1 A scolex of the organism
Echinococcus granulosus, the causative
agent of hydatid disease.
COMPLICATIONS
▪ Arise as cysts migrate, grow in size, rupture
▫ Liver: eosinophilia, pruritus, jaundice,
urticaria, liver abscess, anaphylaxis
▫ Peritoneal cavity: peritonitis, pancreatitis
▫ Pleural space: abscess formation →
pneumothorax/pleural effusion
▫ Bronchial tree: respiratory distress,
hemoptysis
▫ Heart: cardiomegaly/pericardial effusion
▫ Kidney: glomerulonephritis
▪ Large cyst compression effect
▫ Heart: large cyst in liver → compression
of right heart
▫ Cerebral/spinal cord (CNS): neurological
deficits
▫ Liver/biliary tree cysts: obstructive
jaundice/cholangitis; venous drainage
obstruction → portal hypertension →
Budd–Chiari syndrome (abdominal pain,
ascites, hepatomegaly)
33
Chapter 61 Cestodes (Tapeworms)
SIGNS & SYMPTOMS
▪ Initially asymptomatic
▪ Depend on affected organs
▫ Liver: right upper quadrant pain,
hepatomegaly, nausea, vomiting
▫ Lungs: cough, chest pain, dyspnea,
hemoptysis
▪ Other organs (rarely affected)
▫ Heart: jugular venous distention,
dyspnea
▫ Musculoskeletal: diffuse pain, pathologic
fractures
▫ Kidney: hematuria, flank pain
▫ CNS: headache, motor deficit, seizure,
coma
▪ Puncture-aspiration-injection-reaspiration
(PAIR)
▫ Ultrasound/CT scan-guided cyst
puncture
▫ Aspirate cystic fluid
▫ Inject scolicidal solution
▫ Reaspirate cystic solution
▫ Repeat procedure until aspirate clears
▫ Fill cyst with isotonic saline
DIAGNOSIS
DIAGNOSTIC IMAGING
Ultrasound/MRI/CT scan
▪ Cyst presence
LAB RESULTS
▪ Enzyme-linked immunosorbent assay
(ELISA)
▫ Echinococcal antigen detection in cystic
fluid
▪ Indirect hemagglutination
▫ Echinococcal antigen detection
▪ Immunodiffusion/immunoelectrophoresis
▫ Echinococcal-specific antibody detection
▪ Biopsy/cyst aspiration
Figure 61.3 A CT scan of the abdomen in
the axial plane demonstrating a large hepatic
hydatid cyst. The numerous daughter cysts
are faintly visible.
TREATMENT
MEDICATIONS
▪ Albendazole/ mebendazole
▫ Uncomplicated cases
Figure 61.4 A histological section through a
hydatid cyst wall showing a typical laminated
structure.
SURGERY
▪ Complicated cases
▫ Rupture, vital structure compression,
cysts with diameter > 10cm/3.94in
34
NOTES
NOTES
CHLAMYDIA
MICROBE OVERVIEW
▪ Gram-negative, obligate intracellular
bacteria
▪ AKA “energy parasites”; rely on host cell for
adenosine triphosphate (ATP) synthesis
▪ Primarily infects epithelium, mucous
membranes
Morphology
▪ Coccoid; cell walls don’t contain
peptidoglycan
Replication
▪ Intracellular life cycle: infectious stage
(spore-like elementary body) attaches to
host cell via endocytosis → reorganizes
within cellular vacuole into reticulate
body (vegetative form) → reproduces,
forms multiple reticulate bodies → forms
elementary bodies → released from host
cell → continues infectious process
CHLAMYDIA SPECIES
(PNEUMONIA)
osms.it/chlamydia-species
PATHOLOGY & CAUSES
▪ Species of chlamydia; primarily causes
community-acquired pneumonia
▪ Transmitted via respiratory secretions
▪ Can also infect endothelial cells →
atherosclerosis
RISK FACTORS
▪ ↑ risk with age
COMPLICATIONS
▪ Otitis media, sinusitis, parapneumonic
effusions, pericarditis
35
Chapter 62 Chlamydia
SIGNS & SYMPTOMS
▪ Gradual onset
▪ General malaise, myalgia, fever, chills,
pharyngitis, hoarseness, sinusitis, cough
▪ Extra-respiratory manifestations:
meningoencephalitis, reactive arthritis,
myocarditis
DIAGNOSIS
LAB RESULTS
▪ Microbial identification: serology,
polymerase chain reaction (PCR),
nasopharyngeal swab culture, direct
antigen testing
▪ Complete blood count (CBC): normal white
blood cell count
OTHER DIAGNOSTICS
▪ History, physical examination: lung sounds
(e.g. crackles, wheezing)
DIAGNOSTIC IMAGING
Chest X-ray
▪ Unilateral patchy infiltrates
TREATMENT
MEDICATIONS
▪ Antibiotics
CHLAMYDIA TRACHOMATIS
osms.it/chlamydia-trachomatis
PATHOLOGY & CAUSES
▪ Species of chlamydia; primarily affects eyes,
urogenital tract
▪ Different serovars cause diverse disease
states
▫ A, B, Ba, C: trachoma
▫ D–K: urogenital infection, conjunctivitis
▫ L1, L2, L2a, L2b, L3: lymphogranuloma
venereum
RISK FACTORS
▪ Risky sexual practices (e.g. multiple sex
partners, unprotected sex)
▪ Impaired mucous membrane barrier (e.g.
cervical friability)
▪ History of sexually transmitted disease
▪ Exposure during birth
COMPLICATIONS
▪ Ocular: ophthalmia neonatorum
(conjunctivitis), blindness
▪ Genitourinary: pelvic inflammatory disease
(PID), PID-associated ectopic pregnancy,
infertility, proctitis, cervicitis, urethritis
▪ Chlamydial pneumonia, bronchitis,
perihepatitis (Fitz-Hugh–Curtis syndrome);
↑ risk of acquiring/transmitting HIV due to
genital inflammation
SIGNS & SYMPTOMS
Trachoma
▪ Chronic, granulomatous inflammation of
eye → corneal ulceration, scaring, pannus
formation → blindness
Adult conjunctivitis
▪ Inclusion (purulent erythematous injection
of epithelial surface) conjunctivitis,
mucopurulent discharge → keratitis
Urogenital infections
▪ Individuals who are biologically female
▫ May be asymptomatic
▫ Bartholinitis, cervicitis (mucopurulent
endocervical discharge), endometritis,
salpingitis, urethritis (dysuria, pyuria)
▫ PID: uterine/adnexal tenderness
▫ Perihepatitis: right upper quadrant
(RUQ) pain
36
▪ Individuals who are biologically male
▫ Urethritis: dysuria, watery/mucoid
discharge
Neonatal conjunctivitis
▪ Eyelid swelling, watery/purulent discharge,
red, thickened conjunctiva (chemosis)
▪ Conjunctival scarring, corneal
vascularization
▪ ↑ risk of developing C. trachomatis
pneumonia
Infant pneumonia
▪ Diffuse, interstitial disease; rhinitis, staccato
cough
Figure 62.1 Purulent discharge from the
cervix of an individual with chlamydia
infection.
Lymphogranuloma venereum (LGV)
▪ Anorectal disease: anorectal pain,
tenesmus (feeling of incomplete bowel
evacuation), rectal bleeding/discharge,
constipation
▪ Painless ulcer → inflammation of lymph
nodes (preauricular, submandibular,
cervical) → progression to systemic
symptoms
▪ Population at highest risk: individuals who
are biologically male who have same-sex
intercourse (MSM)
DIAGNOSIS
LAB RESULTS
▪ Microbial identification: nucleic acid
amplification test (NAAT)
Figure 62.2 A cervical smear from
an individual infected with Chlamydia
trachomatis. There are numerous organisms
contained within intracellular vacuoles.
OTHER DIAGNOSTICS
▪ Clinical presentation, history
TREATMENT
MEDICATIONS
▪ Antibiotics
▪ Sexual partners should also be treated
37
Chapter 2 Acyanotic Defects
NOTES
COCCOBACILLI: AEROBES
MICROBE OVERVIEW
▪ Intermediate shape between cocci
(spherical bacteria), bacilli (rod-shaped
bacteria)
▪ Gram-negative, obligate aerobe, nonmotile, non-spore forming
BORDETELLA PERTUSSIS
(PERTUSSIS/WHOOPING COUGH)
osms.it/bordetella-pertussis
PATHOLOGY & CAUSES
▪ Infectious agent, causes pertussis
▪ Strict human pathogen
▪ Tropism to respiratory epithelium
Pathophysiology
▪ Bacteria enters upper respiratory tract,
releases toxins
▪ Filamentous hemagglutinin, pertactin,
agglutinogen
▫ Anchors to epithelial cells
▪ Tracheal cytotoxin
▫ Paralysis of respiratory cilia → ↑
accumulation of mucus in airways →
violent cough reflex
▪ Pertussis toxin
▫ Stimulates T cells to divide, blocks
them from leaving blood, migrating into
tissues → lymphocytosis
▫ ↑ sensitivity of respiratory tissues to
histamine → ↑ vascular permeability →
fluid leaks into airway tissues → airway
edema → dyspnea
38
▪ Adenylate cyclase toxin
▫ ↑ conversion of adenosine
triphosphate (ATP) to cyclic adenosine
monophosphate (AMP) within
phagocytes → disbalance in cellular
signaling mechanism → phagocytes
unable to correctly respond to infection,
undergo apoptosis
Stages of infection
▪ Catarrhal: follows incubation period
(approx. one week); lasts two weeks, very
contagious
▪ Paroxysmal: 1–6 weeks
▪ Convalescent: 2–3 weeks
RISK FACTORS
▪ Infants too young to have completed
immunization
▪ Unimmunized individuals
▪ Debilitation of immune system
COMPLICATIONS
▪ Apparent life-threatening event (ALTE)
▫ Young infants; gasping, cyanosis, apnea
▪ Hypoxia
▫ Seizures, encephalopathy, death
▪ Pneumonia, pneumothorax
SIGNS & SYMPTOMS
DIAGNOSIS
LAB RESULTS
▪ Nasopharyngeal swab culture (Bordet–
Gengou agar)
▫ Detects microbe; mercury drop colonies
▪ Polymerase chain reaction (PCR)
▫ Detects microbe
▪ Serology
▫ Detects microbe; antibody levels
▪ Complete blood count (CBC)
▫ Leukocytosis (esp. young infants)
OTHER DIAGNOSTICS
▪ History, physical examination
▫ Sudden, dramatic coughing attacks;
lung sounds (e.g. whooping)
TREATMENT
MEDICATIONS
▪ Prophylactic
▫ Diphtheria, tetanus, acellular pertussis
(DTaP) vaccine
▪ Macrolide antibiotics
OTHER INTERVENTIONS
▪ Compulsory isolation of infected individual
▪ Catarrhal: nasal congestion; mild cough;
sneezing; low-grade fever; red, watery eyes
(similar to common cold)
▪ Paroxysmal: high-pitched whooping during
inhalation; uncontrollable coughing fits →
vomiting, fainting, rib fracture, petechiae in
face, subconjunctival hemorrhages, hernias
▪ Convalescent stage: coughing
improvement; decreased paroxysm,
whooping; healing of airways
39
Chapter 63 Coccobacilli: Aerobes
FRANCISELLA TULARENSIS
(TULAREMIA)
osms.it/francisella-tularensis
PATHOLOGY & CAUSES
▪ Infectious agent that causes zoonosis
tularemia
▪ Facultative, gram-negative intracellular
bacteria
▪ Highly infectious, virulent; potential
bioterrorism agent
Transmission
▪ Direct contact with infected animals (e.g.
inoculation of mucous membranes via
contaminated hands/infected material)
▪ Ingestion of contaminated water/meat
▪ Airborne spread via contaminated materials
(e.g. dust, hay, grass, lab specimens)
▪ Insect vectors
▫ Ticks, mosquitoes, horse flies, fleas, lice
Pathophysiology
▪ Francisella tularensis enters body →
phagocytosed by macrophages → impairs
phagosome-lysosome fusion, rapidly
proliferates within macrophage → infected
macrophage undergoes apoptosis →
bacteria released, infection spread
COMPLICATIONS
▪ Lymph node suppuration, sepsis, renal
failure, rhabdomyolysis, hepatitis,
pneumonia
SIGNS & SYMPTOMS
▪ Fever, chills, malaise, lethargy, anorexia,
chest/muscle soreness
▪ Ulceroglandular: papulo-ulcerative lesion
at point of contact with animal/vector;
lymphadenopathy
▪ Glandular: lymphadenopathy (no skin
lesions)
▪ Oculoglandular: pain/irritation of eye;
periorbital edema/erythema; increased
tearing; photophobia; regional adenopathy
▪ Oropharyngeal: sore throat; cervical lymph
node enlargement; pharyngitis; tonsillitis
▪ Pneumonic: dry cough; breathing
difficulties; substernal chest pain
▪ Typhoidal: very high fever; abdominal
pain; diarrhea; vomiting; diffuse abdominal
tenderness
TYPES
▪ Forms of tularemia: ulceroglandular (75%),
glandular, oculoglandular, oropharyngeal,
pneumonic, typhoidal
RISK FACTORS
▪ Work-related
▫ Lab workers, farmers, veterinarians,
gardeners, hunters, butchers
▪ Skin exposure to vectors (esp. in summer)
Figure 63.1 An ulcer on the skin of an
individual with tularemia.
40
DIAGNOSIS
LAB RESULTS
▪ Culture
▫ Buffered charcoal yeast extract (BCYE)
agar
▪ PCR, direct fluorescent antibody (DFA) test
▫ Detects microbe
▪ Serology
▫ IgM, IgG antibodies appear after 2
weeks
TREATMENT
MEDICATIONS
▪ Antibiotics (e.g. streptomycin, gentamicin,
doxycycline)
OTHER DIAGNOSTICS
▪ History, physical examination
41
Chapter 2 Acyanotic Defects
NOTES
COCCOBACILLI: FACULTATIVE
ANAEROBES
MICROBE OVERVIEW
▪ Intermediate shape between cocci
(spherical bacteria), bacilli (rod-shaped
bacteria)
▪ Gram-negative, facultative anaerobes,
nonmotile, nonspore-forming
BRUCELLA
osms.it/brucella
PATHOLOGY & CAUSES
▪ Characteristics
▫ Zoonotic infection
▫ Urease, catalase positive
▫ Facultatively intracellular
▫ Sensitive to heat, ionizing radiation,
disinfectants, pasteurization
▪ Virulence factors
▫ Lipopolysaccharide (LPS): promotes cell
entry, evasion, intracellular killing
▫ Type IV secretion system (key virulence
factor): injection of effector molecules
into host cell → modifies endoplasmic
reticulum, enables replication
▪ Culture
▫ Isolation specimen: blood, bone marrow,
body fluids, tissues
▫ Media: biphasic (solid, liquid) RuizCastaneda blood culture/modern
automated blood culture systems
(faster, more effective)
▫ Raised, convex colonies with smooth,
shiny surface
▪ Causative agent of brucellosis
▪ Most common zoonotic infection to cause
disease in humans
Transmission
▪ Contact with infected animals (e.g. sheep,
cattle, goats, pigs, etc)
▫ Entry of bacteria through skin lesions,
conjunctival inoculation, inhalation of
contaminated aerosol
▪ Ingestion of contaminated animal
products (e.g. unpasteurized milk, cheese;
undercooked meat)
▫ Remains viable up to two days in milk at
8°C/46.4°F, three weeks in frozen meat,
three months in goat cheese
Pathogenesis
▫ Inoculation of bacteria → ingestion by
polymorphonuclears, macrophages
→ passage to local lymph nodes →
bacteria replicates intracellularly →
some bacteria avoid intracellular killing
by different strategies (e.g. inducing
phagocyte apoptosis, inhibiting
phagocyte-lysosome fusion) → chronic
infection
TYPES
Acute infection
▪ Localized infection (30% of cases), can
affect any organ
42
▫ Skeletal (most common): arthritis,
spondylitis, sacroiliitis, osteomyelitis
▫ Pulmonary: bronchitis, interstitial
pneumonitis, lobar pneumonia,
pulmonary nodules, pleural effusion,
empyema, abscesses
▫ Cardiac: endocarditis, myocarditis,
pericarditis, endarteritis
▫ Alimentary: cholecystitis, ileitis, colitis,
pancreatitis
▫ Reticuloendothelial: reactive hepatitis,
granulomas, acute hepatitis with focal
necrosis (B. melitensis), formation
of noncaseating sarcoidosis-like
granulomas (B. abortus), suppurative
abscess formation (B. suis) in liver
▫ Genitourinary: orchitis, epididymitis
▫ Hematological: anemia, leukopenia,
thrombocytopenia, pancytopenia,
disseminated intravascular coagulation
▫ Neurologic: meningitis, encephalitis,
myelitis, radiculitis, neuritis, mycotic
aneurysms, brain abscess
▫ Ocular: uveitis, keratoconjunctivitis,
corneal ulcers, iridocyclitis, nummular
keratitis, choroiditis, optic neuritis,
papilledema, endophthalmitis
▫ Dermatologic: nonspecific skin
eruptions, ulcerations, petechiae,
purpura, granumanifestationslomatous
vasculitis, abscesses
Chronic infection
▪ Symptoms persist one year after diagnosis;
localized infection, relapse
RISK FACTORS
▪ Occupational exposure
▫ Lab health care workers, farmers,
slaughterhouse workers, veterinarians
COMPLICATIONS
▪ Infection during pregnancy → intrauterine
infection, premature delivery, spontaneous
abortion, miscarriage
▪ Endocarditis damage, destroy heart valves
▪ Leading cause of death by brucellosis
▪ Skeletal
▫ May cause long-term damage, bone/
joint malformations
▪ Neurologic
▫ May lead to permanent brain damage
▪ Ocular
▫ Visual impairment
43
Chapter 64 Coccobacilli: Facultative Anaerobes
SIGNS & SYMPTOMS
▪ Range from asymptomatic to severe illness
▪ Onset of symptoms can be acute/insidious
▪ Incubation
▫ 1–4 weeks to several months
▪ Acute generalized infection
▫ Acute undulating fever (key sign),
arthralgia, myalgia, fatigue, headache,
night sweats, malaise, weight loss
▫ Hepatomegaly, splenomegaly,
lymphadenopathy
▫ Foul-smelling perspiration (characteristic
sign)
▪ Localized infection
▫ Symptoms depend on organ/organ
system affected
DIAGNOSIS
LAB RESULTS
▪ Rising titers of specific antibodies
▫ Initial rise in IgM class titers, followed in
several weeks by predominance of IgG
antibodies; both decrease over time with
treatment
▪ Anemia, thrombocytopenia
Microbe identification
▪ Positive bodily fluids/tissue culture
▪ Serum agglutination, enzyme-linked
immunosorbent assay (ELISA)
▪ Polymerase chain reaction (PCR)
▪ Lysis-centrifugation technique
OTHER DIAGNOSTICS
▪ History of travel, food consumption,
occupation
TREATMENT
MEDICATIONS
▪ Six-week course of doxycycline plus
streptomycin/gentamicin/doxycycline plus
rifampin
▪ In children < eight years old
▫ Trimethoprim-sulfamethoxazole (TMPSMX) plus rifampin
SURGERY
▪ Surgical interventions sometimes necessary
for osteoarticular manifestations (e.g.
pyogenic joint effusions), hepatosplenic
granulomas/abscesses, cardiac
complications (e.g. valve replacement
surgery)
OTHER INTERVENTIONS
▪ Prophylaxis
▫ Biosafety level 3 in laboratories
recommended while handling Brucella
cultures
▫ No vaccines for humans; live attenuated
vaccines containing strains of B.
abortus, B. melitensis used for animals
44
AfraTafreeh.com exclusive
HAEMOPHILUS DUCREYI
osms.it/haemophilus-ducreyi
PATHOLOGY & CAUSES
▪ Virulence factors
▫ Lipooligosaccharide
▫ Pili: provides attachment of bacteria
▫ Soluble cytolethal distending toxin,
cytotoxic hemolysin, hemoglobinbinding protein, copper-zinc superoxide
dismutase, filamentous hemagglutininlike protein, zinc-binding periplasmic
protein
▪ Culture
▫ Isolation specimen: genital ulcer swab,
lymph node aspirate
▫ Media: enriched growth medium
contains factor X (hemin), serum
incubated at 33–35ºC/91.4–95°F
with CO2; small, heterogenous, gray/
translucent colonies
▪ Causative agent of sexually transmitted
genital ulcer called chancroid (AKA
ulcus molle), associated inguinal
lymphadenopathy
▪ Some strains causes cutaneous ulcers in
children in South Pacific, parts of equatorial
Africa
Transmission
▪ Sexual intercourse (genital ulcers)
▪ Nonsexual transmission (cutaneous ulcers)
Pathogenesis
▪ Incubation
▫ 4–10 days
▪ Inoculation through epidermal
microabrasions → attachment of bacteria
to extracellular matrix in skin via pili,
lipooligosaccharide → attachment to cells
via specific heat shock protein (GroEL)
→ cytotoxin release, epithelial injury →
formation of erythematous papule →
evolves into pustule → pustule ruptures,
forms ulcer
RISK FACTORS
▪ Uncircumcised individuals, poverty, multiple
sexual partners
COMPLICATIONS
▪ Increases risk for HIV contraction
SIGNS & SYMPTOMS
▪ Single/multiple painful genital ulcers on
erythematous base, 1–2cm/0.39–0.79in
diameter with sharply demarcated borders;
base of ulcer covered with purulent
exudate, bleeds easily when scraped
▪ Predilection sites
▫ Prepuce, coronal sulcus, glans penis in
individuals who are biologically male
▫ Labia, vaginal introitus, perianal area in
individuals who are biologically female
▪ Individuals who are biologically female
▫ Dysuria, dyspareunia, vaginal discharge,
rectal bleeding, painful defecation
▪ Inguinal lymphadenopathy in approx. 50%
of cases (more common in individuals who
are biologically male)
▫ Painful fluctuant buboes (swollen lymph
nodes); if untreated, may spontaneously
rupture, form draining sinus, releases
pus
45
Chapter 64 Coccobacilli: Facultative Anaerobes
DIAGNOSIS
LAB RESULTS
Microbe identification
▪ Diagnosis of confirmed chancroid
▫ Culture (not widely available)
▪ Nucleic acid amplification tests
▫ Not available outside of clinical research
purposes
▪ Polymerase chain reaction (PCR) multiplex
▫ Detection of bacterial DNA
▪ Histologic characteristics of chancroid
OTHER DIAGNOSTICS
▪ Diagnostic criteria for probable chancroid
▫ ≥ one painful genital ulcers
▫ No evidence of Treponema pallidum
infection (by darkfield microscopy/
serologic testing)
▫ No evidence of Herpes simplex virus
(HSV) infection
▫ Appearance of genital ulcers, regional
lymphadenopathy
▪ Purulent exudate in superficial epidermis
with perivascular, interstitial mononuclear
infiltrate in dermis
Figure 64.1 An ulcer on the glans penis of
a male with chancroid. The ulcer is typically
painful, unlike the ulcer of primary syphilis.
TREATMENT
MEDICATIONS
▪ Single-dose therapy with azithromycin/
ceftriaxone
▪ Alternative
▫ Multiple-dose therapy with
ciprofloxacin/erythromycin
OTHER INTERVENTIONS
▪ Fluctuant lymphadenopathy
▫ Needle aspiration, drainage to prevent
spontaneous rupture
46
HAEMOPHILUS INFLUENZAE
osms.it/haemophilus-influenzae
PATHOLOGY & CAUSES
▪ Haemophilus: blood loving
▪ Characteristics
▫ Catalase, oxidase positive
▪ Virulence factors
▫ Polysaccharide capsule: prevents
phagocytosis; causes ciliostasis, evades
mucociliary clearance of bacteria;
classified into six serotypes based on
capsular antigens (A, B, C, D, E, F);
some strains unencapsulated (AKA
nontypable); most clinical isolates
Haemophilus influenzae type B (Hib)/
nontypable
▫ IgA1 protease, adherence factors,
antigenic variation, biofilm formation
▪ Gram stain of exudate shows bacteria
arranged in chains (“school of fish”)
▪ Culture
▫ Isolation specimen: cerebrospinal fluid
(CSF), urine, serum, synovial fluid
▫ Media: chocolate agar/Fildes medium
(hemolyzed erythrocytes) with factor
X (hemin), V (nicotinamide adenine
dinucleotide) supplementation in
aerobic, only factor X supplementation
in anaerobic environment
▫ Convex, smooth, grey/transparent
colonies
▪ Gram-negative coccobacillus → meningitis,
respiratory tract infections
▪ Nontypeable strains colonize nasopharynx
of 40–80% children, adults
▪ Hib colonizes 3–5% children
TYPES
Hib
▪ Epiglottitis in older children, adults
▪ Cellulitis (most common in young children)
▪ Pneumonia
▫ Sometimes with meningitis, epiglottitis
▪ Meningitis, septic arthritis, osteomyelitis
Nontypable
▪ Less invasive due to lack of capsule; causes
mild localized respiratory tract disease in
children, adults
▪ More severe in immunocompromised/
predisposed individuals
▫ Otitis media, sinusitis, purulent
conjunctivitis, bacterial pneumonia
in children (in low-income countries),
neonatal bacteremia
▫ Community-acquired pneumonia in
adults with underlying lung disease
▫ Exacerbation of chronic obstructive
pulmonary disease (COPD)
▫ Meningitis in individuals with
predisposition/conditions causing
leakage of CSF fluid (e.g. sinusitis, otitis
media, head trauma)
Transmission
▪ Direct contact with respiratory tract
secretions/airborne respiratory droplets
Pathogenesis
▪ Inoculation → passage through upper
respiratory tract → adherence to respiratory
epithelium, LPS inhibits mucociliary
clearance → colonization spreads
throughout respiratory tract → sinuses,
otitis, pneumonia
▪ IgA1 protease, antigenic variation,
paracytosis, biofilm formation →
perseverance of bacteria
RISK FACTORS
▪ Viral infection, sickle-cell disease, asplenia,
HIV infection, malignancies, congenital
deficiencies of complement components
COMPLICATIONS
▪ Nontypable in neonates,
immunocompromised individuals →
septicemia, meningitis, septic arthritis
▪ Hib meningitis → subdural effusion/
empyema; ischemic/hemorrhagic cortical
47
Chapter 64 Coccobacilli: Facultative Anaerobes
infarction; cerebritis (nonviral parenchymal
infection of brain); ventriculitis; intracerebral
abscess; hydrocephalus; neurologic
sequelae (e.g. permanent sensorineural
hearing loss, seizures, intellectual disability)
▪ Hib pneumonia can spread to pericardium
→ purulent pericarditis
SIGNS & SYMPTOMS
Hib
▪ Meningitis
▫ Fever, lethargy, irritability, vomiting,
altered mental status
▫ Fulminant course → rapid neurologic
deterioration, respiratory arrest
▫ Positive Kernig’s sign: inability to
straighten leg when hip flexed to 90º
▫ Positive Brudzinski’s sign: flexing of
neck by examiner → flexing of hips,
knees
▪ Epiglottitis
▫ Fever, sore throat, difficulty speaking,
dyspnea → severe stridor, dysphagia,
pooling of secretions, drooling
▫ “Tripod” posture: individual takes sitting
position with trunk leaning forward,
neck hyperextended, chin thrust forward
to get more air through obstructed
airway
▪ Cellulitis
▫ Fever; warm, tender area of erythema/
violaceous discoloration on cheek/
periorbital area
▪ Septic arthritis
▫ Fever, pain, swelling, tenderness,
decreased mobility of affected joint
Nontypeable
▪ Otitis media
▫ Fever, ear pain, irritability, sleep
disturbance, otorrhea
▫ Red bulging tympanic membrane with
decreased mobility upon pneumatic
otoscopy examination
▫ Often conjoined with conjunctivitis
▪ Sinusitis
▫ Fever, persistent purulent nasal
discharge or cough > 10 days
▫ Tenderness over involved paranasal
sinuses
DIAGNOSIS
DIAGNOSTIC IMAGING
Laryngoscopy
▪ Red, swollen epiglottis; aryepiglottic folds
▪ Examine with caution; possible laryngeal
spasm
X-ray
▪ Thumb sign on epiglottis (radiographic
corollary of omega sign)
LAB RESULTS
Microbe identification
▪ Positive Gram stain, bacterial culture of
CSF, synovial fluid, epiglottis, pleural,
pericardial, other sterile fluids
▪ Latex agglutination, enzyme immunoassay,
coagglutination
▫ Type B capsular antigen detection in
CSF, serum, urine
▪ Definitive diagnosis
▫ Culture of fluid obtained by sinus
aspiration, tympanocentesis, tracheal/
lung aspiration, bronchoscopy,
bronchoalveolar lavage
Figure 64.2 An X-ray image of the chest
demonstrating diffuse airway shadows
in an individual with bronchopneumonia.
H. influenzae is a causative organism of
bronchopneumonia.
48
TREATMENT
MEDICATIONS
Prevention
▪ Conjugate Hib vaccines
▫ Routine vaccination of infants of two
months
▪ Rifampin chemoprophylaxis
▫ Individuals in close contact with
infected; incompletely vaccinated
individuals in households with infants/
children < four years old
Hib with meningitis
▪ Third generation cephalosporins
▫ Adults: ceftriaxone
▫ Children: ceftriaxone plus
dexamethasone (decreases immune
response to released LPS upon bacterial
death, lowers chance for destruction of
neurons, neurologic sequelae)
▪ Epiglottitis (life-threatening condition;
prompt treatment paramount)
▫ Ceftriaxone
Nontypable
▪ Amoxicillin/clavulanate, broad-spectrum
cephalosporins, macrolides (azithromycin/
clarithromycin), fluoroquinolones
SURGERY
▪ Epiglottitis
▫ Placement of artificial airway
PASTEURELLA MULTOCIDA
osms.it/pasteurella-multocida
PATHOLOGY & CAUSES
▪ Characteristics
▫ Zoonotic infection (e.g. birds, cats, dogs,
rabbits, cattle, pigs); oxidase, catalase,
nitrate reduction positive
▪ Virulence factors
▫ Polysaccharide capsule: prevents
phagocytosis; divided into serogroups
based on capsular antigens (A, B, C, D,
E)
▫ Lipopolysaccharide: endotoxin
▫ Sialidases, hyaluronidase, surface
adhesins, iron acquisition proteins,
pasteurella multocida toxin (PMT)
▪ Culture
▫ Isolation specimen: respiratory tract
samples, CSF
▫ Media: sheep blood, chocolate, HS,
Mueller–Hinton agar at 37ºC/98.6F;
opaque/gray colonies 1–2mm in
diameter
▪ Medically important subspecies
▫ P. multocida subsp multocida, P.
multocida subsp septica, P. multocida
subsp gallicida
Transmission
▪ Most commonly cat/dog bites, scratches,
licks
TYPES
▪ Soft tissue infections
▫ Cellulitis at site of inoculation (most
common) → abscess, necrotizing
soft tissue infections, septic arthritis,
osteomyelitis
▪ Respiratory infections
▫ Due to underlying chronic pulmonary
disease; glossitis, pharyngitis,
sinusitis, otitis media, epiglottitis,
tracheobronchitis, pneumonia,
empyema, lung abscess
▪ Invasive infection (immunocompromised,
infants)
▫ Bacteremia, meningitis, intra-abdominal
infections (peritonitis, appendicitis),
endocarditis, septic arthritis, ocular
infection
49
Chapter 64 Coccobacilli: Facultative Anaerobes
Pathogenesis
▪ Inoculation → attachment of bacteria to
ECM, cells soft tissue → PMT secretion →
tissue inflammation within 24 hours
COMPLICATIONS
▪ Sepsis, septic shock
SIGNS & SYMPTOMS
▪ Soft tissue infections
▫ Wound inflammation, cellulitis
with purulent drainage; regional
lymphadenopathy
▪ Respiratory tract infections
▫ Fever, malaise, dyspnea, pleuritic chest
pain
▪ Sepsis
▫ Purpura fulminans (rash rapidly
progresses from petechiae, purpura to
gangrene/limb amputation)
▪ Respiratory infection
▫ Wheezing, rhonchi, dullness
DIAGNOSIS
LAB RESULTS
Microbe identification
▪ Culture, PCR, serological testing
OTHER DIAGNOSTICS
▪ History of animal contact
▫ Cat bites pose higher risk for developing
osteomyelitis, septic arthritis
TREATMENT
MEDICATIONS
▪
▪
▪
▪
Penicillins
Tetracyclines
Cephalosporins
Quinolones
50
NOTES
NOTES
COMMA–SHAPED RODS
MICROBE OVERVIEW
▪ Gram-negative, facultative anaerobes,
motile, non-spore forming
CAMPYLOBACTER JEJUNI
osms.it/campylobacter-jejuni
PATHOLOGY & CAUSES
Characteristics
▪ Zoonotic disease
▫ Reservoir in wild, domestic mammals,
birds (esp. poultry)
▪ Oxidase +; invasive; microaerophilic;
sensitive to heat, desiccation, acidity,
irradiation, disinfectants
Virulence factors
▪ Fimbriae-like filaments
▫ Promote attachment to intestinal
epithelial cells
▪ Possesses single, unsheathed flagellum in
one end (monotrichous)/two flagella each at
both ends (amphitrichous)
▫ Provide motility, chemotaxis (mucin is
chemoattractant → tropism for ileum,
colon, rectum)
▪ Surface proteins (eg, PEB1, CadF) promote
colonization, invasion of intestinal epithelial
cells
▪ Lipopolysaccharide (LPS)
▫ Plays role in adherence, evasion of host
immune response (undergoes antigenic
variation)
51
Chapter 65 Comma-shaped Rods
Culture
▪ Isolation specimen: stool, food
▪ Media: blood/charcoal agar with
microaerophilic atmosphere (5–10% O2,
3–5% CO2), thermophilic environment
(optimal 42°C/107.6°F)
▪ C. jejuni: one of most common bacterial
causes of gastroenteritis with acute
diarrhea
Transmission
▪ Fecal-oral, contaminated water
Pathogenesis
▪ Incubation period 1–7 days; tropism for
distal ileum, colon, rectum
▪ Inoculation of bacteria → passage through
upper GI tract → colonization, adherence to
surface epithelium of distal ileum, colon →
non-inflammatory secretory diarrhea (exact
mechanism unknown)
▪ Invasion of intestinal epithelium,
proliferation → release of cytolethal
distending toxin → cell damage,
inflammatory response → dysentery with
fecal leukocytes
▪ Rare: translocates into lamina propria,
spreads to mesenteric lymph nodes
(mesenteric adenitis) → extraintestinal
infections (e.g. meningitis, cholecystitis,
UTI)
▫ Occurs mostly in immunocompromised
RISK FACTORS
▪ Consumption of undercooked meat/
unpasteurized milk
▪ Underlying conditions/medications that
reduce/buffer gastric acidity (e.g. proton
pump inhibitors)
▪ Individuals with HIV/AIDS
COMPLICATIONS
▪ Toxic megacolon, massive bleeding, colonic
perforation
▪ Reactive arthritis
▪ Associated with Guillain–Barré syndrome
▫ Sialic acid contained bacterial core
oligosaccharide can resemble
gangliosides → cross-activation
of autoreactive T/B cells (molecular
mimicry)
SIGNS & SYMPTOMS
▪ Vary in severity depending upon inoculum
concentrations; range from asymptomatic
carriage to systemic illness
▫ Most episodes mild, self-limiting (up to
one week); rarely persists up to several
weeks
▪ Fever, myalgia, malaise, headache (early
symptoms, 1–2 days); severe periumbilical
abdominal pain, cramping, secretory,
inflammatory diarrhea, vomiting
▫ Abdominal pain may mimic acute
appendicitis
▫ Secretory diarrhea: more common in
children
▫ Inflammatory diarrhea: tenesmus,
bloody stools, fecal leukocytes
DIAGNOSIS
LAB RESULTS
▪ Stool culture
▪ Rapid diagnosis with carbolfuchsin stain/
phase-contrast/dark-field microscopic
examination of fresh stool specimen in case
of acute manifestation
▪ PCR-based methods, enzyme
immunoassays (EIAs) directly from stool
OTHER DIAGNOSTICS
▪ Clinical manifestations
▪ Histology
▫ Acute mucosal inflammation with
edema, cellular infiltration of lamina
propria, crypt abscess formation
TREATMENT
MEDICATIONS
▪ Antibiotics for severe cases,
immunocompromised individuals
▫ Erythromycin/azithromycin
OTHER INTERVENTIONS
▪ Replacement of fluids, electrolytes
52
HELICOBACTER PYLORI
osms.it/helicobacter-pylori
PATHOLOGY & CAUSES
Characteristics
▪ Urease +, catalase +, oxidase +; noninvasive
▪ Microaerophilic
▫ Requires oxygen, lower concentrations
than present in atmosphere
Virulence factors
▪ Possesses 2–7 unipolar sheathed flagella
(H-antigen)
▫ Provide motility, chemotaxis (sense
pH, move bacteria towards beneficial
environment)
▪ Lipopolysaccharide (LPS)
▫ Promotes adherence, causes
inflammation
▪ Coccoid form
▫ More resistant form; occurs as
adaptation to hostile environment
outside human body
▪ Urease
▫ Important for survival, colonization
▪ Mucolytic enzymes
▫ Allow passage through mucus layer to
gastric epithelium
▪ Adhesive proteins (Hop proteins)
▪ Vacuolating cytotoxin A (VacA)
▫ Damages epithelial cells; disrupts tight
junctions, causes apoptosis
▪ Cytotoxin associated gene CagA (CagA)
▫ Triggers inflammation
▪ Type IV secretion system
▫ Pili-like structure for injection of
effectors (e.g. CagA)
▪ Proteases, lipases
▪ Biofilm formation
Culture
▪ Isolation specimen: vomitus, diarrheal
stools
▪ Media: blood agar/selective Skirrow’s media
incubated at 37ºC/98.6°F in 5% oxygen;
small, uniformly sized, translucent bacterial
colonies
▪ H. pylori: causative agent of most common
chronic infection in humans; common cause
of duodenal, gastric ulcers, chronic gastritis
Transmission
▪ Unknown; fecal/oral, oral/oral transmission
suggested
▪ Reservoir
▫ Human (majority of cases); found in
primates in captivity, domestic cats,
sheep
▪ Also found in municipal water in endemic
areas of infection with polymerase chain
reaction (PCR) techniques
Pathogenesis
▪ Bacterial urease hydrolyzes gastric luminal
urea to form ammonia → ↑ gastric pH
→ formation of protective layer around
bacteria → survival in hostile gastric
environment
▪ ↑ pH → mucin liquefies → H. pylori passes
through mucous layer to surface epithelium
via bacterial flagella, mucolytic enzymes
→ attaches to specific gastric epithelial
cell receptors via surface adhesins (Hop
proteins) → release of proteases (VacA,
CagA) + host immune response →
inflammation, tissue injury
▪ Disruption of mucous layer → susceptibility
to acid peptic damage
▪ Chronic inflammation, tissue injury together
with acid peptic damage → chronic
gastritis, peptic ulcers (10–20% risk)
RISK FACTORS
▪
▪
▪
▪
▪
Low socioeconomic status
Increased housing density
Lack of running water
Genetic susceptibility
Swimming in pools, rivers, streams
53
Chapter 65 Comma-shaped Rods
COMPLICATIONS
▪ Gastric carcinoma (1–2% risk): chronic
gastritis → atrophic gastritis, intestinal
metaplasia, carcinoma
▫ Chronic inflammation, ↑ TNF, ↑ IL-6, ↑
bacterial proteases → excessive tissue
damage, cell mutation → intestinal
metaplasia → carcinoma
▪ Gastric mucosa-associated lymphomas due
to persistent immune stimulation of gastric
lymphoid tissue
▫ Omeprazole/pantoprazole +
clarithromycin, amoxicillin: in case of
penicillin sensitivity, replace amoxicillin
with metronidazole
SIGNS & SYMPTOMS
▪ Majority of cases asymptomatic
▪ Acute infection
▫ Upper abdominal pain, nausea, loss of
appetite
▪ Chronic infection
▫ Chronic gastritis: upper abdominal pain,
nausea, bloating, vomiting/melena (black
stool)
▫ Peptic ulcers: stomach pain/ache; occurs
with empty stomach, between meals,
early morning
Figure 65.1 Helicobacter organisms in a
gastric pit.
DIAGNOSIS
LAB RESULTS
▪ Blood antibody test
▪ Stool antigen test
▪ Carbon urea breath test
▫ Individual ingests 14C- or 13C-labelled
urea, which bacterium metabolizes,
yielding labelled carbon dioxide
detectable in breath
▪ Urine enzyme-linked immunosorbent assay
(ELISA) test
TREATMENT
MEDICATIONS
▪ One-week “triple therapy”: antacid/acidreducing drugs (H2-receptor antagonists/
proton pump inhibitors) + two antibiotics
▫ Bismuth salicylate + metronidazole +
tetracycline
▫ Ranitidine bismuth citrate + tetracycline
+ clarithromycin/metronidazole
54
VIBRIO CHOLERAE (CHOLERA)
osms.it/vibrio-cholerae
PATHOLOGY & CAUSES
Characteristics
▪ Oxidase +; non-invasive; halophilic; genome
consists of two circular chromosomes;
sensitive to acid, drying
▪ Reservoir: aquatic environments (saltwater,
brackish)
▪ Fermentation: glucose, sucrose
Virulence factors
▪ Cholera toxin
▫ Only toxigenic strains; responsible
for pathogenesis of massive, watery
diarrhea; coded by filamentous
bacteriophage (CTXΦ)
▪ Other toxins that increase mucosal
permeability
▫ Zona occludens toxin (ZOT), accessory
cholera enterotoxin (ACE), WO7 toxin
17
▪ Lipopolysaccharide (LPS)
▫ > 200 serogroups; O1, O139 associated
with cholera epidemics
▪ Motility
▫ Single polar flagellum (H-antigen)
▪ Toxin-coregulated pilus (TCP)
▫ Present only in toxigenic strains;
promotes adherence, aggregation of
bacteria; coded by genes in Vibrio
pathogenicity island (VPI)
▪ Mucinase
▫ Digests mucous layer of gastrointestinal
(GI) tract
Culture
▪ Isolation specimen: stool, rectal swab
▪ Media: thiosulfate citrate bile salts sucrose
(TCBS) agar/taurocholate tellurite gelatin
agar (TTGA); large, yellow colonies (2–4mm
diameter) with opaque centers, translucent
edges
▪ V. cholerae: diverse species; pathogenic,
toxin-producing (toxigenic) variants cause
cholera
▪ Cholera characterized by profound
secretory diarrhea → rapid, life-threatening
dehydration
▪ Transmitted by fecal-oral route/
contaminated food or water
Pathogenesis
▪ Inoculation → passage through upper GI
tract → rapid movement of bacteria through
mucous via flagellum → colonization of
small intestine via TCP → releases cholera
toxin
▪ Cholera toxin (AB protein toxin) → B
subunit binds to GM1 ganglioside on
intestinal epithelial cell, allows entry of A
subunit → activates G-protein regulated
adenylyl cyclase → ↑ intracellular cyclic
adenosine monophosphate (AMP) →
secretion of chloride, sodium; inhibition
of sodium chloride absorption → massive
fluid secretion; loss of sodium, chloride,
bicarbonate, potassium
TYPES
Pathogenic (toxin-producing), nonpathogenic
Serological classification: O antigen
differences
▪ Serogroup O1 is subdivided into two
serotypes (Inaba, Ogawa), two biotypes
▫ El Tor: cause of current global pandemic
of cholera
▫ Classical: cause of previous V. cholerae
pandemics; now thought extinct
▪ Serogroup O139
▪ Non-O1/O139
RISK FACTORS
▪ Travel to endemic/epidemic areas
▪ Inadequate access to clean water
55
Chapter 65 Comma-shaped Rods
▪ Shellfish consumption in areas with
sporadic cholera
▪ People with blood group O at higher risk for
severe cholera (mechanism unknown)
COMPLICATIONS
▪ If untreated
▫ Dehydration, hypovolemic shock in 4–12
hours, death in 18 hours to several days
▪ Renal failure secondary to hypovolemia
▪ Electrolyte imbalances
▫ Hypokalemia, metabolic acidosis
▪ Pneumonia (esp. in children) due to
aspiration of vomit
SIGNS & SYMPTOMS
▪ Incubation period is 28–48 hours
▪ Asymptomatic to severe depending upon
strain, inoculum concentration (≥ 108 →
severe form)
▪ Abrupt onset of profound watery diarrhea
(grey, cloudy, flecked with mucus; “ricewater stool”); painless, without tenesmus;
loss of 1 liter of fluid per hour in severe
cases
▪ Moderate to severe vomiting, borborygmus,
abdominal discomfort
▪ Dehydration
▫ Thirst, dry mucous membranes,
decreased skin turgor, sunken eyes,
hypotension, weak/absent radial pulse,
tachycardia, tachypnea, hoarse voice,
oliguria
▪ Altered mental status
▫ Somnolence, restlessness, lethargy
▫ Culture on TCBS agar
▫ Dark field microscopy/dipstick test of
stool specimen for rapid confirmation in
non-endemic areas (detectable in stool
for 1–2 weeks without antimicrobial
therapy)
TREATMENT
MEDICATIONS
▪ Oral antibiotic treatment reduces duration,
severity of disease
▫ Doxycycline for adults
▫ Azithromycin for children, pregnant
individuals
OTHER INTERVENTIONS
▪ Prophylaxis
▫ WC-rBS (Dukoral) vaccine: monovalent
inactivated oral cholera vaccine
containing killed whole cells of V.
cholerae O1, additional recombinant
cholera toxin B subunit
▫ BivWC (Shanchol) vaccine: bivalent
inactivated oral vaccine containing killed
whole cells of V. cholerae O1, O139
▫ CVD 103-HgR/Vaxchora vaccine:
attenuated oral vaccine derived from
serogroup O1 classical Inaba strain
▪ Rapid, aggressive volume replacement
(oral/intravenous fluids)
▪ Adequate nutrition to prevent malnutrition
▪ Correction of electrolyte imbalances
▪ Zinc supplementation reduces duration,
severity of disease
DIAGNOSIS
LAB RESULTS
▪ Hypoglycemia/hyperglycemia
▪ Hypercalcemia, hypermagnesemia,
hyperphosphatemia
▪ ↑ hematocrit due to volume depletion
OTHER DIAGNOSTICS
▪ Clinical presentation
▪ Microbiologic diagnosis
56
NOTES
NOTES
CORONAVIRUSES
MICROBE OVERVIEW
▪ Causes respiratory infections
▪ OC43, NL63: most common strains of
human coronaviruses (HCoV)
▪ Associated clinical syndromes: common
cold, pneumonia, bronchitis, Middle East
respiratory syndrome (MERS-CoV), severe
acute respiratory syndrome (SARS-CoV)
Genetic material
▪ Positive-stranded RNA viruses
Taxonomy
▪ Corona: crown; named after spiked
appearance in electron microscopy
▪ Genera: alpha, beta (human pathogens),
gamma, delta
Morphology
▪ Helical capsid; enveloped (outer lipid
membrane)
▪ Structural proteins
▫ Small envelope protein (E) → viral
assembly
▫ Hemagglutinin esterase protein (HE)
→ binds to cell membrane (beta
coronaviruses only)
▫ Membrane protein (M) → viral assembly
▫ Nucleocapsid protein (N) → forms
nucleocapsid
▫ Spike protein (S) → binds, fuses with
host cell membrane
57
Chapter 66 Coronaviruses
CORONAVIRUS (SARS)
osms.it/coronavirus-SARS
PATHOLOGY & CAUSES
▪ Causes severe respiratory syndrome
(SARS)
▪ B-beta coronavirus: viral pulmonary
disease
▪ Viral inoculation of respiratory tract mucosa
→ cell damage → release of cytokines
(interferon-gamma, IL-1, IL-6, IL-12) →
inflammation, ↑ secretions
▪ Incubation period: 2–7 days
CAUSES
▪ Direct contact, airborne droplets, fomites
RISK FACTORS
▪ Immunosuppression, healthcare-related
occupation, comorbid conditions
COMPLICATIONS
▪ Acute respiratory distress syndrome,
multiorgan failure
SIGNS & SYMPTOMS
▪ Prodrome: fever, malaise, headaches,
myalgia, chills
▪ Non-productive cough, dyspnea, chest pain
▪ Diarrhea, rhinorrhea, sore throat
▪ Interstitial pulmonary infiltrates
LAB RESULTS
Histologic pulmonary tissue observation
▪ Hyaline membranes, edema, fibroblast
proliferation
Reverse-transcriptase polymerase chain
reaction (RT-PCR)
▪ Nasopharyngeal, oropharyngeal, stool,
serum samples
Serologic tests
▪ Enzyme-linked immunosorbent assays
(ELISA)
▪ Fluorescence antigen detection assays
Lab tests
▪ Lymphopenia, thrombocytopenia, ↑
lactate dehydrogenase (LDH), ↑ alanine
aminotransferase (ALT)
TREATMENT
OTHER INTERVENTIONS
▪ Mechanical ventilation in respiratory failure
Prevention
▪ Hand washing
▪ Proper disposal of infected materials (e.g.
used tissues)
▪ Mask use within healthcare environment
DIAGNOSIS
▪ Center for Disease Control and Prevention
(CDC)
▫ No alternative diagnosis after 72 hours
of clinical evaluation
▫ Individual at risk
DIAGNOSTIC IMAGING
Chest X-ray
58
NOTES
NOTES
CUTANEOUS FUNGAL INFECTIONS
GENERALLY, WHAT ARE THEY?
DIAGNOSIS
PATHOLOGY & CAUSES
▪ Noninvasive fungal infections of skin and its
annexes
▫ Limited to stratum corneum
▫ Caused by human skin’s commensal
flora
LAB RESULTS
▪ Microscopic observation
OTHER DIAGNOSTICS
▪ Clinical findings upon examination
SIGNS & SYMPTOMS
▪ Skin pigmentation changes
▪ Characteristic lesions: macule, patch, scale,
plaque
▪ Occasional pruritus
TREATMENT
MEDICATIONS
▪ Antifungal
MALASSEZIA
(TINEA VERSICOLOR &
SEBORRHOEIC DERMATITIS)
osms.it/malassezia
PATHOLOGY & CAUSES
▪ Genus of yeast-like fungi
▪ Cause cutaneous infections
▪ Cutaneous commensal flora, mostly lipiddependent (thrive on human sebum),
saprophytic (nutrients obtained from dead,
organic matter), dimorphic (yeast, hyphal/
mycelial forms)
TYPES
Tinea versicolor
▪ Superficial cutaneous mycosis
▪ AKA pityriasis versicolor
▪ Most common causes: M. globosa, M.
furfur, M. sympodialis
▪ Fungus produces azelaic acid → tyrosinase
activity (activated by sunlight) → skin
pigmentation changes → hypopigmented/
hyperpigmented macules, patches, plaques
59
Chapter 67 Cutaneous Fungal Infections
Seborrheic dermatitis
▪ Chronic, inflammatory dermatitis
▫ Tends to flare, relapse
▫ Likely caused by Malassezia spp.
▪ Fungus produces acids, enzymes, oxygen
radicals → cell damage → inflammatory
response → erythema, greasy, yellowish
scaling (range from mild, flaky to coarse,
thick lesions)
RISK FACTORS
Tinea versicolor
▪ Most common
▫ Adolescents/young adults
▪ Excessive heat, humidity, perspiration,
sunlight
▪ Immunosuppression
Figure 67.1 Tinea versicolor on the
abdomen.
Seborrheic dermatitis
▪ Biphasic occurrence
▫ Infants (cradle cap), adolescents/adults
▪ Biologically-male > biologically-female
individuals
▪ Comorbidities
▫ HIV/AIDS, Parkinson’s disease
SIGNS & SYMPTOMS
Tinea versicolor
▪ Characteristic skin changes
▫ Usually located on abundant sebaceous
gland areas (torso, proximal extremities,
face, neck)
▪ Light brown in light-skinned individuals;
dark brown to gray-black in dark-skinned
individuals
▪ Mild erythema, pruritus, scaling
▪ Lesions fail to tan with sun exposure
Seborrheic dermatitis
▪ Characteristic skin changes
▫ Usually located on trunk (“petaloid
pattern”), scalp (dandruff), eyebrows,
eyelids, nasolabial folds, external
auditory meatus, anogenital area
▪ Pruritus, erythema, blepharitis
▪ Tends to flare during stress, cold weather
▪ Infants: adherent yellowish scales primarily
on vertex of scalp
Figure 67.2 Seborrhoeic dermatitis
affecting the nasolabial folds.
DIAGNOSIS
LAB RESULTS
Tinea versicolor
▪ KOH preparation: microscopic observation
→ “spaghetti and meatballs”
▫ Spaghetti: hyphae
▫ Meatballs: yeast
60
OTHER DIAGNOSTICS
Tinea versicolor
▪ Wood’s lamp examination: yellow to
yellow-green fluorescence
Seborrheic dermatitis
▪ Clinical findings upon examination
Seborrheic dermatitis
▪ No known cure
▪ Chronic topical agent treatment: antifungal
medications, corticosteroids, calcineurin
inhibitors
▫ Other topical agents: selenium sulfide,
zinc pyrithione, salicylic acid or coal tar
(keratolytics)
▪ Oral antifungal agents if non-responsive
TREATMENT
MEDICATIONS
Tinea versicolor
▪ Topical agents: antifungal medications,
selenium sulfide, zinc pyrithione
▪ Oral antifungal medications if nonresponsive
61
Chapter 2 Acyanotic Defects
NOTES
DIPLOCOCCI: AEROBIC
MICROBE OVERVIEW
▪ Spherical-shaped bacteria (cocci), appear in
pairs as joined cells (diplo)
▪ Gram-negative, aerobes/facultative
anaerobes, non-motile, non-spore forming
TYPES
Moraxella catarrhalis
▪ Oxidase +, nitrate reduction +
(characteristic)
▪ Part of normal respiratory flora, causes
opportunistic infections
▪ Virulence factors
▫ Beta-lactamase production → penicillin
resistant
▫ DNase production
▪ Culture
▫ Isolation specimen: respiratory
secretions, sputum
▫ Media: blood, chocolate agar (round,
opaque colonies that turn pink after
48 hours; positive “hockey puck sign”:
able to slide colonies across agar with
wooden stick without disruption)
Neisseria gonorrhoeae
▪ Facultatively intracellular
▪ Oxidase +, catalase +
▪ Fermentation
▫ Glucose; differentiation from N.
meningitidis
▪ Virulence factors
▫ LOS/endotoxin: triggers inflammation;
undergoes antigenic variation
▫ IgA1 protease: cleaves IgA antibodies;
aids in evasion of humoral immune
response
▫ Type IV pili: promote adhesion of
bacteria to epithelium; undergo phase,
antigenic variation
▫ Porins (PorA, PorB): allow movement
of ions, nutrients into bacteria, promote
invasion into cells
▫ Opa, Opc: promote adhesion, invasion;
undergo phase, antigenic variation
▪ Culture
▫ Isolation specimen: urine, vaginal/
endocervical swab, urethral swab;
pharyngeal, rectal swab
▫ Media: Thayer–Martin VCN, chocolate
agar
Neisseria meningitidis
▪ Facultatively intracellular
▪ Oxidase +, catalase +
▪ Fermentation
▫ Maltose, glucose
▪ Present as normal non-pathogenic flora of
nasopharynx in 10% of adults
▪ Virulence factors
▫ Capsule: prevents phagocytosis;
N. meningitidis subdivided into
13 serogroups based on capsular
polysaccharides; A, B, C, W135, Y
account for most disease cases
▫ Lipooligosaccharide (LOS)/endotoxin:
released in blebs/vesicle-like structures
→ sepsis, vascular necrosis, hemorrhage
into surrounding tissue; levels of LOS
closely correlate with prognosis
▫ IgA1 protease: cleaves IgA antibodies;
aids in evasion of humoral immune
response
▫ Pili: promote adherence of bacteria to
nasopharyngeal epithelium; undergo
phase, antigenic variation → protect
against host immune response, vaccines
▫ Opacity proteins (Opa, Opc): promote
adhesion, invasion
62
▫ Factor H binding protein:
downregulates alternative complement
pathway
▪ Culture
▫ Isolation specimen: blood, cerebrospinal
fluid (CSF), petechial scrapings
▫ Media: Thayer–Martin vancomycin,
colistin, nystatin (VCN), chocolate agar
MORAXELLA CATARRHALIS
osms.it/moraxella-catarrhalis
PATHOLOGY & CAUSES
▪ Gram-negative diplococcus → respiratory
tract infections, otitis media
▪ Infections caused by M. catarrhalis
▫ Respiratory tract infections
(bronchitis, rhinosinusitis, laryngitis,
bronchopneumonia, communityacquired bacterial pneumonia)
▫ Otitis media in children < three years of
age
▫ Exacerbations of chronic obstructive
pulmonary disease (COPD)
RISK FACTORS
▪ Immunocompromised individuals
▪ Individuals with chronic respiratory disease
(e.g. COPD, emphysema)
▪ Children < two years of age, elderly
COMPLICATIONS
▪ Rare: bacteremia, septicemia, urethritis,
septic arthritis
SIGNS & SYMPTOMS
▪ Acute bacterial rhinosinusitis: fever, nasal
obstruction, purulent nasal discharge, facial
pain, headache
▪ Otitis media: fever, ear pain, bulging
tympanic membrane
▪ Exacerbations of COPD: increased cough,
sputum production/change in color,
dyspnea
63
Chapter 68 Diplococci: Aerobic
DIAGNOSIS
OTHER DIAGNOSTICS
▪ Clinical presentation
▫ Sufficient for diagnosis
▪ Microbiologic diagnosis
TREATMENT
MEDICATIONS
▪
▪
▪
▪
Amoxicillin-clavulanate
Trimethoprim-sulfamethoxazole
Third-/second-generation cephalosporins
Macrolides (e.g. azithromycin,
clarithromycin)
NEISSERIA GONORRHOEAE
osms.it/neisseria-gonorrhoeae
PATHOLOGY & CAUSES
▪ Gram-negative diplococcus → gonococcal
disease (gonorrhea, disseminated
gonococcemia, gonococcal ophthalmia
neonatorum)
▪ Portal of entry
▫ Unprotected sex (vaginal, oral, anal):
bacteria attaches, invades genitourinary,
rectal, oral epithelium via Opa, Opc, pili
→ gonorrhea
▫ Rare: invades bloodstream →
disseminated gonococcemia, septic
arthritis
▫ Perinatal transmission: birth canal
of infected mother → gonococcal
ophthalmia neonatorum
TYPES
▪ Gonorrhea
▫ Urethritis, cervicitis, proctitis,
pharyngitis
▪ Disseminated gonococcemia
▫ Result of spread, intravascular
multiplication of N. gonorrhoeae; joints,
skin (dermatitis-arthritis syndrome)
▪ Gonococcal ophthalmia neonatorum
▫ Causes gonococcal conjunctivitis
RISK FACTORS
▪ Unprotected sex
▫ Individuals with multiple sexual
partners, sex between individuals who
are biologically male (MSM), recent new
sexual partner
▪ Low educational, socioeconomic levels
▪ Substance abuse
▪ History of gonorrhea
COMPLICATIONS
Gonorrhea
▪ Epididymitis, prostatitis, penile
lymphangitis, urethral strictures in
individuals who are biologically male;
cervical gonorrhea → pelvic inflammatory
disease → infertility in individuals who are
biologically female
Gonococcal ophthalmia neonatorum
▪ Corneal scarring/perforation, blindness
SIGNS & SYMPTOMS
Gonorrhea
▪ Some individuals who are biologically
male, most individuals who are biologically
female (50–80%) asymptomatic
▪ Urethritis: dysuria, urinary urgency,
purulent foul-smelling urethral discharge
▪ Cervicitis: lower abdominal discomfort,
dyspareunia (pain during sexual
intercourse), vaginal pruritus, purulent foulsmelling vaginal discharge
64
AfraTafreeh.com exclusive
▪ Proctitis: anal pruritus, tenesmus, rectal
fullness, constipation, purulent anorectal
discharge, bleeding
▪ Pharyngitis: sore throat, swollen lymph
nodes
Disseminated gonococcemia
▪ Fever, chills, generalized malaise
▪ Polyarthralgia (multiple joint pain)
▪ Tenosynovitis (tendon inflammation)
▪ Pustular/vesiculopustular lesions on skin
Gonococcal ophthalmia neonatorum
▪ Purulent conjunctival discharge
▪ Swollen eyelids
▪ Conjunctival hyperemia, chemosis
DIAGNOSIS
LAB RESULTS
Blood tests
▪ ≥ two white blood cells in urethral
secretions
▪ ≥ 10 white blood cells on microscopic
examination of first void urine
▪ Disseminated gonococcemia
▫ Positive blood culture
▫ Synovial fluid leukocyte count: increased
values (50,000 cells/microL)
Gram stain
▪ Polymorphonuclear leukocytes with
intracellular gram-negative diplococci
Nucleic acid amplification testing (NAAT)
▪ For initial diagnosis
TREATMENT
MEDICATIONS
Figure 68.1 A neonate with gonococcal
ophthalmia neonatorum.
▪ Uncomplicated gonorrhea
▫ Intramuscular injections of ceftriaxone
+ azithromycin/doxycycline (in
case of gonococcal resistance to
cephalosporins, potential chlamydia
regardless of chlamydial coinfection
status)
OTHER INTERVENTIONS
▪ Prophylaxis
▫ No vaccines
▫ Extensive antigenic variations of
bacterial components (pili, LOS, opa
proteins) prevents development of
immunological memory
Figure 68.2 Creamy discharge emanating
from the external urethral meatus is typical of
genital gonorrhea infection.
65
Chapter 68 Diplococci: Aerobic
NEISSERIA MENINGITIDIS
osms.it/neisseria-meningitidis
PATHOLOGY & CAUSES
SIGNS & SYMPTOMS
▪ Gram-negative diplococcus →
meningococcal disease (meningitis,
meningococcemia)
▪ Portal of entry
▫ Inhalation of respiratory droplets
→ bacteria attaches to respiratory
epithelium via Opa, Opc, pili →
nasopharynx colonization → usually
resolves asymptomatically (carriers)
▫ Invades bloodstream → meningococcal
disease (rare)
Meningococcemia
▪ Petechial rash caused by destruction
of blood vessels, hemorrhage due to
endotoxin release; fever, chills; joint, muscle
pain
RISK FACTORS
Meningitis
▪ Infants
▫ Early nonspecific: irritability, vomiting,
inactivity, poor feeding, temperature
instability
▫ Late specific: bulging anterior fontanelle,
seizures
▪ Children, adolescents, adults
▫ Early nonspecific: sudden onset of fever,
headache, nausea, vomiting, myalgia
▫ Late specific: altered mental status,
lethargy, neck stiffness (nuchal rigidity),
photophobia
▫ First specific symptoms of sepsis:
abnormal skin color (pallor/mottling)
which can evolve from nonspecific rash
to petechial to hemorrhagic over several
hours; cold hands, feet; leg pain
▪ Signs upon physical examination
▫ Positive Kernig’s sign: inability to
straighten leg when hip flexed to 90º
▫ Positive Brudzinski’s sign: flexing of
neck by examiner → flexing of hips,
knees
COMPLICATIONS
Fulminant meningococcemia
▪ Abrupt onset
▪ Rapid enlargement of petechiae/
ecchymoses
▪ Hypotension, tachycardia due to vascular
collapse, shock
TYPES
▪ Meningococcemia
▫ Result of intravascular multiplication
of N.meningitidis; can occur alone/in
conjunction with meningitis
▪ Meningitis
▫ Most common; occurs upon spreading
of bacteria to meninges during
meningococcemia; usually affects
children, adolescents
▪ Fulminant meningococcemia
▫ AKA Waterhouse–Friderichsen
syndrome; most severe form of
meningococcal sepsis; massive bilateral
hemorrhage into adrenal glands
▪ Infants 6–24 months (due to immature
immune system, inability to vaccinate)
▪ Living in close quarters (military barracks,
dormitories)
▪ Adrenal insufficiency; disseminated
intravascular coagulation (DIC); purpura
fulminans (cutaneous hemorrhage, necrosis
due to vascular thrombosis, DIC); acute
respiratory distress syndrome (ARDS);
coma, death
66
DIAGNOSIS
LAB RESULTS
▪ Fulminant meningococcemia
▫ Adrenal insufficiency signs: ↓ blood
glucose, ↑ K+, ↓ Na+, adrenocorticotropic
hormone (ACTH) stimulation test (low
response)
▫ Thrombocytopenia due to DIC
▫ Metabolic acidosis
▪ Meningococcemia and meningitis
▫ Blood culture and CSF analysis
OTHER DIAGNOSTICS
▪ Physical examination
▫ Characteristic findings of meningitis
TREATMENT
MEDICATIONS
Prophylaxis
▪ Quadrivalent immunization with purified
capsular polysaccharides from serogroups
A, C, Y, W135 (group B not available)
Figure 68.3 A petechial rash can be seen in
the late stages of meningococcal septicemia.
Meningococcal disease
▪ Antibiotics
▫ Third-generation cephalosporins (e.g.
cefotaxime, ceftriaxone)/penicillin G
▪ Chloramphenicol
▫ In case of beta-lactam antibiotics
hypersensitivity
▪ Chemoprophylaxis of individuals in close
contact with the infected
▫ Rifampin of ciprofloxacin
▪ Hydrocortisone
▫ For adrenal insufficiency
SURGERY
▪ Plastic surgery, skin grafting, amputation to
treat tissue necrosis
Figure 68.4 The brain of an individual at
post mortem following death from bacterial
meningitis. Nesseiria meningitidis is the
most common causative organism amongst
adolescents and young adults.
67
NOTES
NOTES
ECTOPARASITES
GENERALLY, WHAT ARE THEY?
PATHOLOGY & CAUSES
▪ Ectoparasites: arthropods that live on
outside of host, extract nutrients at host’s
expense
▫ Uncommon: viable parasites transferred
without direct host contact
RISK FACTORS
DIAGNOSIS
OTHER DIAGNOSTICS
History
▪ Including close contacts/living quarters
Physical examination
▪ Dermatologic examination
▪ Commonly poor hygiene, close living
quarters
TREATMENT
MEDICATIONS
COMPLICATIONS
▪ Predicated on individual’s immune status,
housing situation
▪ Topical neurotoxins, topical/oral parasidal
drugs
OTHER INTERVENTIONS
SIGNS & SYMPTOMS
▪ Pruritus, bite marks, visible body parasites
▪ Proper hygiene, household measures,
isolation (if necessary)
PEDICULOSIS CORPORIS, CAPITIS,
AND PUBIS (LICE)
osms.it/lice
PATHOLOGY & CAUSES
▪ Infestation of easily transmissible sucking
lice species
▫ Commonly in hairy bodily areas,
characterized by local pruritus
▪ Sucking lice infection
▫ (Phylum) arthropoda → (class) insecta
→ (order) phthiraptera → (suborder)
anoplura → (family) pediculidae/
pthiridae
PATHOLOGY
▪ Lice live human hair → suck blood for
nutrients
▫ Bite → saliva injection →
anticoagulation effect, ↑ histamine
release → maculae cerulea (blue/copperhued bite marks); pruritus
68
▪ Lice require humans for nutritional source
(parasites)
▫ Most climates allow mature louse 24
hours of viable life away from human
source
Transmission
▪ Physical contact
▫ Ideal location: slow-moving, parallel hair
fibers
▫ Louse on one hair fiber → transfer to
another individual’s hair → lay nits
(eggs) on hair 1–2mm off of scalp →
nymphs hatch within one week →
mature over one week → female lice lay
eggs for one month
▪ Fomites
TYPES
▪ Pediculus humanus capitis → head louse
▫ Can survive 24–48 hours without blood
meal/separated from host
▪ Pediculus humanus humanus → body louse
▫ Larger than head louse; can survive <
72 hours without blood meal
▪ Phthirus pubis → pubic louse
▫ AKA crab louse; can spread to body
RISK FACTORS
▪ School-aged children
▪ Homeless population
▪ Refugee population (if living in close
quarters)
COMPLICATIONS
▪ Co-infections (also carried by louse)
▫ Bartonella quintana → endocarditis
▫ Epidemic typhus
▫ Louse-borne relapsing fever
▫ Trench fever
▪ Adolescents with pubic lice → ↑ gonorrhea/
chlamydial infection risk
▪ Pruritus → skin excoriation → secondary
infection
▫ Commonly staphylococcal infection
SIGNS & SYMPTOMS
▪ Site pruritis (head, body, pubis)
DIAGNOSIS
OTHER DIAGNOSTICS
Dermatologic examination
▪ Examination of hair follicles, clothing seams
▪ Nits: more visible than nymphs/lice; most
visible (white) after louse released from
egg; does not dislodge easily from hair
follicle
▪ Body louse → widespread dermatitis
▪ Often linear excoriations
▪ Maculae ceruleae → may have punctal
hemorrhages → recent bites
▪ Hyperpigmentation/lichenification → older
bites
Lymph node examination
▪ Capitis infection → posterior
lymphadenopathy
TREATMENT
MEDICATIONS
Topical benzyl alcohol
▪ Mechanism of action → louse asphyxiation
▫ Difficult for resistance to develop
Neurotoxic agents
▪ Resistance develops with
▫ Pyrethrin: botanically-derived
neurotoxin
▫ Permethrin: Na+ channel blocker →
paralysis → death
▫ Malathion: organophosphate
cholinesterase inhibitor
OTHER INTERVENTIONS
Hair shaving
▪ Eradicate current infection
Mechanical removal
▪ Wet combing → tedious, poor compliance
▫ Pre-treatment: vinegar/formic acid →
flattened hair cuticle → better combing
efficiency (does not dissolve/loosen nits)
Prevention
▪ Proper hygiene
69
Chapter 69 Ectoparasites
▪ Household
▫ Housemates: examination
▫ Bedmates: prophylactic treatment
▫ Household cleaning: washing >
54°C/130°F; unwashable material →
place in sealed plastic bag for two
weeks
▪ School
▫ No nit policy (infected children stay
home), education, screening during
outbreaks
SARCOPTES SCABIEI (SCABIES)
osms.it/sarcoptes-scabiei
PATHOLOGY & CAUSES
▪ Sarcoptes scabiei mite infection
▫ Elicits strong immune response
▫ Nocturnal pruritus
Mite infection
▪ (Phylum) mite → (class) arachnida →
(subclass) acari → (order) astigmata →
(family) sarcoptidae
▫ Usually obligate human parasite → vars
hominis
▫ Sometimes animal mange mites can
infest
PATHOLOGY
Mite transfer
▪ Direct skin-to-skin contact for 15–20
minutes
▪ Average infested individual carries 5–12
mites
▫ Crusted scabies individuals: > 1000
mites can be shed (transmission through
objects more likely)
Type IV hypersensitivity reaction
▪ House dust mite cross reactivity
▪ Infestation → ↑ IL-6, vascular epithelial
growth factor (VEGF) →TH1-cell activation
→ IL-2 release → lymphocyte proliferation,
differentiation
RISK FACTORS
▪ Overcrowding (including long-term care
facilities, prisons), poor hygiene/nutrition,
homelessness, dementia, sexual contact
COMPLICATIONS
▪ Infestation → secondary staphylococcal
infection
▫ Low-income countries (mostly)
▫ Impetigo → chronic kidney disease
▫ Ecthyma, paronychia, furunculosis
Crusted scabies
▪ AKA Norwegian scabies
▪ Infection commonly scalp, hands, feet →
diffuse spread over entire body
▪ Occurs in compromised cellular immunity
setting
▫ Acquired immunodeficiency syndrome
(AIDS)
▫ Human lymphocytic virus type 1 (HTLV1)
▫ Leprosy
▫ Lymphoma
▫ Long-term topical corticosteroid use
▪ Risk factors: age, Down syndrome
▪ Complications: fissional lesions develop →
bacterial entryway → infection
▫ Sepsis, poststreptococcal
glomerulonephritis
70
SIGNS & SYMPTOMS
Classic scabies
▪ Intense, intractable, generalized pruritus
▪ Nodules, pustules at most intense pruritus
sites
▪ Common areas → intertriginous spaces
▫ Anterior axillary folds, webs of fingers,
volar aspect of hand/wrist, beltline,
penis, areolar region (biologically-female
individuals)
Dermatologic examination
▪ Serpiginous keratotic lines (1–4mm) →
burrow marks
▫ Often with vesicle on end (housing mite)
Nodular scabies
▪ Hypersensitivity reactions → large,
persistent, intensely pruritic 5–6mm
nodules
▫ Commonly groin, buttock, axillary folds
Crusted scabies
▪ Poorly defined, erythematous patches →
scale
▫ Untreated → entire integumental spread
→ warty appearance (especially over
bony prominences); lesions crust, fissure
develop → malodorous; nail involvement
→ thickened, dystrophic, discolored
DIAGNOSIS
LAB RESULTS
Microscopy
▪ Confirmatory scraping: fluorescein stain →
highlights fecal material, ova fragments
▫ Epithelial milieu (eosinophils,
lymphocytes, histiocytes)
▫ Crusted scabies: mate capture more
likely due to disease burden
Polymerase chain reaction assays
▪ S. scabiei DNA polymerase
OTHER DIAGNOSTICS
History
▪ Close contact commonly present with
concurrent symptoms
▪ Infected individual contact history (may be
many weeks prior)
Figure 69.1 A high-magnification photograph
of a single mite burrow in the skin of an
individual with scabies. The mite is at the end
of the burrow at the top right of the image.
TREATMENT
MEDICATIONS
Classic scabies
▪ Permethrin (5%) → synthetic neurotoxin →
Na+ channel blocker → paralysis → death
▪ Precipitated sulfur (6%, 10%) in petroleum
▪ Benzyl benzoate (10%, 25%)
▫ Adverse reactions: allergic dermatitis
▫ Contraindications: pregnancy/lactation
(neurotoxicity); children < two years old
▪ Oral ivermectin
▫ One dose (200mcg/kg) repeated in
7–10 days
Nodular scabies
▪ Topical steroids
▪ Intralesional steroid injection
Crusted scabies
▪ Topical, systemic treatment required
▪ Oral ivermectin, topical permethrin (5%)/
benzyl benzoate (5%)
▫ Two week oral regimen, topical therapy
71
Chapter 69 Ectoparasites
persisting after that twice weekly until
cure
▫ Treatment cure → active lesion
resolution, nocturnal pruritus absence
for one week
OTHER INTERVENTIONS
▪ Isolation for infected
▪ Nail clipping
▫ +/- brushing with scabicidal agent
▪ Thorough personal, household material
laundering
Prevention
▪ Monosulfiram soap in communities with ↑ ↑
incidence
72
NOTES
NOTES
ENTEROCOCCUS
ENTEROCOCCUS
osms.it/enterococcus
PATHOLOGY & CAUSES
▪ Gram-positive spherical-shaped bacteria
(cocci)
▪ Grow in pairs (diplococci)/short chains
▪ Non-spore forming
▪ Optimal growing conditions: 45ºC/113°F,
can withstand up to 60ºC/140°F
▫ Facultatively anaerobic bacteria
▫ 6.5% NaCl, bile-containing media
▪ Characteristics
▫ Catalase negative, pyrrolidonyl
arylamidase (PYR) positive
▫ Variable hemolytic activity on blood
agar plates; most commonly gamma
hemolytic
▪ Common human pathogens: E. faecalis, E.
faecium
▪ Important cause of hospital-acquired
infections, esp. infective endocarditis,
urinary tract infections (UTIs), infections
of prosthetic devices (e.g. venus/urinary
catheters)
▫ Surface carbohydrates → adherence to
cardiac valves → fibrinogen synthesis
→ valve vegetations → infective
endocarditis
▫ Adherence to renal epithelial cells → UTI
▪ Less common associations
▫ Wound, bloodstream, biliary tract, pelvic
infections; intraabdominal abscesses
RISK FACTORS
▪ Indwelling medical devices (e.g. central
venous, urinary catheterization)
▪ Cardiovascular abnormalities
▪ Prolonged hospitalization, mechanical
ventilation
▪ Prior antibiotic use, immunodeficiency
COMPLICATIONS
▪ Bacteremia
▫ Immunocompromised individuals
SIGNS & SYMPTOMS
▪ UTIs
▫ Cystitis, pyelonephritis; dysuria,
frequency, urgency, suprapubic/flank
tenderness
▪ Infective endocarditis
▫ Subacute onset of fever, malaise,
peripheral signs (e.g. Janeway lesions,
Osler’s nodes), cardiac murmurs (usually
left-sided), splenomegaly
▪ Infection of prosthetic devices, wounds
▫ Erythema, swelling, tenderness, warmth
DIAGNOSIS
DIAGNOSTIC IMAGING
Transthoracic echocardiography; abdominal CT scan; ultrasound
▪ Identify organ involvement
LAB RESULTS
▪ Cultures from infected sites (e.g. blood,
urine)
▪ Susceptibility testing
▫ Detect antibiotic resistance
73
Chapter 70 Enterococcus
▪ Complete blood count (CBC)
▫ ↑ polymorphonuclear cells
▪ Urinalysis
▫ UTI → pyuria, proteinuria, hematuria
TREATMENT
MEDICATIONS
Antimicrobial therapy
▪ Localized infections
▫ Ampicillin/beta lactamase inhibitors
(clavulanate/sulbactam)
▫ Alternative: nitrofurantoin
▪ Serious infections with bacteremia,
meningitis, endocarditis
▫ Penicillin/ampicillin + aminoglycoside
▫ Alternative: vancomycin,
aminoglycoside
▪ Vancomycin-resistant enterococcal (VRE)
infection
▫ Linezolid/daptomycin
SURGERY
▪ Drain abscess
▪ Valve replacement
OTHER INTERVENTIONS
▪ Remove venous/urinary catheters
74
NOTES
NOTES
FILAMENTS
GENERALLY, WHAT ARE THEY?
PATHOLOGY & CAUSES
▪ Gram ⊕ slender bacteria with ⊕ branches
(atypical lung disease organisms, capable
of affecting any body organ
▪ Atypical organisms → indolent disease →
insidious growth → severe disease
RISK FACTORS
▪ Immunodeficiency
▪ Corticosteroid use → iatrogenic
immunosuppression
SIGNS & SYMPTOMS
▪ Cough, dyspnea
▫ Indolent course
▫ Common in fever’s absence
▪ Other symptoms
▫ Dependent on organ systems affected
by organism
LAB RESULTS
▪ Tissue biopsy → histological
OTHER DIAGNOSTICS
Physical examination
▪ Pulmonary examination
▫ Auscultation: rhonchi (crackles), ↓ breath
sounds
▫ Palpation: ↓ tactile fremitus
▫ Percussive dullness
TREATMENT
MEDICATIONS
▪ Antibiotics
SURGERY
▪ Resection
▫ Medication non-responsive
▫ Large infections → significant
dysfunction
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Localized alveolar infiltrate
▫ Homogeneous, non-segmental, cavitary
appearance
75
Chapter 71 Filaments
ACTINOMYCES ISRAELII
osms.it/actinomyces-israelii
PATHOLOGY & CAUSES
Microbe characteristics
▪ ⊕ Gram stain
▪ Shape
▫ Filamentous, non-spore-forming,
pleomorphic bacilli
▪ Metabolism
▫ Catalase negative, anaerobic/
microanaerobic bacilli
▪ Types
▫ 21 species found in humans
▫ Actinomyces israelii most common
▪ Locations
▫ Normal mouth (by two years old),
gastrointestinal (GI) tract, female
genitourinary tract flora
PATHOLOGY
Thoracic
▪ Pulmonary → pneumonia
▫ Oropharyngeal content aspiration →
bacterial alveoli seeding → immune
response, bacterial growth →
pneumonia
Abdominal & pelvic
▪ Gastrointestinal → appendicitis
▫ Preceding colonic mucosa perforation →
unrecognized → months–year course →
symptomatic infection
▪ Pelvic → female genitourinary infections
▫ Complicated abortions, infected
intrauterine devices (IUDs), endometritis,
tubo-ovarian abscess (TOA)
RISK FACTORS
▪ Chronic granulomatous disease
Cervicofacial
▪ Chronic tonsillitis, dental decay, periodontal
disease, mastoiditis, otitis media
▪ Uncommon infection source
▪ Mucosal membrane violated → indolent,
invasive disease
▫ Commonly co-occurs with another
pathogen → micro-O2 Actinomyces
environment
▫ Can burrow through soft tissue, bone →
small abscesses, drainage tracts
▫ Abscesses: yellow sulfur-containing
granules in granulomatous reactive
material setting (bacteria found in
microfilament tangles, surrounded by
neutrophils)
COMPLICATIONS
TYPES
Thoracic
▪ Pneumonitis
Cervicofacial
▪ Osteomyelitis of mandible/maxilla
▫ Resident flora in periodontal pockets,
carious teeth, dental plaque, tonsillar
crypts
Abdominal & pelvic
▪ Gastrointestinal
▫ Peritoneal, hepatic, pelvic infectious
spread
Thoracic
▪ Aspiration history
Cervicofacial
▪ Deep neck tissue infection →
retropharyngeal space → mediastinitis
▪ Meningitis → if sinus tracts to posterior
neck, spinal cord
76
SIGNS & SYMPTOMS
Cervicofacial
▪ Lumpy jaw
▫ Usually in fever’s/other infectious signs’
absence
▪ Progression → oral mucosa, trismus sinus
tract draining
Thoracic
▪ Fever, cough > three day duration
▪ Auscultation
▫ Rhonchi
▫ ↓ breath sounds
▪ Palpation
▫ ↓ tactile fremitus
▪ Percussive dullness
Abdominal & pelvic
▪ Gastrointestinal → appendicitis
▫ Asymptomatic colonic mucosa (micro)
perforation → months–years prodrome
→ symptomatic appendicitis
▫ Nonspecific prodrome: chronic fever,
weight loss, diarrhea, constipation, night
sweats
▫ Appendicitis: nausea, vomiting, anorexia
▪ Pelvic → female genitourinary infections
▫ Painful abdominal, cervical examination
▫ Purulent vaginal discharge
CT scan
▪ Abdominal, pelvic
▫ Disrupted tissue planes
Colonoscopy
▪ Abdominal, pelvic
▫ Normal/thickened mucosal appearance,
colitis, ulceration, nodular lesion, buttonlike appendiceal orifice elevation
Bedside ultrasound
▪ Abdominal, pelvic
▫ TOA evaluation
LAB RESULTS
▪ Cervicofacial
▫ Monoclonal antibody staining
▫ Polymerase chain reaction (PCR) of 16S
rRNA
Cultures (needle aspiration)
▪ Cervicofacial
▫ Histology: granulation tissues with
neutrophils, foamy macrophages,
lymphocytes, plasma cells (with
surrounding fibrosis)
▪ Abdominal, pelvic
▫ Histology: granulation tissue
surrounding oval, eosinophilic zones
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Thoracic
▫ Localized alveolar infiltrate:
homogeneous; non-segmental, cavitary
appearance; can extend past fissure
lines → into chest wall
Barium enema
▪ Abdominal, pelvic
▫ Luminal narrowing, fistualization,
extrinsic compression
Figure 71.1 Sulphur granules formed by
Actinomyces organisms.
77
Chapter 71 Filaments
OTHER DIAGNOSTICS
History
▪ Thoracic
▫ May have community-acquired
pneumonia diagnosis, treatment
(without relief within 3–5 days)
Physical examination
▪ Cervicofacial
▫ Lymphatic examination
▪ Thoracic
▫ Pulmonary examination
TREATMENT
MEDICATIONS
▪ Antibiotics
▫ Prolonged (weeks–months) penicillin V
(oral)/penicillin G (intravenous)
▫ Amoxicillin (alternative)
SURGERY
▪ Necrotic disease/especially large abscess
formation cases
NOCARDIA
osms.it/nocardia
PATHOLOGY & CAUSES
▪ Microbe characteristics
▪ ⊕ Gram stain
▪ Shape
▫ Filamentous, branch-forming, bacillus
▫ Branches → beaded appearance
(delicate nature of stain → cocci/bacilli
fragmentation)
▪ Metabolism
▫ Aerobic, catalase ⊕, urease ⊕
▪ Types
▫ > 80 species
▫ Around 30 disease-causing in humans
▪ Locations
▫ Saprophyte (organic pathogen) → found
in soil, house dust, water (fresh/salt),
bathing pools
PATHOLOGY
▪ Direct tissue inoculation
▫ Saprophyte → aerosolization common
(↑ ↑ pulmonary infections)
▫ Soil/water contamination (contaminated
food → GI disease, skin trauma →
cutaneous disease, eye trauma → ocular
disease)
▪ Facultative intracellular organism →
requires innate host defense mechanisms
▫ Inhalation entry: deficient/ineffective
mucociliary clearance, host response →
bronchopulmonary disease
▫ Skin trauma entry: deficient keratinized
skin barrier → local subcutaneous
infection; deficient keratinized cornea →
ocular infection
▪ Rapid filamentous growth → ↓ phagocytic
clearing
▫ Phagocytosed → ↓ lysosomal
destruction (phagocyte-lysosome
fusion inhibition; catalase, dismutase
production → ↓ reactive oxygen species
→ pathogen survival)
TYPES
Pneumonia
▪ Nocardia asteroides (common pathogen)
▪ > 2⁄3 of total disease
▪ Progression
▫ Empyema, pericardial effusion
Primary cutaneous infections
▪ Nocardia brasiliensis (common pathogen)
▪ Cellulitis, ulcers, pyoderma, myocetma
78
▪ Progression
▫ Abscess/nodular development,
lymphangitis
RISK FACTORS
▪ ↓ mucociliary clearance
▫ Cystic fibrosis, asthma, bronchiectasis
▪ Immunosuppression
▫ Iatrogenic (most commonly
corticosteroid use); lymphoreticular
malignancy; chronic obstructive
pulmonary disease; chronic
granulomatous disease;
dysgammaglobulinemia; HIV infection;
bone marrow, organ transplant
COMPLICATIONS
▪ Organ dissemination
▫ Pulmonary infection → hematogenous
spread (commonly)
▫ Most common: central nervous system
(meningitis, cerebral abscess)
▪ Pulmonary
▫ Empyema, pericardial effusion
▪ Cutaneous
▫ Lymphangitis
▪ Ocular
▫ Endophthalmitis (ocular infection)
SIGNS & SYMPTOMS
Pneumonia
▪ Acute, subacute, chronic suppurative
course
▫ Symptoms may also relapse/remit
▪ Cough, dyspnea common
▫ Anorexia, weight-loss (uncommon)
▫ Hemoptysis (cavitary disease)
▪ Rhonchi (crackles)
▪ ↓ breath sounds, tactile fremitus, +/egophony
Local cutaneous infection
▪ Local erythema, warmth, +/- ulceration,
nodular growth
Neurologic infection
▪ Meningismus, fever, rigors, seizure
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Pulmonary
▫ Homogeneous, non-segmental, cavitary
alveolar infiltrate
Brain MRI
▪ Neurologic
▫ ↑ T1 imaging intensity → ↑
enhancement (gadolinium)
LAB RESULTS
▪ Tissue biopsy: histology (acid fast stain)
▫ Gram ⊕, branching, beaded filamentous
growth
TREATMENT
MEDICATIONS
Antibiotic monotherapy
▪ Mild/moderate disease
▪ 3–6 months treatment duration
▪ Sulfonamides → trimethoprimsulfamethoxazole (TMP-SMX)
▪ Linezolid
▫ Nocardia 100% sensitive
▫ Limited treatment duration (2–3 weeks)
→ ineffective monotherapy for complete
therapy duration
Antibiotic multi-agent therapy
▪ Severe disease
▪ Up to 6–12 months treatment duration
▪ Agents
▫ TMP-SMX + amikacin/carbapenem/
linezolid
▪ Commonly for progressive disease in
immunosuppressed individuals/pulmonary,
disseminated disease
Prevention & vaccine
▪ Daily, full-strength TMP-SMX → secondary
prophylaxis
▪ P. jirovecii TMP-SMX prophylaxis (3x/week)
→ ineffective
79
Chapter 71 Filaments
SURGERY
▪ Indicated for
▫ Antibiotic-resistant, large cutaneous/
cerebral abscess (craniotomy/aspiration
effective)
▫ Empyemas, large fluid collections
▫ Pulmonary nocardiosis → pericarditis
(fatal if not performed)
80
NOTES
NOTES
FILOVIRUSES
MICROBE OVERVIEW
Genetic material
▪ Single-stranded negative-sense RNA virus
family; causes viral hemorrhagic fever
Taxonomy
▪ Genera: Ebolavirus, Marburgvirus,
Cuevavirus
Morphology
▪ Enveloped virions; filamentous,
nonsegmented morphology
Replication
▪ Transcription, replication mediated by
virus-encoded polymerase in infected cell
cytoplasm
▫ Transcription: negative-sense RNA
genome transcribed into monocistronic,
polyadenylated RNA species using host
cell ribosomes, tRNA etc. → translated
into seven proteins
▫ Replication: positive-sense antigenome
serves as template for negative-sense
genomes
Transmission
▪ Zoonotic infection
▫ Natural host: unknown (bats, especially
fruit bats considered infection source)
▫ Intermediate host: Often nonhuman
primates (gorillas, chimpanzees)
Structural proteins
▪ Viral RNA encodes seven structural
proteins
▫ Nucleoprotein
▫ Polymerase cofactor (VP35)
▫ Viral proteins VP40, VP24
▫ Glycoprotein: projecting spikes from
lipid bilayer (attachment to host cell
receptors)
▫ Transcription activator
▫ RNA-dependent RNA polymerase
EBOLA VIRUS
osms.it/ebola-virus
PATHOLOGY & CAUSES
▪ Ebola virus: causative agent of severe,
often fatal hemorrhagic fever
▪ Species: Zaire, Sudan, Tai Forest,
Bundibugyo, Reston
▪ Transmission
▫ Animal-human: direct infected-animal
tissue, body-fluid contact; butchering,
consuming undercooked meat
▫ Human-human: direct tissue, body fluid
contact with ill/deceased
▫ Potential transmission: contaminated
surface/object contact
PATHOLOGY
▪ Inoculation → incubation: 6–12 days
average (ranges 2–21 days)
▫ Host cell attachment, virus endocytosis
→ nucleocapsid release in cytoplasm →
81
Chapter 72 Filoviruses
▪
▪
▪
▪
replication → nucleocapsid formation →
viral shedding, cell necrosis
Initially macrophages, dendritic cells
infected → sentinel lymph node spread →
bloodstream → many cell types infected
(endothelial cells, fibroblasts, hepatocytes,
epithelial cells, adrenal-gland cells) with
lymphocyte/neuron exception
▫ Although uninfected, inflammatory
mediators; support signal loss from
dendritic cells → “bystander” apoptosis
of lymphocytes
Multifocal necrosis in various tissues (e.g.
liver, spleen)
Systemic inflammatory syndrome:
proinflammatory mediator, cytokine release
from infected macrophages, dendritic cells,
necrotic cell breakdown products, etc. →
vasodilation, ↑ vascular permeability →
vascular leakage, shock, multiorgan failure
Dendritic cell dysfunction, “bystander”
lymphocyte apoptosis → impaired adaptive
immunity
▪ Spontaneous abortion, vaginal bleeding in
infected pregnant individuals (100% third
trimester maternal, fetal mortality)
▪ Fatality ranges
▫ 40% to 80–90% (depending upon
species)
RISK FACTORS
▪ Healthcare workers without appropriate
protective equipment, adequate training
▪ Inadequate infected waste product, corpse
handling
▪ Sexual intercourse with person recovering
from Ebola in previous three months
▪ Travel to endemic/Ebola epidemic areas
▪ Wild animal contact (mostly nonhuman
primates)
COMPLICATIONS
▪ Gastrointestinal dysfunction
▫ Fluid loss, hypotension, acute kidney
injury, shock
▪ Neurologic
▫ Meningoencephalitis, meningitis
▪ Coagulopathy
▫ Infected macrophages produce
tissue factor (TF), stimulate extrinsic
coagulation pathway → disseminated
intravascular coagulation (DIC)
▪ Respiratory failure
▪ Coinfection/superinfection
▪ Impaired adaptive immunity → bacterial
sepsis
Figure 72.1 A scanning electron micrograph
of an ebola virus demonstrating the typical
filamentous structure seen in its class,
Filoviridae.
SIGNS & SYMPTOMS
Initial nonspecific symptoms
▪ Fever, chills, fatigue, headache, appetite
loss, malaise, sore throat, myalgias,
lumbosacral pain
Dermatological
▪ Diffuse erythematous; nonpruritic
maculopapular/morbilliform rash, especially
on torso/face
Gastrointestinal
▪ Watery diarrhea (up to 10L/2.2gal); nausea;
vomiting; epigastric, abdominal pain;
abdominal tenderness
82
Hemorrhage
▪ Commonly hematochezia, followed by
hematemesis, melena, metrorrhagia,
purpura, petechiae, ecchymoses, mucosal
bleeding, venipuncture site bleeding
▫ Often occur later in disease course;
not all infected individuals develop
significant bleeding; host susceptibilitydependent (genetically determined viral
immune response difference), different
species’ pathogenicity
Neurologic
▪ Meningoencephalitis symptoms (altered
level of consciousness, hyperreflexia,
myopathy, stiff neck, gait instability, seizure)
Ocular
▪ Conjunctival injection, uveitis symptoms
(blurred vision, photophobia, blindness)
Respiratory
▪ Tachypnea, dyspnea
Convalescent period
▪ Can persist > two years often followed by
symptoms such as arthralgia, blurred vision,
retro-orbital pain, hearing loss, alopecia,
difficulty swallowing, insomnia
▪ Infectious virus/viral RNA can persist
in body fluid (semen, breast milk, urine,
cerebrospinal fluid, aqueous humor) < nine
months after not detectable in blood →
variable transmission risk
DIAGNOSIS
▪ Diagnostic criteria
▫ Clinical manifestation
▫ Travel to endemic/Ebola epidemic areas
(within three weeks prior to disease
onset)
▫ Infected individual contact during acute
disease
▪ Laboratory findings
▫ Leukopenia; thrombocytopenia;
↑ ↓ hematocrit; ↑ aspartate
aminotransferase (AST), alanine
aminotransferase (ALT)
▫ Prolonged prothrombin (PT), partial
thromboplastin time (PTT)
▫ Proteinuria, ↑ blood urea nitrogen, ↑
creatinine
▫ Hyponatremia, hypocalcemia,
hyperkalemia
TREATMENT
▪ No cure
▪ Infected individual isolation; placement in
negative airflow room advised
MEDICATIONS
▪ Complications treatment
▫ Coinfection/superinfection: empiric
antimicrobial therapy with broad
spectrum antibiotics
▫ Shock: intravenous fluids,
vasoconstrictors
▫ Fever reduction: antipyretic agents
OTHER INTERVENTIONS
▪ Complications treatment
▫ Fluid, electrolyte loss correction
▫ Acute kidney injury: renal replacement
therapy (dialysis)
▫ Respiratory failure: supplemental
oxygen therapy, mechanical ventilation
(if necessary, barotrauma risk)
▪ Prevention
▫ Ervebo: FDA-approved vaccine
▫ Proven highly effective, safe during trial
conducted in Guinea (2015)
LAB RESULTS
▪ Reverse-transcription polymerase chain
reaction (RT-PCR)
▫ Detectable Ebola virus in blood samples
within three days after symptom onset
▪ Rapid chromatographic immunoassay
(ReEBOV)
83
NOTES
NOTES
FLAVIVIRUSES
GENERALLY, WHAT ARE THEY?
PATHOLOGY & CAUSES
▪ Single-stranded, ⊕-sense, enveloped RNA
viruses
▪ AKA arboviruses
▪ > 40 types identified
Replication
▪ Cell surface attachment → cytoplasm entry
→ viral protein translation → viral RNA
genome replication → virion formation
(encapsidation) → cellular release
Transmission
▪ Primarily transmitted via arthropod bites
RISK FACTORS
▪ Recent endemic area travel/residence
▪ Poor insect repellant use
▪ Improper skin coverage (e.g. long sleeve
clothing)
SIGNS & SYMPTOMS
▪ Mainly asymptomatic
▪ Others commonly have acute-onset flu-like
symptoms
▪ Serious complications/sequelae commonly
characterized by neurologic disease (e.g.
seizure, encephalopathy)
▫ Fever, nausea/vomiting
DIAGNOSIS
LAB RESULTS
▪ Serology
▪ Time-dependent on infection course,
immune response
▪ Reverse transcription polymerase chain
reaction (RT-PCR) for viral antigens
OTHER DIAGNOSTICS
▪ Endemic area travel/residence
▪ Presence of mosquito bite(s)
TREATMENT
▪ No cure
MEDICATIONS
▪ Variable vaccine availability
▫ Untenable mainly due to antigenic
mutations, multiple genotypes
OTHER INTERVENTIONS
▪ Fluid, electrolyte balance
84
AfraTafreeh.com exclusive
DENGUE VIRUS
osms.it/dengue-virus
PATHOLOGY & CAUSES
▪ Viral disease, mosquito transmission
▫ Characterized by febrile illness
▫ AKA break-bone fever
▪ Four serotypes
▫ DENV-1 through DENV-4
▫ IgG response to specific type → lifetime
immunity
▫ Limited, transient cross-protectivity
across viral types
▪ Vector
▫ Primarily Aedes aegypti and A.
albopictus
Pathogenesis
▪ Aedes mosquito bite → dengue virus skin
introduction → local infection, response →
dissemination → viremia (2–6 days later) →
circulating leukocyte infection (especially)
monocytes → viral replication, release →
fever
RISK FACTORS
▪ Endemic area residence
▪ Recent travel
COMPLICATIONS
Severe dengue
▪ AKA dengue hemorrhagic fever
▪ Viremia → local hemorrhage → systemic
hemorrhage → hemodynamic collapse →
shock
▫ Dengue shock syndrome (rare
complication—DENV-2 confers highest
risk)
▪ Direct bone marrow infection →
hematopoietic stem cell infection → ↓
megakaryocyte differentiation (among all
other cell lines) → ↓ platelets formed
▫ ↓ Circulating lifetime: virus-antibody
complex → circulating platelet
adherence → complement destruction
▪
▪
▪
▪
of virus-antibody complex → adherent
platelet destruction
Capillary leak
▫ Direct effect: viral endothelial cell
infection → cellular dysfunction → tight
junction widening → capillary leak
▫ Indirect: virus-infected monocytes,
dendritic cells, mast cells → TNFalpha, IFN-gamma, IL-2, IL-8, vascular
endothelial growth factor release,
complement activation → ↑ capillary
permeability
Molecular mimicry
▫ Viral E protein antibody response
→ plasminogen cross-reaction →
inefficient coagulation cascade →
hemorrhage
Liver failure
▫ Direct viral infection → Kupffer
cells, hepatocytes → apoptosis →
hepatocellular necrosis, Councilman
bodies
▫ Sever disease → shock → liver
hypoperfusion → hepatocellular injury,
death
Central nervous system (CNS) involvement
▫ Rare, direct viral infection of brain
parenchyma
SIGNS & SYMPTOMS
▪ High-grade fever (> 38.5°C/101.3°F)
▪ Generalized pain
▫ Abdominal pain/tenderness, headache,
joint pain, muscle pain, eye/retro-orbital
pain
▪ Nausea/vomiting
▪ Mucosal bleeding
▪ Rash
Severe symptoms and signs
▪ Severe bleeding
▪ Fluid accumulation → respiratory distress
▫ Ascites, pleural effusion
85
Chapter 73 Flaviviruses
▪ Neurologic impairment
▫ Lethargy, seizure, encephalopathy
DIAGNOSIS
LAB RESULTS
▪ Leukopenia
▪ Thrombocytopenia
▪ ↑ Hematocrit
TREATMENT
MEDICATIONS
▪ No antiviral therapy available
▪ Fever control
▫ Antipyretics
OTHER INTERVENTIONS
Antibody testing
▪ Antigen assay
▫ ⊕ < five days of infection
▪ IgM
▫ ⊕ ≧ three days of infection
▪ IgG
▫ ⊕≧ seven days of infection
▫ May be ⊕ due to prior infection
OTHER DIAGNOSTICS
Positive tourniquet test
▪ Blood pressure cuff insufflation → maintain
pressure midway between systolic,
diastolic → hold for five minutes → deflate
→ observe for petechiae
▫ Positive test:10+ petechiae in 2.5cm/1in
▪ Maintain adequate intravascular volume
▪ Shock
▫ Crystalloid fluid resuscitation
▪ Bleeding
▫ Platelet transfusion if severe
thrombocytopenia and/or active,
uncontrolled bleeding
Prevention
▪ Mosquito control
▫ ↓ Standing water breeding sites
▫ Copepod use (organisms feed on
mosquito larvae)
▫ Insecticides
▪ Personal protective measures
▫ Repellant use
▫ Permethrin-treated clothing (Na+
channel blockade → neurotoxicity
→ paralysis → death; low toxicity in
humans)
WEST NILE VIRUS
osms.it/west-nile-virus
PATHOLOGY & CAUSES
▪ Virus causing mosquito-borne, self-limited
disease
▫ Characterized by flu-like symptoms
▫ Potential for severe neurologic sequelae
▪ Vector
▫ Culex mosquito
Pathogenesis
▪ Culex mosquito bite → viral replication
in skin’s dendritic cells → lymph node
migration → further replication →
enters bloodstream → visceral organ
dissemination
▫ Complication arise when virus enters
bloodstream → crosses blood-brain
barrier
Transmission
▪ Infected bird (amplifying host) → prolonged
viremia → Culex mosquito blood meal
→ human/other vertebrate bite → blood
meal, virus-laden saliva injected→ virus
transmission
▪ Viral-load organ/blood donation
▪ Transplacental infection
86
RISK FACTORS
▪ Elderly
▪ Immunosuppressed individuals
▪ Malignancy (especially hematologic
malignancies)
▪ Chemokine receptor CCR5 deficiency
▫ Chemokine involved in viral infection
immune response (especially brain
parenchyma)
COMPLICATIONS
▪ Neurological
▫ Meningitis, encephalitis, tremor, flaccid
paralysis, Guillain–Barré syndrome
▪ Ocular
▫ Chorioretinitis, uveitis, optic neuritis
▪ Cardiac
▫ Myocarditis, cardiac arrhythmias
▪ Muscular
▫ Myositis, rhabdomyolysis
▪ Other
▫ Pancreatitis, orchitis, hepatitis, central
diabetes insipidus
SIGNS & SYMPTOMS
▪ Abrupt-onset fever, headache, myalgias
▫ Usual presentation is self-limited (AKA
West Nile Fever)
▪ Abdominal pain, anorexia, nausea/vomiting,
diarrhea, maculopapular rash
Neurologic complications
▪ Motor
▫ Tremor, flaccid paralysis (Guillain-Barré
complications)
▪ Meningoencephalitis
▫ Meningismus, altered mental status
DIAGNOSIS
DIAGNOSTIC IMAGING
Nerve action potentials
▪ Normal sensory conduction velocity
▪ Abnormally ↓motor neuron velocity,
potentials
LAB RESULTS
▪ Reverse transcription polymerase chain
reaction (RT-PCR)
▪ Viral cultures
▪ Cerebrospinal fluid
▫ Severe cases
▫ If present in CSF → neuroinvasive
disease likely
▫ Neutrophilic pleocytosis (<500 cells/
microL) → lymphocytic predominance
as disease progresses
▫ ↑ Protein (<150mg/dl)
▪ Serology
▫ Enzyme-linked immunosorbent assay
(MAC-ELISA) for IgM detection →
positive 4-10 days after viremia
detection
OTHER DIAGNOSTICS
▪ Integument examination
▫ Maculopapular rash
▪ Neurological
▫ Meningismus (in severe cases)
Electroencephalography (EEG)
▪ Generalized, continuous slowing prominent
in frontal/temporal regions
TREATMENT
▪ No cure
OTHER INTERVENTIONS
Prevention
▪ Personal protection measures
▪ Mosquito control programs
▪ Blood donor screening
MRI
▪ Neurologic sequelae → ↑ signal in T2
imaging
87
Chapter 73 Flaviviruses
YELLOW FEVER VIRUS
osms.it/yellow-fever-virus
PATHOLOGY & CAUSES
▪ Virus that causes mosquito-borne illness
in endemic, tropical areas (Africa, South
America)
▫ Characterized by hemorrhagic fever,
coagulopathy, shock (most severe
cases)
▪ Seven major genotypes, conserved in each
specific endemic region
▫ Only one serotype that has remained
antigenically conserved
▪ Vector
▫ Aedes, Haemagogus mosquitoes
Pathogenesis
▪ Female Haemagogus mosquito bite →
viral transmission → skin dendritic cells
infected→ lymphatic spread → monocyte/
macrophages, histiocytes in lymph node →
replication → lymphatic, hematogenous
spread throughout body
▫ 3–6 day incubation period until
symptoms appear
Transmission
▪ Infected monkey → Haemagogus mosquito
bite → human bite → blood meal, viral
transmission
▪ During epidemics/urban spread
▫ Aedes mosquito feeds on infected
humans → infected Aedes mosquito
→ feeds on another human → viral
transmission
COMPLICATIONS
▪ Hepatic injury → viral hepatitis
▫ Councilman bodies (hepatocytes
characterized by eosinophilic
degeneration with condensed nuclear
chromatin) present on histology
▫ Jaundice results
▫ Severe disease → ↓ vitamin
▪
▪
▪
▪
▪
K-dependent coagulation factors →
hemorrhagic diathesis
Shock
▫ Due to cytokine dysregulation
▫ ↑ IL-1, IL-6, TNF-alpha, IFN-10
Renal failure
▫ Hypotension/hepatorenal syndrome
→ eosinophilic degeneration, renal
parenchyma fatty change
Hemorrhage
Secondary bacterial infection
Myocardial infection
Neurological
▪ Delirium, convulsions, coma
SIGNS & SYMPTOMS
▪ Infection period (3–4 days)
▫ Acute-onset headache, fevers, chills,
myalgia
▫ Photophobia, restlessness, anorexia,
vomiting
▪ Remission period (up to 48 hours)
▫ Afebrile
▫ No other symptoms
Severe yellow fever
▪ Intoxication period (3–6 days after infection
onset)
▪ Nausea/vomiting, fever, oliguria → anuria,
epigastric tenderness, jaundice
▪ Hemorrhage
▫ Coffee-ground emesis, melena,
hematuria, petechiae, ecchymoses
▪ Myocardial injury
▫ Cardiac enlargement, conduction
abnormalities
▪ Neurological dysfunction
▫ Altered mental status, convulsions,
delirium
88
OTHER DIAGNOSTICS
DIAGNOSIS
LAB RESULTS
▪ Renal dysfunction
▫ Azotemia, proteinuria, creatinine (3–-8x
normal value)
▪ PCR
▫ Viral genome
▪ CSF analysis
▫ ↑ opening pressure, ↑ protein, ⊝ cells
Serology
▪ Enzyme-linked immunosorbent assay
(ELISA)
▫ IgM antibodies
▪ Reverse-transcriptase loop-mediated
isothermal amplification (RT-LAMP)
▫ Field test for quick diagnostics
▫ Easy-to-read visual test, good sensitivity
ECG
▪ Cardiac conduction abnormalities
▪ ST-T abnormalities
TREATMENT
OTHER INTERVENTIONS
▪ Fluid management, resuscitation
▪ Bleeding diathesis reversal
▫ Fresh frozen plasma
▪ Adequate nutrition
Prevention
▪ Live attenuated vaccine
▫ Recommended for individuals > nine
months old residing in/traveling to
endemic areas
▫ Contraindication in children < six
months old
▪ Passive maternal immunization
▫ Safest immunoglobulin transfer
ZIKA VIRUS
osms.it/zika-virus
PATHOLOGY & CAUSES
▪ Mosquito-borne virus
▫ Characterized by mild disease in
infected individuals, serious neurologic
sequelae in developing fetus
▪ Vector
▫ Aedes aegypti/albopictus mosquitoes
Pathogenesis
▪ Aedes mosquito bite → dendritic cell
infection, replication → bloodstream →
dissemination
▪ Maternal infection → placental injury →
fetus infection → neuronal progenitor cell
infection → neuronal cell death
Transmission
▪ Human-to-human close contact
▫ E.g. sexual contact
▫ Possible transmission via other
bodily secretions (e.g. tears, sweat) in
individuals with very high viral loads
▪ Blood transfusion
▪ Organ transplantation
▪ Maternal-fetal spread
COMPLICATIONS
▪ Microcephaly in developing newborns
▪ Greatest risk for sequelae estimated to
occur with first, second trimester infection
▫ Serious neurological, intellectual deficits
▫ Seizures
▫ Vision, hearing problems
▪ Neurological sequelae
▫ Guillain–Barré syndrome
▫ Myelitis
89
Chapter 73 Flaviviruses
▫ Meningoencephalitis
SIGNS & SYMPTOMS
▪
▪
▪
▪
Mild fever
Maculopapular rash
Nonpurulent conjunctivitis
Joint, muscle pain
DIAGNOSIS
DIAGNOSTIC IMAGING
Ultrasound
▪ Pregnant individuals
▫ Microcephaly, growth retardation
evaluation
LAB RESULTS
▪ RT-PCR
▫ If < 14 days from symptoms onset
▫ Confirmatory testing with plaque
reduction neutralization test (PRNT)
▫ PRNT: measure of antibodies that
neutralize virus-specific antigens
Serology
▪ IgM against Zika virus
▫ If > 14 days from onset of symptoms
TREATMENT
▪ No cure
MEDICATIONS
▪ Antipyretics
OTHER INTERVENTIONS
Mother
▪ Rest
▪ Fluid management, resuscitation
Newborn
▪ Nutritional support
▪ Physical therapy
▪ Specialty referral (especially neurology)
Prevention
▪ No vaccine available
▪ Personal protective measures in endemic
areas
▪ Consultation with physician prior to travel
for pregnant individuals
▪ Safe sexual practice
▫ Biologically-male individuals wait at
least six months after endemic-area
travel for unprotected sex
▫ Biologically-female individuals wait at
least eight weeks after endemic-area
travel for unprotected sex
▪ Blood donation
▫ Do not donate for at least six months
after endemic-area travel
90
91
NOTES
NOTES
GASTROINTESTINAL INFECTIONS
GENERALLY, WHAT ARE THEY?
PATHOLOGY & CAUSES
▪ Gastrointestinal tract (GIT) inflammation
caused by virus, bacteria, other parasites
▪ GIT mucosa inflammation → ulceration →
epithelial disruption → edema, bleeding
→ fluid, electrolyte loss (diarrhea) →
dehydration, electrolyte imbalance, anemia
(bloody diarrhea)
▪ Mainly fecal-oral transmission
RISK FACTORS
▪ Living/traveling to endemic areas, youth,
immunosuppression (e.g. corticosteroid
treatment, HIV co-infection), malnutrition,
poor hygiene
DIAGNOSIS
▪ Pathogen-dependent
LAB RESULTS
▪ Stool culture
TREATMENT
▪ Rehydration
▪ Antimicrobial therapy (pathogendependent)
SIGNS & SYMPTOMS
▪ Fever, diarrhea, abdominal pain (cramps)
▪ Dehydration
▫ Sunken eyes, dry mouth, decreased
urination, dark yellow urine (deep
amber—severe), dry skin, syncope
92
CRYPTOSPORIDIUM
osms.it/cryptosporidium
PATHOLOGY & CAUSES
▪ Cryptosporidiosis: diarrheal disease caused
by Cryptosporidium (intestinal intracellular
protozoan parasite)
▪ Life-cycle can be completed in one host
▫ Immunocompetent hosts: self-limited
diarrhea
▫ Immunocompromised hosts: lifethreatening complications
CAUSES
▪ Cryptosporidium oocysts (infective form)
transmitted via fecal → oral route
▫ Infected individual/animal feces
contaminates food; drinking, swimming
water → fecally-contaminated food/
water ingestion
▪ Parasites → intestinal epithelial
inflammation → villi structure distortion →
↓ absorption, ↑ secretion → watery diarrhea
▫ Sclerosing cholangitis/acalculous
cholecystitis, respiratory
cryptosporidiosis, pancreatitis
RISK FACTORS
▪ Endemic-area exposure (tropical countries,
Kuwait), immune deficiencies, poor hygiene
▪ Interpersonal transmission: sexual
partners, daycare centers, household
members
COMPLICATIONS
DIAGNOSIS
LAB RESULTS
Microscopic oocyte identification
▪ Stool; bile secretion, affected GIT aspirates;
affected GIT tissue biopsy; respiratory
secretion
Polymerase chain reaction (PCR)
▪ More sensitive, specific
▪ Differentiates between Cryptosporidium
genotypes
Monoclonal antibodies and enzyme immunoassays (EIA)
▪ Monoclonal antibody test against oocyst
wall
▪ More sensitive, specific than light
microscope
TREATMENT
MEDICATIONS
▪ Immunocompetent host: antidiarrheal,
antimicrobial agents
▪ Immunocompromised host: antiretroviral
therapy (HIV-infected individuals),
antimicrobial agents, azithromycin (severe
diarrhea)
OTHER INTERVENTIONS
▪ Immunocompetent host: oral/IV fluid/
electrolyte-loss replacement
▪ Dehydration, fluid and electrolyte imbalance
SIGNS & SYMPTOMS
▪ Host’s immune status-dependant
93
Chapter 74 Gastrointestinal Infections
Figure 74.1 Cryptosporidium organisms
lining the crypt epithelium in an infected
individual.
94
ENTAMOEBA HISTOLYTICA
(AMOEBIASIS)
osms.it/entamoeba-histolytica
PATHOLOGY & CAUSES
SIGNS & SYMPTOMS
▪ Amebiasis
▫ Caused by Entamoeba histolytica
(anaerobic parasitic protozoan)
▪ Trophozoites bind to intestinal epithelial
cells in colon, release lytic enzymes (e.g.
cysteine proteinases) → epithelial cell lysis
→ trophozoites lyse inflamed/attracted
immune cells → immune cell’s lytic
enzymes ↑ release
▫ Intestinal mucosa ulcers → colitis →
bowel necrosis → perforation → sepsis
▫ Tissue destruction → mucosa blood
vessel injury, malabsorption, ↑ intestinal
secretion → bloody diarrhea, amebic
dysentery
▫ Blood vessel injury → trophozoites
in blood stream → extraintestinal
amebiasis (liver, pulmonary, cardiac,
brain)
▪ Mostly asymptomatic; bloody diarrhea,
mucus in stool (severe dysentery);
abdominal pain; fever; weight loss; right
upper-quadrant pain, jaundice (liver); cough
(pulmonary); dehydration
RISK FACTORS
▪ Endemic-area exposure (Africa, Southern
Asia, Central America)
▪ Intimate partner transmission possible
▪ Youth
▪ Malnutrition
▪ Immunodeficiency (e.g. malignancy,
corticosteroid treatment, HIV)
▪ Poor hygiene
COMPLICATIONS
▪ Amebic liver abscess rupture
▫ Pericarditis, peritonitis
▪ Toxic megacolon
▪ Cerebral amebiasis → brain abscess → ↑
intracranial pressure
▪ Cutaneous amebiasis
▪ Dehydration
DIAGNOSIS
DIAGNOSTIC IMAGING
X-ray
▪ Liver
CT scan, MRI, ultrasound
▪ Cystic intrahepatic cavity detection
LAB RESULTS
Microscopic identification
▪ Cysts/trophozoites in stool/pus (e.g. liver
abscess)
Antigen detection
▪ Enzyme-linked immunosorbent
assay (ELISA), radioimmunoassay,
immunofluorescence
PCR
▪ Entamoeba DNA detection
Serology
▪ Entamoeba antibodies detection
Sigmoidoscopy/colonoscopy
▪ Histological examination biopsies
95
Chapter 74 Gastrointestinal Infections
TREATMENT
MEDICATIONS
▪ Antibacterial agents
▫ Invasive amoebic colitis
▪ Luminal agents
▫ Intraluminal cysts, trophozoites
▪ Metronidazole
▫ Amebic liver abscess ≤ 10cm/3.94in
▪ Broad-spectrum antibiotics
▫ Suspected perforation, bacterial
superinfection
SURGERY
▪
▪
▪
▪
▪
Figure 74.2 Trophozoites of Entamoeba
histolytica in a colonic biopsy. The
trophozoites have ingested red blood cells.
Massive GIT bleeding
Amebic liver abscess > 10 cm/3.94 in
Ruptured amebic liver abscess
Perforated amebic colitis
Toxic megacolon
OTHER INTERVENTIONS
▪ Rehydration
GIARDIA LAMBLIA
osms.it/giardia-lamblia
PATHOLOGY & CAUSES
▪ Giardiasis
▫ Diarrheal disease caused by Giardia
intestinalis/Giardia duodenalis
(flagellated protozoan parasite,
colonizes small intestine)
▪ Pathogenesis not well understood
▪ Infection causes microvilli shortening →
intestinal malabsorption, hypersecretion →
diarrhea
CAUSES
▪ Contaminated/untreated water ingestion
▪ Contaminated food (uncommon)
RISK FACTORS
▪ Endemic-area exposure (tropical countries),
immunosuppression, comorbidities (e.g.
cystic fibrosis), poor sanitation
COMPLICATIONS
▪ Weight loss; dehydration; zinc,
disaccharidase deficiency; malabsorption
syndrome (adult); growth delay (children)
96
SIGNS & SYMPTOMS
▪ Usually asymptomatic
Acute giardiasis
▪ 7–14 days after infection exposure
▪ Diarrhea, malaise, abdominal pain,
flatulence, nausea/vomiting, malodorous
stool, steatorrhea, fever (uncommon)
Chronic giardiasis
▪ > 18 days after infection exposure
▪ Loose stools (not typical diarrhea), profound
weight loss (occasionally), abdominal
pain, borborygmus (moving gas/fluid →
GIT gurgling sound), flatulence, burping,
malaise, fatigue, depression
Figure 74.3 Giardia lamblia in a cytology
specimen.
DIAGNOSIS
LAB RESULTS
Antigen detection assays
▪ Trophozoite antigen (stool) detection
Nucleic acid amplification assays (NAAT)
▪ Giardia DNA detection
Stool microscopy
▪ Giardia cyst detection
Figure 74.4 A duodenal biopsy
demonstrating giardia organisms in the
duodenal crypt.
TREATMENT
MEDICATIONS
▪ Antimicrobial therapy
▫ Paromomycin (pregnant/lactating
individuals)
97
NOTES
NOTES
GENERAL INFECTIONS
GENERALLY, WHAT ARE THEY?
TREATMENT
PATHOLOGY & CAUSES
▪ Localized, systemic disorders caused by
microbial infections
SIGNS & SYMPTOMS
MEDICATIONS
▪ Antimicrobials
OTHER INTERVENTIONS
▪ Drainage
▪ See individual conditions
DIAGNOSIS
▪ See individual conditions
ABSCESSES
osms.it/abscesses
PATHOLOGY & CAUSES
▪ Localized, circumscribed pus collection
surrounded by inflamed tissue
▪ May develop in any body region
▫ Superficial (e.g. skin, soft tissue)
▫ Internal (e.g. lung, liver, brain)
▪ Caused by pyogenic bacteria (e.g. S.
aureus, S. pyogenes, S. epidermidis, P.
aeruginosa)
▪ Bacterial invasion → local inflammatory
response
▫ Suppuration (pus production)
▫ Necrosis, liquefaction
▫ Cellular debris accumulation
▪ Loculation, walling off of abscess by
adjacent, healthy cells
RISK FACTORS
▪
▪
▪
▪
▪
Trauma
Foreign body presence (e.g. body piercing)
Intravenous (IV) drug use
Dermatological conditions (e.g. cellulitis)
Anatomical involvement-related risks
▫ Examples: ascending infection → pelvic
abscess, hematologic spread → central
nervous system (CNS) abscess, local
spread → liver abscess
COMPLICATIONS
▪ Spread to other tissue
▪ Fistula formation
▪ Sepsis
98
SIGNS & SYMPTOMS
▪ Localized warmth, erythema, swelling,
tenderness/pain, induration
▪ Fluctuant mass
DIAGNOSIS
▪ History, physical examination: characteristic
findings
DIAGNOSTIC IMAGING
CT scan
▪ Central decreased attenuation area with
circumferential enhancement ring
MRI
▪ T1: central hypointense area
▪ T2: hyperintense
▪ T1, T2: rim is iso- to hypointense
Ultrasound
▪ Internal abscess
▫ Homogeneous fluid collection appears
as hypoechoic region within tissue
▫ Edema: cobblestone appearance—thin
hyperechoic (dark gray) bands, anechoic
(black) fluid
Figure 75.1 The clinical appearance of a
pilonidal abscess.
LAB RESULTS
▪ Ultrasound-guided needle aspiration
▫ Specimen collection, culture
TREATMENT
MEDICATIONS
▪ Antimicrobials
OTHER INTERVENTIONS
▪ Incision, drainage
Figure 75.2 A CT scan of the brain in the
coronal plane demonstrating a cerebral
abscess of the left frontal lobe. The abscess
shows typical ring enhancement.
99
Chapter 75 General Infections
SEPSIS
osms.it/sepsis
PATHOLOGY & CAUSES
▪ Serious, life-threatening, systemic infection
reaction
▫ Microbial host barrier breach (skin,
mucous membranes) → provokes host
dysregulated immune response →
proinflammatory mediator release (e.g.
TNF-α, interleukins) → detrimental
physiological effects, tissue damage
▫ Endothelial damage, dysfunction; ↑
vascular permeability; microvascular
dysfunction; coagulopathies
RISK FACTORS
▪ Bacteremia (most common) Gram-positive
organisms
▪ Hospital/intensive care unit (ICU) admission
→ nosocomial infection
▪ Immune system deficiency
▫ E.g. HIV/AIDS, hematologic malignancy,
immunosuppressant medication
▪ Recent surgery/hospitalization → altered
microbiome
▪ Indwelling medical device presence
▫ E.g. urinary catheter, venous access
device
▪ Bimodal age distribution
▫ Infants, adults ≥ 65
▪ Community-acquired pneumonia
▪ Chronic disease
▫ E.g. diabetes, heart failure (HF), chronic
obstructive pulmonary disease (COPD)
▪ Genetic factors
▪ Respiratory failure
▪ Disseminated intravascular coagulation
(DIC)
▪ Metabolic acidosis
▪ Stress ulcer
▪ Treatment complications
▫ E.g. ventilator-associated pneumonia,
venous thrombosis
▪ Death
SIGNS & SYMPTOMS
General presentation
▪ Fever; tachypnea; tachycardia; hypotension;
hypoxemia; ↓ urine output; ↓ PaO2; edema;
ileus, ↓ bowel sounds
▪ Altered mental status
▫ Malaise, agitation, lethargy, stupor
▪ Signs of shock
▫ E.g. cool skin, cyanosis, ↑ capillary refill,
mottling
Quick Sequential Organ Failure Assessment (qSOFA)
▪ Score ≥ 2 → ↑ mortality risk
▪ See table
COMPLICATIONS
▪ Severe sepsis → septic shock
▫ Sepsis-induced vasodilation,
hypotension
▫ Unresponsive to fluid resuscitation
▪ Multiple organ dysfunction syndrome
(MODS)
100
DIAGNOSIS
▪ History, clinical presentation, physical
examination
LAB RESULTS
Blood studies
▪ ↑ white blood cell (WBC) count, left shift
▪ ↑ C-reactive protein (CRP), erythrocyte
sedimentation rate (ESR)
▪ Indications of organ hypoperfusion, injury,
dysfunction
▫ ↑ lactate, ↓ platelets (may precede DIC),
↑ glucose, ↑ creatinine, ↑ bilirubin
▪ Coagulation studies
▫ ↑ international normalized ratio (INR),
activated partial thromboplastin time
(aPTT)
▪ Blood cultures
▫ Identify pathogen
TREATMENT
SURGERY
▪ Source control
▫ E.g. infected device removal, abscess
drainage)
OTHER INTERVENTIONS
Hemodynamic support
▪ Invasive monitoring
Respiratory support
▪ Supplementary oxygen; mechanical
ventilation
Infection prevention
▪ Infection control practices
▫ ↓ hospital-acquired infections
(ventilator-associated pneumonia,
catheter-associated bloodstream
infections)
Complication prevention
▪ Deep-vein thrombosis prophylaxis
▪ Glycemic control
MEDICATIONS
▪ Antimicrobials
▫ Initial broad-spectrum antibiotics until
pathogen identified
▪ Hemodynamic support
▫ Fluid resuscitation
▫ Vasopressors
▪ Infection prevention
▫ Vaccinations for at risk individuals
(influenza, pneumonia)
▪ Complication prevention
▫ Proton pump inhibitor
101
NOTES
NOTES
GRAM VARIABLE
GARDNERELLA VAGINALIS
(BACTERIAL VAGINOSIS)
osms.it/gardnerella-vaginalis
PATHOLOGY & CAUSES
DIAGNOSIS
▪ Facultative, anaerobic, gram-variable
coccobacilli
▪ Microscopically
▫ Very thin gram-positive cell wall
appears gram-positive/negative
▪ Non-spore forming, non-motile
▪ Causative agent for bacterial vaginosis (BV)
▫ Disruption of vaginal microbiome
▪ Most common vaginal infection in
individuals who are biologically female, age
15–44
LAB RESULTS
RISK FACTORS
OTHER DIAGNOSTICS
▪ Multiple sex partners, douching, smoking
COMPLICATIONS
▪ ↑ risk of contracting HIV, other STDs
▪ Acute cervicitis, endometritis, postabortal
infection
▪ During pregnancy
▫ Premature birth, low birthweight
SIGNS & SYMPTOMS
▪ Malodorous (fishy), thin white/gray vaginal
discharge
▪ Vaginal pain, itching, burning
▪ Burning with urination
▪ Gram-stain smear of vaginal discharge;
using criteria (e.g. Nugent, Hay/Ison)
▪ Affirm VP III
▫ Commercial automated DNA probe
assay detects G. vaginalis
▪ OSOM BVBlue test
▫ Chromogenic identifies presence of
elevated sialidase enzyme produced
by G. vaginalis, other bacteria (e.g.
Bacteroides, Prevotella, Mobiluncus)
▪ Requires three of the following Amsel
criteria
▫ Characteristic vaginal discharge
▫ pH > 4.5
▫ Clue cells visible on saline wet mount
(vaginal epithelial cells covered with
bacteria)
▫ Positive whiff-amine test (combine
vaginal discharge sample with 10%
KOH → fishy odor)
TREATMENT
MEDICATIONS
▪ Antibiotics: metronidazole
102
103
NOTES
NOTES
HEMATOLOGIC INFECTIONS
GENERALLY, WHAT ARE THEY?
PATHOLOGY & CAUSES
Taxonomy
▪ Apicomplexan phylum protozoa →
infectious hematological diseases
Replication/multiplication
▪ Arthropod transmission
Transmission
▪ Multiple life cycle stages (host-, vectordependent)
DIAGNOSIS
LAB RESULTS
▪ Blood smear: best intra-erythrocyte
location diagnosis
TREATMENT
▪ See individual infections
SIGNS & SYMPTOMS
▪ See individual infections
104
BABESIA
osms.it/babesia
PATHOLOGY & CAUSES
RISK FACTORS
▪ Malaria-like parasitic infection
▫ Ixodes tick transmission
▪ Phylum Apicomplexa (same as
Plasmodium, Toxoplasmosis)
▪ Common species
▫ Babesia microti most predominant
(northeast, upper midwest United
States)
▫ B. duncani (Western United States)
▫ B. divergens (Europe)
▪ Endemic area resident
▪ Endemic area travel, especially May–
September
▪ Blood transfusion (last six months)
▪ Age > 50
▪ Biologically male
▪ Asplenia
▪ Decreased immunity (malignancy, HIV/
AIDS infection, immunosuppressive drugs)
▪ Coinfection with Borrelia and/or Anaplasma
▪ Premature birth
CAUSES
COMPLICATIONS
▪ Ixodes scapularis tick: only recognized
vector
▫ Same tick transmits Borrelia burgdorferi
(→ Lyme disease; concurrently infects
⅔ babesiosis-infected individuals) and
Anaplasma phagocytophilum (→ human
granulocytic anaplasmosis; concurrently
infects ⅓ babesiosis-infected
individuals)
▫ Contaminated blood transfusion → rare
human-to-human transmission
▪ Life cycle
▫ Two hosts: white-footed mouse, Ixodes
tick (definitive)
▫ Blood meal → Babesia-infected
tick sporozoites into mouse host →
sporozoites directly invade mature
mouse erythrocytes → sporogony
(asexual reproduction; budding) → in
blood, some parasites differentiate into
male, female gametes → gametes unite
→ sporogonic cycle → sporozoites
→ tick bites human → 1–4 week
incubation → sporozoites enter
erythrocytes → sporogony
▪ Sporogony
▫ Asynchronous with host → no massive
hemolysis (versus malaria)
▪ Tick’s nymph stage is most infectious
▪ ↑ occurence in anemic/parasitemic
individuals
▫ Congestive heart failure
▫ Noncardiac pulmonary edema
▫ Acute respiratory distress syndrome
▫ Splenic infarct
▫ Splenic rupture
▫ Septic shock
▫ Myocardial infarction
▫ Disseminated intravascular coagulation
▫ Death
SIGNS & SYMPTOMS
▪ Mostly asymptomatic, may persist
undiagnosed months/years
▪ Blood-stage parasite multiplication →
clinical manifestation
▪ Non-specific flu-like symptoms
(misdiagnosis common)
▫ Fatigue (gradual onset)
▫ Fever, chills, sweats
▫ Headache, myalgia, arthralgia
▫ Anorexia, nausea
▫ Cough
105
Chapter 77 Hematologic Infections
▪ Severe manifestations include malaria-like
illness (fever, fatigue, malaise) + hemolytic
anemia manifestation
▪ B. microti case-fatality rate is 5%
▪ B. divergens infections case-fatality rate is
42%
▫ Disseminated intravascular coagulation,
bleeding diathesis
▫ Acute renal failure
▫ Cardiopulmonary complications (e.g.
hypotension, poor perfusion, pulmonary
edema)
OTHER INTERVENTIONS
▪ Severe disease (parasitemia > 4%)
▫ Antimicrobial + exchange transfusion
Prevention
▪ Personal: avoid endemic areas; long pants,
shirts minimize exposed skin
▪ Tick repellant on skin (e.g. DEET)
▪ Post-exposure tick checks
▪ No antibiotic prophylaxis
DIAGNOSIS
LAB RESULTS
Blood smear
▪ Wright/Giemsa staining
▫ Thin: rapid, ↓ microscopy experience
required
▫ Thick: ↑ accurate, ↑ microscopy
experience required
▫ Pathognomic tetrad “Maltese cross”
visible in erythrocyte
Laboratory
▪ Thrombocytopenia
▪ Reticulocytosis
▪ ↓ Hematocrit
▪ ↓ Hemoglobin
▪ ↑ Lactate dehydrogenase
▪ ↓ Haptoglobin
▪ ↑ Liver function tests
▪ ↑ Creatinine
Polymerase chain reaction (PCR)
▪ Speciation (useful for low-level parasitemia)
Serology
▪ Immunofluorescent antibody testing;
species specific
TREATMENT
▪ Asymptomatic: no treatment
MEDICATIONS
▪ Symptomatic disease: azithromycin +
atovaquone
106
PLASMODIUM SPECIES (MALARIA)
osms.it/malaria
PATHOLOGY & CAUSES
▪ Anopheles mosquito vector → parasitic
hematologic infection
▪ Four major malarial parasite species
▫ Plasmodium falciparum: most lethal,
most drug-resistant (sub-Saharan
Africa)
▫ P. vivax: widest geographic distribution,
relapsing species
▫ P. ovale: relapsing species (western
areas of sub-Saharan Africa)
▫ P. malariae: AKA ‘benign’ malaria (mild
course)
▪ P. knowlesi: normally infects macaques,
recent cause of human malaria cases
▪ Erythrocytic life cycle stage
▫ In erythrocytes → parasites can
undergo asexual schizogony/
sexual differentiation (necessary for
transmission) → gametocytes
▪ Asexual schizogony
▫ Trophozoites (parasite name once inside
erythrocyte) digest host cell hemoglobin
(amino acids, energy source) → schizont
→ undergoes mitosis → differentiates
into merozoites → erythrocyte rupture
→ merozoite bloodstream release →
fever, malarial symptoms
CAUSES
▪ Exoerythrocytic (sporogonic; blood) life
cycle stage
▫ Blood-stage gametocytes in other
host (e.g. human) → female Anopheles
mosquito blood meal → mosquito
multiplication, growth cycle → 10–18
day incubation → sporozoites in
mosquito salivary gland → human
bite → sporozoite inoculation →
hematogenous translocation to liver →
rapid hepatic parenchymal cell invasion
→ parasites undergo exoerythrocytic
schizogony (asexual multiplication) →
development, multiplication → 7–14 day
incubation → merozoite development
→ invade erythrocytes → symptomatic
stage develops → P. ovale, P. vivax
can differentiate into quiescent stage
→ hypnozoite → can re-enter into
schizogony → reemerge to invade
erythrocytes
▫ Blood stage → malaria symptoms
▫ Mosquito vector does not have parasite
presence
Figure 77.1 A peripheral blood film taken
from an individual with Plasmodium malariae
infection. There is a mature schizont,
composed of 6-12 merozoites, contained
within a red blood cell.
▪ P. malariae
▫ Low-level persistence possible for
decades without diagnosis, treatment
▪ P. vivax
▫ Prefers erythrocytes with Duffy blood
group antigen (rare in persons from
West and Central Africa); prefers
reticulocytes but will also invade mature
erythrocytes
▪ Others preferentially invade mature
erythrocytes
107
Chapter 77 Hematologic Infections
▪ P. falciparum
▫ Develop ‘knobs’ on infected
erythrocytes in late trophozoite stage
→ parasitized erythrocytes adhere to
capillary endothelium → various organ
sequestration (notably brain) → cerebral
malaria
▪ Most malaria deaths
▫ Children < five years old in hightransmission areas
COMPLICATIONS
Coma
Seizure
Severe anemia
Acute renal failure
▫ Acute tubular necrosis secondary to
hypoperfusion, hypovolemia
▪ Acute respiratory distress syndrome
▪ Shock
▪ Septicemia
▪
▪
▪
▪
SIGNS & SYMPTOMS
RISK FACTORS
▪ Poor rural populations in endemic areas
▪ Proximity to standing water
▫ Larvae breeding site (e.g. agriculture,
irrigation ditches)
▪ Travel in endemic areas
▪ Blood transfusion (last six months)
▪ Lack of insect repellant, chemoprophylaxis,
personal protective measures (long pants,
shirts, mosquito nets)
▪ Duffy blood group antigen-positive
individuals
Protective factors
▪ P. falciparum protection: sickle cell trait
(heterozygotes)
▪ P. vivax: Duffy blood group antigennegative individuals
▪ Febrile paroxysm (hallmark feature)
▫ 10–12 hours of intense rigors, chills →
high fever, profuse diaphoresis
▫ Febrile seizure potential
▪ Non-specific complaints
▫ Headache, malaise, myalgia, arthralgia
▪ Abdominal pain, diarrhea, vomiting
▪ Possibly remarkably asymptomatic
between episodes
▪ P. falciparum cerebral effects
▫ Significant morbidity, mortality (delirium,
confusion, seizures → declining mental
status → coma → death)
▪ Parasitemia, cytokine disturbance
→ hypotension, metabolic acidosis,
hypoglycemia
▪ Anemia (high parasitemia → more
pronounced)
▪ Sequestration, red blood cell destruction by
spleen → hypersplenism
DIAGNOSIS
LAB RESULTS
Giemsa stain blood smear
▪ Thin: rapid, ↓ microscopy experience
required
▪ Thick: ↑ accurate, ↑ microscopy experience
required
▪ P. falciparum: red blood cells contain
multiple parasites, ring forms, lack of
schizonts
▪ Banana-shaped gametocyte form
108
Rapid diagnostic test
▪ 15–20 minute results
▪ Histidine-rich protein 2: detects P.
falciparum
PCR
▪ Supportive laboratory results
(nondiagnostic)
▫ Normocytic hemolytic anemia
▫ Thrombocytopenia
TREATMENT
MEDICATIONS
▪ Severe malaria
▫ Quinidine + doxycycline/tetracycline/
clindamycin/artesunate (test for G6PD
deficiency)
▪ Uncomplicated malaria
▫ Chloroquine-sensitive strain:
chloroquine phosphate/
hydroxychloroquine (blocks Plasmodium
heme polymerase)
▫ Chloroquine-resistant strain:
atovaquone-proguanil, mefloquine,
artesunate, quinine-based regimens
▪ P. ovale, P. vivax: + primaquine for
hypnozoite (test for G6PD deficiency)
OTHER INTERVENTIONS
Figure 77.2 A histological section of the
placenta from an individual infected with
malaria. The numerous black dots within the
red blood cells are malaria organisms.
Prevention
▪ Chemoprophylaxis
▪ Avoid outdoors (dusk/dawn)
▪ Reduce exposed skin (long pants, shirts)
▪ Bed netting/mosquito nets
▪ Insect repellent (DEET 30–50%)
109
NOTES
NOTES
HEPADNAVIRIDAE
MICROBE OVERVIEW
▪ Hepadnaviridae: family of DNA viruses,
causes liver disease
▪ Produces DNA polymerase with reverse
transcriptase, RNAse activity
Replication/multiplication
▪ Reverse transcription (RNA intermediate)
Structure
▪ Enveloped
▪ Icosahedral capsid
▪ Partially double stranded, partially single
stranded circular DNA
COMPLICATIONS
▪ Chronic hepatitis, acute liver failure
(fulminant hepatitis), liver cirrhosis,
hepatocellular carcinoma (HCC)
HEPATITIS B VIRUS
osms.it/hepatitis-b-virus
PATHOLOGY & CAUSES
▪ Hepatitis B virus (HBV)
▫ DNA virus: can cause acute/chronic liver
disease
▫ Member of Hepadnaviridae family (only
human pathogenic species)
▪ Target: hepatocytes, zone I (periportal area)
▪ Incubation period: six weeks to six months
Antigens
▪ Hepatitis B surface antigen (HBsAg/
Australia antigen)
▫ Key infection marker
▪ Hepatitis B core antigen (HBcAg)
▫ Not detectable in serum; found in liver
with acute/chronic hepatitis B
▪ Hepatitis B e antigen (HBeAg)
▫ Secreted by infected cells; indicates
active infection, replication
Transmission
▪ Perinatal (childbirth), parenteral (e.g. IV
drug use, blood transfusions), sexual
▪ HBV survives ≥ seven days in environment
Highest prevalence
▪ Parts of sub-Saharan Africa, mainly due to
perinatal transmission
▪ Lower prevalence areas have greater
association with parenteral, sexual
transmission
TYPES
Acute hepatitis
▪ Liver inflammation for < six months)
▪ HBV penetrates hepatocytes → CD8+
T-lymphocyte activation → cytotoxic killing
→ hepatocyte apoptosis → liver damage,
inflammation
▪ Phases
▫ Early, window (antigens undetectable),
recovery
110
▪ Acute liver failure progression: < 1%
▪ Chronic hepatitis progression (in adults):
< 5%
▪ Acute liver failure
▫ Acute liver injury (severe hepatocyte
necrosis), hepatic encephalopathy,
coagulopathy
▫ Excessive immune response → massive
hepatocyte lysis
▫ Preexisting liver disease absence
▫ Associated with HBV genotype D
Chronic hepatitis
▪ HBsAg persistent for > six months
▪ Pathogenesis
▫ HBV penetrates hepatocytes →
insufficient CD8+ T-lymphocyte
activation → “immune tolerance” →
HBV persistence
▪ Progression to HCC
▫ HBV integrates to host DNA →
oncogene activation → oncogenesis
▪ Chronicity depends highly on age at time of
infection
▫ Younger age, ↑ chronicity risk
▫ Immunosuppressed, elderly people also
more susceptible
▪ Phases
▫ Immune tolerant: no liver inflammation/
fibrosis
▫ Immune active: liver damage,
inflammation, possible fibrosis
▫ Immune inactive: ↓ inflammation
▫ Reactivation: liver damage,
inflammation, possible fibrosis
▫ “Recovery”: occult HBV, HBsAg
negative; still infected, disease inactive
(best prognosis)
RISK FACTORS
▪ IV drug use
▪ Healthcare workers
▫ Frequent contact with blades, needles,
body fluids
▪ High-risk sexual behavior
▪ Anal intercourse
▪ Previous HIV/hepatitis C infection
COMPLICATIONS
▪ HCC, fulminant hepatitis, liver cirrhosis
Hepatic encephalopathy
▪ Excessive nitrogen load, electrolyte
disturbances → altered neurologic
functions in individuals with severe liver
disease
Hepatorenal syndrome
▪ Portal hypertension → splanchnic
vasodilation → ↓ effective circulatory
volume → ↑ renin-angiotensin-aldosterone
system → renal vasoconstriction →
hepatorenal syndrome (liver dysfunction →
kidney failure)
Bleeding diathesis
▪ Hypocoagulability → ↑ hemorrhage risk
SIGNS & SYMPTOMS
Acute hepatitis
▪ Can be anicteric (not accompanied by
jaundice) with non-specific symptoms (e.g.
fever, malaise, nausea, vomiting)
▪ Some progress to icteric hepatitis with
hepatomegaly, right upper-quadrant pain,
jaundice (30%), dark-colored urine (due to
conjugated hyperbilirubinemia), pale stool
Chronic hepatitis
▪ Mostly asymptomatic/non-specific
symptoms until late disease stages
▪ Exacerbations may present as acute
hepatitis
▪ Jaundice, ascites, splenomegaly,
encephalopathy (e.g. personality changes,
↓ level of consciousness, intellectual
impairment, asterixis) may present
▪ Extrahepatic manifestations (occasionally)
▫ Fever, rash, arthralgia, arthritis,
glomerulonephritis
DIAGNOSIS
LAB RESULTS
▪ HBV DNA detection
▫ Polymerase chain reaction (PCR), in-situ
hybridization, Southern hybridization
111
Chapter 78 Hepadnaviridae
Laboratory testing
▪ ↑ alanine aminotransferase (ALT), aspartate
aminotransferase (AST)
▫ ALT > AST
▫ Usually ALT in acute hepatitis > 1000
U/L (may be ↓ in chronic hepatitis)
▫ ALT levels take longer than AST to
return to normal
▪ ↑ ALT for > six months indicates chronicity
▪ ↑ alpha-fetoprotein (AFP) in HCC
▪ Liver fibrosis
▫ ↓ leukocytes, platelets
▫ AST/ALT > 1 (normal ≈ 0,8)
▫ ↑ total bilirubin
▫ ↓ serum albumin
▫ Delayed prothrombin time, ↑
international normalized ratio (INR)
Serologic marker detection through enzyme immunoassay
▪ Hepatitis B surface antigen (HBsAg)
▫ Indicates infection (acute/chronic)
▪ Hepatitis B surface antibodies (Anti-HBs)
▫ Provides HBV infection immunity
appears after vaccination/resolved acute
hepatitis
▪ IgM antibodies against hepatitis B core
antigen (IgM anti-HBc)
▫ Acute infection/chronic hepatitis
reactivation phase
▪ IgG antibodies against hepatitis B core
antigen (IgG anti-HBc)
▫ Non-specific antibody; may be ↑ during
acute, resolved, chronic hepatitis
▪ Hepatitis B e antigen
▫ Active replication, high infectivity
▪ Hepatitis B e antibodies
▫ Low replication, infectivity
Liver biopsy
▪ Acute hepatitis
▫ Mononuclear infiltrate
▫ Pericentral inflammation, necrosis
▫ Eosinophilic hepatocytes
▪ Chronic hepatitis
▫ Fibrosis
▫ Nodule formation
▫ Mononuclear portal infiltrate
▫ Some hepatocytes have uniformly dull
cytoplasm due to endoplasmic reticulum
swelling (“ground glass” hepatocytes)
112
Figure 78.1 Ground glass hepatocytes seen
in the liver of an individual with hepatitis B
infection.
TREATMENT
MEDICATIONS
▪ Antiviral monotherapy
▫ Severe acute hepatitis, pre-existing
liver disease, concomitant hepatitis C/D
infection, immunocompromised, elderly
Acute hepatitis
▪ Post-exposure prophylaxis
▫ HBV vaccine and immunoglobulin
Prevention
▪ HBV vaccine
▪ Recombinant type most commonly used
▫ HBsAg inserted in yeast cells
▪ HBsAg → development of anti-HBsAg →
HBV infection immunity
▪ Intramuscular administration
▪ Three doses for coverage (very effective)
▪ Administration regime: 0, 1–2 months,
6–12 months
▫ Infant immunization: first dose at birth
▫ Does not require booster dose (longterm protection)
SURGERY
▪ Acute liver failure
▫ Consider liver transplantation
OTHER INTERVENTIONS
▪ Acute liver failure
▫ Fluid resuscitation, early nutritional
support, antiviral therapy (nucleoside/
nucleotide analogues)
Acute hepatitis
▪ Supportive treatment (e.g. fluid therapy,
nutrition)
Chronic hepatitis
▪ Combination therapy (e.g. lamivudine,
interferon)
113
Chapter 78 Hepadnaviridae
HEPATITIS D VIRUS
osms.it/hepatitis-d-virus
PATHOLOGY & CAUSES
SIGNS & SYMPTOMS
▪ Hepatitis D virus (HDV/delta virus):
incomplete RNA virus; contributes to
acute liver failure development, chronic
hepatitis exacerbation in people coinfected/
previously infected with HBV
▪ HDV not member of Hepadnaviridae family
▪ HDV infection inhibits HBV replication
due to HBV, HDAg interaction during viral
replication
▫ Coinfected individuals: HDV
predominant
▫ ↑ inflammatory response (compared to
HBV alone)
▫ Poor response to existing HBV
treatment
▪ Incubation period
▫ 6–24 weeks
Coinfection
▪ Biphasic course with acute hepatitis
symptoms
Structure
▪ Outer envelope made of HBsAg, inner HDV
RNA, delta antigen (HDAg)
LAB RESULTS
Transmission
▪ Parenteral, sexual, perinatal (very rare)
Satellite virus
▪ Can only infect if host also infected with
HBV
▫ Coinfection: simultaneous infection
▫ Superinfection: HDV infection after
established HBV infection; more severe
RISK FACTORS
▪ IV drug use, high-risk sexual behavior, HBV
presence
Acute liver failure
▪ Systemic symptoms
▫ E.g. fever, malaise, nausea, vomiting)
▫ Hepatomegaly, right upper quadrant
pain; sometimes jaundice, dark colored
urine, pale stool
▪ Hepatic encephalopathy
▫ Personality changes, ↓ level of
consciousness, intellectual impairment,
asterixis
DIAGNOSIS
▪ Serologic marker detection
▫ IgM/IgG anti-HDV
▪ PCR assays
▫ HDV RNA detection
TREATMENT
MEDICATIONS
▪ Pegylated interferon alpha
▪ Prevention
▫ HBV vaccine
SURGERY
▪ Consider liver transplantation for chronic
hepatitis D, acute liver failure
114
AfraTafreeh.com exclusive
NOTES
NOTES
HERPESVIRUSES
MICROBE OVERVIEW
▪ Large family of DNA viruses
Species known to infect humans
▪ Herpes simplex virus (HSV)
▫ HSV-1, HSV-2
▪ Varicella-zoster virus (VZV)
▪ Epstein-Barr virus (EBV)
▪ Human cytomegalovirus
▪ Human herpesvirus 6 (roseola)
▪ Human herpesvirus 8 (Kaposi’s sarcoma)
Genetic material
▪ Double-stranded, linear DNA genome
encoding 84 proteins
Morphology
▪ Icosahedral capsid; enveloped virus
Life cycle
▪ Obligate intracellular parasites
▪ Can perpetuate in latent phase
▪ Expression of lytic genes → lytic phase→
host cell death → shedding
CYTOMEGALOVIRUS
osms.it/cytomegalovirus
PATHOLOGY & CAUSES
▪ Primary infection of immunocompetent
individuals usually asymptomatic; lifelong
latency
▪ Infects mononuclear leukocytes, endothelial
cells
CAUSES
▪ Person-to-person transmission
▫ Kissing; intimate, sexual contact
▪ Vertical transmission
▫ Congenital infection
▪ Blood products/transfusion
▪ Organ/stem cell transplant
RISK FACTORS
COMPLICATIONS
▪ Congenital CMV infection
▫ Cognitive, sensorineural deficits
SIGNS & SYMPTOMS
▪ Usually asymptomatic
▪ Fever, malaise, sore throat, splenomegaly
▪ Individuals with AIDS
▫ Retinitis, colitis, encephalitis,
pneumonitis
▪ Individuals with congenital CMV infection
▫ Positive CMV-IgG (if pregnant);
intrauterine fetal growth restriction
(IUGR), hydrocephalus, microcephaly,
intracranial calcification, poly/
oligohydramnios, hepatosplenomegaly
▪ Immunocompromised
▫ HIV/AIDS; organ transplant; medications
(e.g. steroids, chemotherapy)
115
Chapter 79 Herpesviruses
DIAGNOSIS
LAB RESULTS
▫ Highly active antiretroviral therapy
(HAART)
▫ CMV immunoglobulin G (CMV-IGIV)
▪ Complete blood count
▫ Atypical lymphocytosis, anemia,
leukopenia, thrombocytopenia
▪ ↑ serum creatinine, AST, ALT
▪ Serology tests
▫ ↑ CMV-IgM titre; acute infection
▫ ↑ CMV-IgG titre; past infection
TREATMENT
▪ Usually self-limiting
MEDICATIONS
▪ At-risk individuals
▫ Antiviral prophylaxis; IV ganciclovir; oral
valganciclovir
Figure 79.1 A colonic biopsy taken from an
individual with CMV colitis. The large nuclei
that give the virus its name can be clearly
seen.
EPSTEIN–BARR VIRUS
(INFECTIOUS MONONUCLEOSIS)
osms.it/epstein-barr_virus
PATHOLOGY & CAUSES
▪ EBV (human herpesvirus 4)
▪ Causes Infectious mononucleosis (glandular
fever)
▪ Infection in children
▫ Asymptomatic/mild flu-like symptoms
▪ Infection in adolescents, young adults
▫ Fever, sore throat, enlarged lymph
nodes
▪ Lytic, latent life cycle
▫ Lytic in oropharyngeal B cells; latent in
lymphocytes
CAUSES
▪ Transmitted through saliva (“kissing”
disease), sexual transmission
RISK FACTORS
▪ Multiple sexual partners
COMPLICATIONS
▪ Splenic rupture
▪ EBV infection associated with Hodgkin’s,
Burkitt’s lymphoma
▪ Neurologic syndromes (2–4 weeks after
initial symptoms)
▫ Guillain-Barré syndrome, cranial
nerve palsies (e.g. facial nerve palsy);
meningoencephalitis (e.g. aseptic
meningitis), transverse myelitis,
peripheral neuritis, optic neuritis
▪ EBV can affect, manifest in any organ
system
▫ Hepatitis, pneumonia, myocarditis,
pancreatitis, acute renal failure, gastric
pseudolymphoma
116
▪ Classic triad
▫ Fever; cervical/generalized
lymphadenopathy; tonsillar pharyngitis
(exudative/non-exudative)
▪ Malaise, hepatosplenomegaly, rash,
jaundice, myalgia
morphology (large, irregular nuclei)
▪ ↑ hepatic transaminase (50% of infected
individuals)
▪ Monospot test
▫ Positive heterophile antibodies (IgM)
▪ Identification of EBV
▫ EBV-specific antibodies, real-time PCR,
EBV DNA detection
DIAGNOSIS
TREATMENT
SIGNS & SYMPTOMS
MEDICATIONS
LAB RESULTS
▪ Complete blood count (CBC)
▫ Lymphocytosis (50%) with atypical
▪ Antipyretics, analgesics
▫ Avoid aspirin; may lead to Reye’s
syndrome
▪ Corticosteroid for upper airway obstruction
(e.g. prednisolone)
▪ Intravenous immunoglobulin (IVIG) for
thrombocytopenia
Figure 79.2 Atypical lymphocytes with
bizarre nuclear forms in a peripheral
blood film from an individual infected with
infectious mononucleosis.
HERPES SIMPLEX VIRUS
osms.it/herpes-simplex-virus
PATHOLOGY & CAUSES
▪ HSV-1, HSV-2
▫ Members of herpesviridae family
▪ Causes oral, genital, ocular ulcers
▪ Portal of entry
▫ Mucosal surfaces/skin breaks; vertical
transmission during pregnancy/
childbirth
▪ Primary infection often symptomatic;
replicates in epidermis of skin → travels
down nerve endings, axon→ sensory
ganglia
▪ Periodic reactivation, subsequent episodes
▫ Virus becomes active in ganglia
→ transported via axon to skin →
replicates in epidermis → sheds → new
sores
▫ Often asymptomatic shedding; may feel
tingling, burning sensation
117
Chapter 79 Herpesviruses
TYPES
▪ Infection types
▫ Primary: first infections; seronegative;
symptoms more extensive, systemic;
greater viral shedding loads
▫ Non-primary: previously infected
individuals; serum antibody, humoral
immunity (e.g. genital HSV-2 infection
in adulthood after oral mucosa HSV-1
infection in childhood)
▫ Recurrent: previously infected
individuals; reactivation episodes (e.g.
genital HSV-2 infection in adulthood
after genital HSV-2 infection in
adolescence)
▪ Herpes labialis (AKA oral herpes)
▫ HSV-1 infection of oral mucosa, lips
▪ Genital herpes
▫ HSV-2, HSV-1 infection of genital area
▫ Healing takes 2–4 weeks
SIGNS & SYMPTOMS
▪ Herpes labialis
▫ Painful ulcers around mouth; high
fever; sore throat; pharyngeal oedema;
myalgia, cervical lymphadenopathy
▫ Recurrent infection: pain, burning,
tingling, vesicle formation
▪ Genital herpes
▫ Genital ulceration, vesicles: vulva,
cervix, vagina, penis shaft/glans,
perineum, buttocks
▫ Genital pain, dysuria, fever, neuralgia
▫ Constipation, rectal pain, tenesmus,
proctitis
CAUSES
▪ Portal of entry
▫ Mucosal surfaces/skin breaks; vertical
transmission during pregnancy/
childbirth
▪ Spread person to person; sexual
transmission
RISK FACTORS
▪ Genital herpes
▫ Contact with infected individual
(producing, shedding virus)
▫ Immunosuppression (e.g. medications,
HIV/AIDS)
▫ High-risk sexual behavior (e.g
multiple sexual partners, unprotected
intercourse, first sexual activity at early
age)
COMPLICATIONS
▪ Neonatal HSV infection, meningitis,
encephalitis, acute retinal necrosis, uveitis,
keratitis, esophagitis
Figure 79.3 Blisters on the lips caused by
infection with herpes simplex virus.
DIAGNOSIS
LAB RESULTS
▪
▪
▪
▪
Viral culture
HSV polymerase chain reaction (PCR)
Direct fluorescent antibody (DFA) test
Serological HSV-1/HSV-2 specific IgG
assay
118
TREATMENT
MEDICATIONS
Herpes labialis
▪ Antivirals (e.g. oral aciclovir, valaciclovir,
famciclovir)
▪ Topical antivirals
Genital herpes
▪ Antivirals (e.g. oral aciclovir, valaciclovir,
famciclovir)
▪ Pregnant: antiviral prophylaxis
OTHER INTERVENTIONS
Genital herpes
▪ Pregnant: possible cesarean delivery
Figure 79.4 Blisters on the dorsum of the
penis of an individual infected with genital
herpes simplex.
HUMAN HERPESVIRUS 6
(ROSEOLA)
osms.it/human-herpesvirus-6
PATHOLOGY & CAUSES
▪ Causes roseola (infantum)
▪ Common cause of fever, followed by rash
in early childhood, associated with febrile
seizures
▪ Incubation period
▫ 1–2 weeks
CAUSES
▪ Asymptomatic contacts
▫ Person-to-person spread by respiratory
secretions
▪ Trophic for CD4+ T lymphocytes
▪ Following acute infection, remains latent in
various tissues
RISK FACTORS
▪ More common in young children (six
months–two years)
▪ Immunosuppressed
▫ Bone marrow/organ transplant
▫ Reactivation of latent virus
COMPLICATIONS
▪ Afebrile seizures
▪ Associated with chronic fatigue syndrome,
multiple sclerosis, systemic lupus
erythematosus
119
Chapter 79 Herpesviruses
SIGNS & SYMPTOMS
▪ 3–5 days of high fever; peak in early
evening
▪ Exanthem; morbilliform rash appears with
defervescence; 3–5mm pink/red macules,
papules along trunk, extremities
▪ Diarrhea, tympanic membrane
inflammation, upper respiratory symptoms
▪ Nagayama’s spots: enanthem of red
papules on soft palate, uvula
▪ Rare
▫ Seizures, periorbital oedema, bulging
anterior fontanelle, cervical/occipital/
postauricular lymphadenopathy
TREATMENT
▪ Usually self-limiting
MEDICATIONS
▪ Immunocompromised
▫ Antiviral compounds (e.g. acyclovir,
ganciclovir, cidofovir, foscarnet)
▪ Oral hydration, antipyretics (e.g.
paracetamol, ibuprofen)
▪ Avoid aspirin; may lead to Reye’s syndrome
DIAGNOSIS
LAB RESULTS
▪ Complete blood count
▫ ↓ white blood cells
▪ Detection of HHV-6B/HHV-7
▫ Viral culture, specific IgG detection
Figure 79.5 A child with roseola infantum,
also known as sixth disease. The most
common causative agent is human herpes
virus six.
HUMAN HERPESVIRUS 8
(KAPOSI'S SARCOMA)
osms.it/human-herpesvirus-8
PATHOLOGY & CAUSES
▪ Causes Kaposi’s sarcoma
▫ Low-grade vasoformative neoplasm
associated with human herpesvirus-8
(HHV-8) infection/Kaposi’s sarcoma
herpesvirus (KSHV) infection
▪ Lesions on mucocutaneous areas; may
involve lymph nodes, viscera
▪ Skin lesions
▫ Patch → plaque → ulcerating tumor
nodules
▪ Morphology
▫ spindle-shape, vasoformative cells with
vascular proliferation, inflammatory cells
120
▪ Spontaneous regression may occur after
HAART/immunosuppressive treatment
TYPES
▪ Epidemic AIDS-associated Kaposi’s
sarcoma
▫ HIV-positive individuals
▪ Classic sporadic Kaposi’s sarcoma
▫ More common in individuals who are
male, older, with Mediterranean/Jewish
background
▪ Iatrogenic, transplant-associated Kaposi’s
sarcoma
▫ Solid organ transplant individuals
▪ African endemic Kaposi’s sarcoma
▫ More common in central Africa,
unrelated to HIV
RISK FACTORS
▪ More common in individuals who are
biologically male, > 50 years
▪ Immunosuppression, HIV (co-infection
synergistic → aggressive, widespread);
drug abuse; immunosuppression therapy
COMPLICATIONS
▪ Secondary skin lesion infection
SIGNS & SYMPTOMS
▪ Skin lesions
▫ Multifocal; asymmetrically distributed;
size, colour variation; non-pruritic;
papular; nodular; plaque-, bullous-like;
indurated; hyperkeratotic
▪ Oral lesions
▫ Hard palate, gingiva, dorsum of tongue
▫ Macules, papules, nodules, exophytic
masses
▪ Lymphadenopathy, lymphoedema
▪ Fever, weight loss, night sweats
Figure 79.6 The skin of an individual with
Kaposi’s sarcoma.
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Nodular/interstitial/alveolar infiltrates; hilar/
mediastinal lymphadenopathy; nodules
LAB RESULTS
▪ HIV test
▪ Positive in AIDS-associated Kaposi’s
sarcoma
▪ Fecal occult blood
▫ Positive may indicate intestinal lesions
OTHER DIAGNOSTICS
▪ Lesion, lymph node biopsy of vascular
lesion
TREATMENT
MEDICATIONS
▪ Delay disease progression
▫ HAART; in HIV infected individuals,
HIV suppression may shrink
Kaposi’s sarcoma lesions; systemic
chemotherapy
▪ Cosmetic
▫ Topical retinoids; intralesional
vinblastine
121
Chapter 79 Herpesviruses
SURGERY
▪ Cosmetic
▫ Surgical excision; cryotherapy; laser
therapy (external beam radiation)
Figure 79.7 The histological appearance of
a Kaposi sarcoma. The tumor is composed
of spindle cells and numerous branching
vascular spaces. The occasional spindle cell
contains hyaline globules.
VARICELLA ZOSTER VIRUS
osms.it/varicella-zoster-virus
PATHOLOGY & CAUSES
▪ Double-stranded, linear DNA virus
▫ Causes chickenpox, shingles
▫ Chickenpox: generally benign, selflimited disease in immunocompetent
children
▫ Shingles: painful skin rash, can occur in
individuals who have recovered from
primary VZV infection
Primary infection (chickenpox)
▪ Transmission: airborne spread through
aerosolized droplets, direct lesion contact
▪ VZV enters respiratory system → lymph
node spread → targets skin, mucous
membranes → small blood vessel vasculitis
→ epithelial cell degeneration → fluid-filled
vesicles
▫ Incubation period: 14 days
▫ Most infectious: 1–2 days before rash;
infectious 3–4 days, until lesions crusted
▫ After symptoms resolve, VZV dormant
in nervous system; latent in trigeminal,
dorsal root ganglia
Figure 79.8 A child with chickenpox.
122
Secondary infection (shingles)
▪ Occurs when dormant VZV reactivates
▫ Reactivation in dorsal root, cranial
nerve ganglia → travels down axons →
local skin inflammation innervated by
ganglion
▫ Prodromal 2–4 day tingling/localized
pain before rash onset
RISK FACTORS
Chickenpox
▪ More common in children
▫ 1–9 years old (highest risk)
▪ Non-immunized status
▪ Immunocompromised status
Shingles
▪ Primary varicella infection history
▪ More common in adults
▫ > 50 years old (highest risk)
▪ Immunocompromised status
▪ Stress
SIGNS & SYMPTOMS
Chickenpox
▪ Fever, headache, malaise, sore throat,
tachycardia
▪ Rash characteristics
▫ Generalized pruritic, vesicular rash
▫ “Dew drop on rose petal” appearance
▫ Formation: macules → vesicles →
rupture → scab
▫ Vesicles on mucous membranes (e.g.
nasopharynx, conjunctiva, mouth, vulva)
Shingles
▪ Erythematous, maculopapular lesions
evolve into painful vesicular rash
▫ Rash follows dermatomal distribution of
cranial nerve/dorsal root ganglion
▫ Thoracic, lumbar dermatomes most
commonly affected
COMPLICATIONS
Chickenpox
▪ Secondary bacterial infection, cutaneous
scarring, encephalopathy, varicella
pneumonitis/pneumonia
▪ Central nervous system (CNS)
complications
▫ Meningitis, encephalitis, intracranial
vasculitis
▪ Congenital varicella syndrome
▫ Limb hypoplasia, paresis, microcephaly,
ophthalmic lesions
Shingles
▪ Ramsay Hunt syndrome
▫ VZV of geniculate ganglion affects facial
nerve; hearing loss, facial weakness
▪ Postherpetic neuralgia, superinfection
of skin lesions, encephalitis, Mollaret’s
meningitis, zoster multiplex/sine herpete,
stroke, myelitis
▪ Herpes zoster ophthalmicus (sightthreatening)
Figure 79.9 Herpes zoster, or shingles,
affecting the ophthalmic branch of the
trigeminal nerve.
123
Chapter 79 Herpesviruses
TREATMENT
MEDICATIONS
Chickenpox
▪ Low risk: usually self-limiting
▪ Moderate risk: oral antiviral therapy
▪ High risk: intravenous antiviral therapy;
zoster-immune globulin (ZIG)
Figure 79.10 Herpes zoster in a dermatomal
distribution on the chest.
DIAGNOSIS
LAB RESULTS
▪ Microbe identification
▫ PCR: VZV DNA
▫ Viral culture: positive VZV
▫ DFA: VZV antigen
Shingles
▪ Oral/intravenous antiviral therapy
▪ Analgesics
▫ E.g. paracetamol, ibuprofen; topical;
opioid
▪ Calamine lotion
▪ Varicella-zoster immune globulin
OTHER INTERVENTIONS
▪ Prevention
▫ Live attenuated VZV vaccine
124
NOTES
NOTES
LEISHMANIA
MICROBE OVERVIEW
▪ Flagellated, parasitic protozoan
▪ Contains kinetoplast (form of mitochondrial
DNA)
▪ Hosts: humans, canids, rodents, hyraxes
Classification
▪ “Old World”
▫ Usually found in Africa, Asia, Middle
East, India
▫ Includes Leishmania infantum, L.
donovani, L. tropica, L. aethiopica, L.
major
▪ “New World”
▫ Usually found in Central, South America;
Mexico
▫ Includes L. braziliensis, L. mexicana, L.
infantum (chagasi), L. amazonensis, L.
panamensis, L. guyanensis
Life stages
▪ Amastigote
▫ Oval; 3–6µm
▫ Intracellular, nonmotile form (no external
flagella)
▫ Found in human circulatory system in
mononuclear phagocytes
▪ Promastigote
▫ Spindle-shaped; 15–30µm
▫ Extracellular, motile form (long external
flagellum)
▫ Found in alimentary tract of sandflies
LEISHMANIA
osms.it/leishmania
PATHOLOGY & CAUSES
▪ Causes disease leishmaniasis
▪ Spread by female sandflies of genus
Lutzomyia/Phlebotomus
▪ Human leishmaniasis characterized by
ulcers (e.g. skin, oral, nasal mucosa),
systemic illness
▪ Lipophosphoglycan layer helps it survive
immune system
TYPES
Cutaneous leishmaniasis (CL)
▪ Most common
▪ Usually caused by L. major, L. tropica, L.
aethiopica
Mucocutaneous leishmaniasis (ML)
▪ Causes mucosal, skin ulcers
▪ Usually caused by L. braziliensis
Visceral leishmaniasis (VL)
▪ AKA kala-azar
▪ Most severe, systemic involvement
▪ Usually caused by L. donovani, L. infantum
▪ If recurs post-treatment: post kala-azar
leishmaniasis
▪ Resists host’s complement system,
prevents natural killer cells
▪ Infected macrophages spread infection
▫ Spleen → splenomegaly
▫ Liver → Kupffer cells with increased size
125
Chapter 80 Leishmania
→ liver dysfunction
▫ Bone marrow → hyperplastic → ↓
hematopoiesis → pancytopenia
▫ Lymph nodes → lymphadenopathy
RISK FACTORS
▫ Poverty, malnutrition, poor hygiene;
deforestation, urbanization
SIGNS & SYMPTOMS
CL
▪ Skin ulcers resembling leprosy lesions
ML
▪ Ulcers on oropharyngeal, nose mucosa
▪ Midfacial destruction: nose cartilage/septal
granulation, tear; ulcers on palate, uvula,
lips
▪ Hoarse voice, gingivitis, periodontitis
▪ Genital mucosal (severe cases)
VL
▪ Fever, weight loss, hepatosplenomegaly,
pancytopenia, hypergammaglobulinemia,
abdominal tenderness
TREATMENT
MEDICATIONS
CL
▪ Topical paromomycin, miltefosine,
pentamidine isethionate, fluconazole,
ketoconazole
ML
▪ Liposomal amphotericin B, miltefosine,
pentamidine
VL
▪ Liposomal amphotericin B, miltefosine
OTHER INTERVENTIONS
Prevention
▪ Nets treated with insecticide while
sleeping; over doors, windows
▪ Insect repellents, insecticide-impregnated
dog collars
▪ Treat infected dogs
DIAGNOSIS
LAB RESULTS
▪ Enzyme-linked immunosorbent assay
(ELISA)
▪ Antigen-coated dipsticks
▪ Direct agglutination test
Light microscope
▪ Amastigotes/Leishman-Donovan body
visualization: blood, aspirates from marrow,
spleen, lymph nodes, skin lesions
▫ Blood, spleen monocytes
▫ Circulating neutrophils, aspirated tissue
macrophages
Figure 80.1 A leishmaniasis ulcer on the leg.
Polymerase chain reaction (PCR)
▪ Detects Leishmania kinetoplast DNA (most
sensitive, specific)
126
NOTES
NOTES
MYCOPLASMA
MICROBE OVERVIEW
▪ Smallest free living organisms
▪ Prokaryotic with absence of cell wall,
presence of flexible cell membrane
containing cholesterol
▫ Pleomorphic
▫ Not visible on Gram stain
▫ Resistant to beta lactam, glycopeptide
antibiotics
▪ Limited metabolic activity → not culturable
on standard culture media, require
specialized medium (e.g. Eaton’s agar) with
sterols, nutrients provided by natural animal
protein (e.g. blood serum)
▪ Can grow under aerobic, anaerobic
conditions
MYCOPLASMA PNEUMONIAE
osms.it/mycoplasma-pneumoniae
PATHOLOGY & CAUSES
▪ Species of mycoplasma; primarily affects
respiratory tract; usually causes upper
respiratory tract infections; can also cause
atypical pneumonia
▪ Transmitted through respiratory droplets
after close contact with infected individual
→ attaches to respiratory epithelium
with P1 surface protein → hydrogen
peroxide, superoxide radicals synthesized
by mycoplasma interact with endogenous
toxic molecules synthesized by host cells →
oxidative stress in respiratory epithelial cells
▪ Macrophages migrate to site of infection
→ activation and phagocytosis → initiate
inflammatory response → T, B lymphocyte
proliferation → antibody production, release
of inflammatory cytokines → control
infection/initiate immune-mediated lung
injury
▪ Extrapulmonary disease (rare)
▫ Due to immune mediated injury/cross
reactive antibody mechanism/direct
invasion
▪ Central nervous system (CNS), joints, skin,
blood, heart, liver, pancreas affected
▫ CNS is the most common
extrapulmonary site; usually encephalitis
▪ Can develop cold agglutinin response
(60%)
▫ Autoimmune hemolytic anemia
▫ Coombs positive
▫ Cold (active below 37°C/98.6°F) IgM
antibodies against erythrocyte surface
antigen due to cross reaction of antigen
with mycoplasma antigens → can
agglutinate/lyse erythrocytes
RISK FACTORS
▪ Common in children, young adults
▪ Immunocompromised status, smoking,
close community living (e.g. nursing homes,
dorms)
COMPLICATIONS
▪ Asthma exacerbations
▪ Respiratory failure
▪ CNS involvement
127
Chapter 81 Mycoplasma
▫ Encephalitis with high mortality rate
▪ Heart involvement
▫ Rhythm disorders, heart failure
SIGNS & SYMPTOMS
▪ Can be asymptomatic
▪ Gradual onset
▫ General fatigue, myalgias, headache,
low grade fever, sore throat, cough
(worsening, frequent, non-productive),
chills
▪ Less common
▫ Sinus/ear pain, wheezing
▪ Chest auscultation
▫ Scattered rales, wheezes, rhonchi,
crackles
▪ Sinus tenderness
▪ Erythema of tympanic membrane
▪ Pharyngeal erythema
▪ Pulse-temperature dissociation: normal
pulse despite fever indicative of atypical
pneumonia
DIAGNOSIS
LAB RESULTS
▪ Molecular testing with polymerase chain
reaction (PCR); most accurate
▪ Serological tests
▫ ≥ four-fold rise in IgM antibodies titers
of acute, convalescent sera 2–3 weeks
apart using enzyme immunoassay
▫ High titer of IgM antibodies
▫ Cold agglutinins titer
▪ Isolation with culture
▫ Limited use due to slow growth (2–3
weeks), need for specialized media
DIAGNOSTIC IMAGING
Chest X-ray/CT scan/high resolution CT
scan
▪ Diffuse reticulonodular pattern indicative of
interstitial pneumonia
▪ Areas of airspace consolidation (esp. lower
lobes)
▪ Thickening of bronchovascular bundle
OTHER DIAGNOSTICS
▪ Histopathology
▫ Inflammation in trachea, bronchioles,
peribronchial tissues
▫ Airspaces filled with purulent exudate
with polymorphonuclear cells
▪ Physical examination
▫ Vague symptoms (e.g. fatigue) indicative
of atypical/“walking” pneumonia
TREATMENT
▪ Most cases mild, self-limited
MEDICATIONS
▪ Atypical pneumonia
▫ Macrolides (e.g, erythromycin,
azithromycin); tetracyclines (e.g.
doxycycline); fluoroquinolones (e.g.,
levofloxacin, moxifloxacin)
128
NOTES
NOTES
NEMATODES (ROUNDWORMS)
GENERALLY, WHAT ARE THEY?
PATHOLOGY & CAUSES
▪ AKA roundworms
▫ Microfilariae: immature worms
▫ Microfilariae: mature worms
▪ Parasites usually enter body cutaneously
▫ Produce gastrointestinal (GI),
occasionally cutaneous symptoms,
disease
COMPLICATIONS
▪ Loffler syndrome
▫ Transient pulmonary disease during
select nematode development (Necator
Americanus, Strongyloides, Ascaris
Lumbricoides, Toxocara, Ancylostoma
duodenale)
▫ Symptoms: irritating, nonproductive
cough, substernal burning discomfort,
dyspnea, wheezing, fever, blood-tinged
sputum production
SIGNS & SYMPTOMS
TREATMENT
MEDICATIONS
▪ Antihelminthic therapy
▫ Nematode location-dependent
Intestinal nematodes
▪ Mebendazole/albendazole
▫ Bind tubulin colchicine-sensitive sites
→ inhibit microtubule assembly →
inhibited glucose uptake (without
human effect) → organism death
Systemic nematodes
▪ Diethylcarbamazine (DEC)
▫ Piperazine derivative (uncertain
mechanism of action)
▪ Ivermectin
▫ Synthetic lactone → negative-charged
ion influx via glutamate-sensitive
chloride channels → hyperpolarizes cells
→ helminth cell death
▫ Not macrofilaricidal, pregnancy
contraindication
▪ GI/localized tissue symptoms, organismdependent
DIAGNOSIS
LAB RESULTS
▪ Eosinophilia
▪ Gammaglobulinemia abnormalities (↑/↓
depending on organism)
Serology
▪ IgG levels
▪ Enzyme linked immunosorbent assays
(ELISA), western blot
129
Chapter 82 Nematodes (Roundworms)
ANCYLOSTOMA DUODENALE &
NECATOR AMERICANUS
osms.it/ancylostoma-duodenale
osms.it/necator-americanus
PATHOLOGY & CAUSES
▪ Infectious hookworm species
Adult morphology
▪ Ancylostoma duodenale
▫ Two ventral plates on buccal capsule’s
anterior margin; two large teeth fused at
base (male 8–11mm, female 10–13mm
long)
▪ Necator americanus
▫ Two dorsal/ventral cutting plates
around buccal capsule’s anterior margin,
paired subdorsal/subventral teeth (male
7–9mm, female 9–11mm long)
▪ A. duodenale, Necator americanus: most
common hookworm species causing
helminth gastrointestinal infection
▫ Light infection (< 100 worms)
▫ Moderate infection (100–500 worms)
▫ Heavy infection (500–1000 worms)
▪ Pathophysiology
▫ Hookworm attachment, hyaluronidase
release → intestinal wall degradation,
capillary laceration → anticoagulant
peptides, inhibits platelet function
inhibitor production → bleeding
facilitation → extravasated blood
ingested (approx. 0.5mL daily)
▫ Moderate–heavy infections → chronic
iron-deficiency anemia; protein
malnutrition
Infectious form
▪ Filariform larva
Life cycle
▪ Infectious form maturation
▫ Female A. duodenale lays 10,000–
30,000 eggs daily
▫ Female N. americanus lays 5,000–
10,000 eggs daily
▫ Human feces egge release → soil
deposition → eggs hatch (24–48
hours—favorable conditions)
→ rhabditiform larvae (L1) →
transformation to infectious filariform
larvae (L3) in 5–10 days
▪ Human stages
▫ Filariform larvae skin penetration →
passage through veins to heart, lung
→ pulmonary alveoli penetration,
ascend bronchi to pharynx → coughed
up/swallowed into small intestine →
maturation → intestinal wall attachment
via buccal capsule
▪ Some A. duodenale larvae undergo
developmental arrest in gut tissues/muscle
▫ Await more favorable environmental
conditions (hypobiotic larvae)
▪ Natural life-span
▫ A. duodenale: approx. one year
▫ N. americanus: 3–5 years
Transmission
▪ Contaminated soil contact → percutaneous
larval penetration
▪ Oral route
▪ Transmammary route
RISK FACTORS
▪ Inadequate clean-water access
▪ Poor sanitary conditions
▪ Walking barefoot (endemic areas)
COMPLICATIONS
▪ Severe iron-deficiency anemia; protein
malnutrition → impaired growth, cognitive
130
development (children); heart failure
(adults)
▪ Pregnancy
▫ Low birth weight, maternal anemia, ↑
infant mortality
SIGNS & SYMPTOMS
▪ May be asymptomatic
Three phases
▪ Cutaneous phase
▫ Local pruritic dermatitis (ground itch)
with papular, sometimes vesicular, focal
rash at larval penetration site (usually
between toes)
▪ Pulmonary phase
▫ Usually asymptomatic, may involve mild
cough, pharyngeal irritation, sore throat,
fever
▪ Gastrointestinal phase
▫ Midepigastric pain, appetite loss,
nausea, diarrhea, vomiting
▫ A. duodenale, N. americanus eggs are
morphologically indistinguishable
OTHER DIAGNOSTICS
▪ History
▫ Contaminated-soil skin exposure;
endemic-area travel; physical
examination
TREATMENT
MEDICATIONS
▪ Anthelmintic treatment
Heavy infection
▪ Hypoproteinemia → weight loss, anasarca,
edema
▪ Anemia → fatigue, mental dullness,
dyspnea, pallor
▪ Overt gastrointestinal bleeding
DIAGNOSIS
DIAGNOSTIC IMAGING
Abdominal X-ray
▪ Intestinal worm visualization
LAB RESULTS
▪ Acute infection
▫ Eosinophelia
▪ Chronic infection
▫ Anemia
▪ Kato–Katz method (thick smear)
▪ Polymerase chain reaction (PCR) test
Direct microscopy
▪ Stool specimen egg visualization
131
Chapter 82 Nematodes (Roundworms)
ANGIOSTRONGYLUS
(EOSINOPHILIC MENINGITIS)
osms.it/angiostrongylus
PATHOLOGY & CAUSES
▪ Parasitic nematode
▫ Causes human GI/CNS disease
▪ Angiostrongylus cantonensis
▫ Medically important species
▫ Causes angiostrongyliasis (most
common eosinophilic meningitis cause)
Adult morphology
▪ Three outer protective collagen layers;
contains fully-developed gastrointestinal
tract, simple stomal opening
Hosts
▪ Primary intermediate host: snails
▪ Paratenic hosts: fish, frogs, freshwater
prawns
▫ Not needed for developmental cycle
▪ Definitive hosts: wild rodents (brown, black
rat)
▪ Incidental hosts: humans
Life cycle
▪ Infectious form maturation
▫ Worms lay eggs in rat pulmonary
artery → spread from lung capillaries
to alveoli, larvae hatch → migrate up
bronchi, trachea, across epiglottis →
swallowed → fecal larvae passage →
soil deposition → intermediate-host
ingestion
▪ Human stages
▫ Intermediate host larval ingestion
(contaminated water/vegetable)
→ CNS tropism → migration into
meningeal capillaries, brain tissue →
meningoencephalitis
▪ Larvae usually fail to complete life-cycle,
rarely → adult form in human host
Transmission
▪ Intermediate/paratenic host ingestion
(raw/undercooked snails, fish, frogs),
contaminated vegetables/water
Pathophysiology
▪ Post-inoculation, A. cantonensis larvae
exhibit neurotropism → meninges, deeper
brain tissue invasion → neural-tissue
mechanical, toxic byproducts damage;
antigen release → meningoencephalitis
▪ Migration to mesenteric arterioles →
arteritis, thrombosis, small infarctions
▫ May cause necrotic ulcers → peritonitis,
fistula formation
COMPLICATIONS
▪ Long-term encephalitis → permanent
nerve damage, intellectual disability,
permanent brain damage, death
SIGNS & SYMPTOMS
▪ Incubation period: three weeks–two
months
▪ Meninges invasion: meningitis picture
▫ Severe headache, photophobia, stiff
neck, fatigue, fever, hyperesthesia,
vomiting, paresthesias
▪ Brain parenchyma invasion: encephalitis
symptoms (brain location-dependent)
▫ Cognitive impairment, slowed reactions,
neuropathic pain, ascending weakness
▫ t quadriparesis, areflexia, respiratory
failure, muscle atrophy, death (rare)
▪ Eye invasion: visual impairment, pain,
keratitis, retinal edema
132
DIAGNOSIS
TREATMENT
LAB RESULTS
▪ Self-limiting infection (usually)
▪ Cerebrospinal fluid (CSF)
▫ Eosinophilia
▫ ↓ glucose levels (severe
meningoencephalitis)
MEDICATIONS
▪ Analgesics, sedatives
▫ Treat headache, hyperesthesia
▪ Corticosteroids
▪ Antihelminthic therapy not advised
▪ Dying parasites, neurologic damage
exacerbation → potential inflammatory
response
OTHER DIAGNOSTICS
▪ Endemic-area travel history
▪ Physical examination
▪ Clinical presentation
OTHER INTERVENTIONS
▪ CSF drainage
▫ Reduces intracranial pressure, relieve
headache
ANISAKIS
osms.it/anisakis
PATHOLOGY & CAUSES
▪ Zoonotic roundworm
▫ Causes human gastrointestinal, extragastrointestinal disease
▫ Causative anisakiasis agent
Adult morphology
▪ Anteriorly-located mouth surrounded by
projections; length (2cm/0.8in)
Infectious form
▪ L3 larvae
Hosts
▪ Natural hosts: marine mammals
▪ Incidental hosts: humans
Life cycle
▪ Infectious form maturation
▫ Eggs hatch into larvae (seawater) →
larval crustacean ingestion → fish/
squid ingest crustaceans → muscular/
subdermal larval encystation (L3
larvae) → marine mammals ingest fish
→ excystation, maturation, nematode
reproduction → fecal egg release
▪ Human stages
▫ Larval ingestion (infected fish) → larvae
fail maturation, cannot complete lifecycle
Transmission
▪ Raw/undercooked fish ingestion
Pathophysiology
▪ Infected fish larvae ingestion → larvae
burrow into intestinal wall, die → dead
organism → inflammatory response
→ possible allergic reaction, abscess,
mechanical obstruction
▪ If parasites pass into large intestine
▫ Eosinophilic granulomatous response →
mimic appendicitis/Crohn’s disease
RISK FACTORS
▪ Raw/undercooked seafood ingestion
(common in Japan)
133
Chapter 82 Nematodes (Roundworms)
COMPLICATIONS
▪ Small bowel obstruction
▪ Intestinal perforation
▪ Peritoneal cavity perforation (extraintestinal
anisakiasis)
▪ Eosinophilic gastroenteritis/enterocolitis
SIGNS & SYMPTOMS
▪ Gastric anisakiasis
▫ Acute epigastric pain, nausea, vomiting
▪ Intestinal anisakiasis
▫ Severe abdominal pain, abdominal
distension, palpable abdominal mass,
intestinal obstruction, bloody diarrhea
▪ Allergic reactions
▫ Urticaria, bronchoconstriction,
angioedema, anaphylactic shock
DIAGNOSIS
DIAGNOSTIC IMAGING
▪ Parasite visualization
▫ Gastroscopic examination, emesis
examination
LAB RESULTS
▪ ↑ serum IgE
TREATMENT
SURGERY
▪ Parasite removal (endoscopically, surgically)
ASCARIS LUMBRICOIDES
osms.it/ascaris-lumbricoides
PATHOLOGY & CAUSES
▪ Intestinal roundworm parasite
▫ Causative ascariasis agent
▪ Adult worm size: 15–30cm/5.9–11.8in
▫ Largest intestinal nematode
Life cycle
▪ Female A. lumbricoides lays 200,000 eggs
daily; begins egg-laying 9–11 weeks postinfection
▪ Infectious-form maturation
▫ Eggs passed in stool → soil deposition
→ eggs embryonate; infectious after
2–4 weeks; can survive < ten years
(favorable conditions)
▪ Human stages
▫ Egg ingestion → larvae hatch → invade
intestinal mucosa → portal circulation
→ systemic circulation → liver → lungs
→ larvae mature in alveoli (10–14 days)
→ ascend bronchial tree, pharynx,
swallowed → larvae develop into adult
form in small intestine
▪ Life-span: 10–24 months
Pathophysiology
▪ Varies upon life-cycle stage
▫ Pulmonary phase (early): caused
by larval migration into lungs →
pneumonitis
▫ Intestinal phase: manifestations caused
by adult-form presence → mechanical
obstruction (degree worm-burdendependent)
RISK FACTORS
▪ Egg-contaminated food/water ingestion
(especially pig/chicken liver)
▪ Infected soil (children)
▪ Feco-oral route reinfection
COMPLICATIONS
▪ Intestinal obstruction
▫ May → volvulus, ileocecal
134
▪
▪
▪
▪
intussusception, gangrene, intestinal
perforation
Children
▫ Malnutrition/malabsorption, impaired
growth, cognitive development
Hepatobiliary involvement
▫ Biliary colic, biliary strictures, obstructive
jaundice, liver abscesses
Pancreatic duct obstruction → pancreatitis
Aspiration pneumonia during esophageal
migration to trachea
SIGNS & SYMPTOMS
▪ Often asymptomatic
▪ Pulmonary phase (Loffler syndrome):
develops 4–16 days post-infection
▫ Dry cough, dyspnea, fever, wheezing,
substernal discomfort, blood-tinged
sputum; symptoms subside after 5–10
days
▪ Intestinal phase: develops 6–8 weeks postinfection
▫ Abdominal discomfort, anorexia,
nausea, vomiting, diarrhea
Barium swallow
▪ Intestinal phase
▫ Defects in filling; if worms ingest
contrast, tram-track appearance
demonstrated
Microscopic examination
▪ Pulmonary phase
▪ Eosinophils, Charcot–Leyden crystals in
sputum
▪ Intestinal phase
▪ Eggs/adult form stool visualization (KatoKatz/FLOTAC method)
LAB RESULTS
▪ Pulmonary phase
▫ Peripheral eosinophilia
OTHER DIAGNOSTICS
▪ Exposure/endemic-area travel history
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Pulmonary phase
▫ May show migratory bilateral oval
infiltrates, range from several mm–
several cm
▪ Intestinal phase
▫ Adult Ascaris worm visualization,
intestinal obstruction
CT scan
▪ Pulmonary phase
▫ Multiple nodules, “ground-glass”
attenuation
▪ Intestinal phase
▫ “Bull’s eye” appearance (worm crosssection)
Figure 82.1 A barium study demonstrating
ascariais. There are numerous worms in the
distal part of the duodenum and the ileum.
135
Chapter 82 Nematodes (Roundworms)
TREATMENT
MEDICATIONS
▪ Anthelmintic treatment
▫ Only effective in intestinal phase
▫ Pregnancy: pyrantel pamoate
SURGERY
▪ Complete intestinal obstruction, volvulus,
intussusception, appendicitis, perforation
cases
OTHER INTERVENTIONS
▪ Endoscopic retrograde
cholangiopancreatography (ERCP)
▫ Hepatobiliary-involvement cases
Figure 82.2 Ascaris worms emerging from
the two free ends of the small intestine
during an operation to remove a length of
ischemic bowel.
Figure 82.3 Small bowel packed with
Ascaris lumbricoides worms. The bowel
is distended so greatly that the visceral
peritoneum on the antimesenteric border is
split.
136
ENTEROBIUS VERMICULARIS
(PINWORM)
osms.it/enterobius-vermicularis
PATHOLOGY & CAUSES
▪ Small roundworm
▫ May infect human colon, rectum
▫ Causative enterobiasis agent
Life cycle
▪ Female E. vermicularis lays > 10,000 eggs
daily
▪ Perianal fold egg deposition →
autoinfection by scratching; contaminated
hand, mouth contact → eggs hatch into
larvae (small intestine) → adult-form
maturation (cecum, appendix)
▪ Life-span: 2–3 months
Transmission
▪ Scratching perianal area (autoinfection) →
hand–mouth contact
▪ Contaminated hands (person–person)
▪ Contaminated surface contact
▪ Eggs may become airborne, inhaled
▪ High worm burden → abdominal pain,
nausea, vomiting
DIAGNOSIS
OTHER DIAGNOSTICS
▪ Pinworm paddle/Scotch tape test
▫ Adhesive clear plastic paddle pressed
against perianal areas → placed
onto glass slide; reveals eggs upon
microscopic examination
TREATMENT
MEDICATIONS
▪ Anthelmintic treatment
RISK FACTORS
▪ Crowded living conditions
▪ Children 5–10 years old
COMPLICATIONS
▪ Persistent perianal-area scratching, skin
tearing → bacterial dermatitis, folliculitis
▪ Adult worms migrate to extraintestinal
sites; cause vulvovaginitis, salpingitis,
oophoritis, cervical granuloma, peritoneal
inflammation
Figure 82.4 Pinworm found incidentally in
an appendectomy specimen.
SIGNS & SYMPTOMS
▪ Mostly asymptomatic
▪ Perianal itching (pruritus ani) occurs
nocturnally
137
Chapter 82 Nematodes (Roundworms)
GUINEA WORM (DRACUNCULIASIS)
osms.it/guinea-worm
PATHOLOGY & CAUSES
▪ Water-borne nematode disease
▫ Characterization: rash, GI illness with
subcutaneous worm visualization
Life cycle/transmission
▪ Infected water → human copepod ingestion
→ GI, subcutaneous migration, sexual
reproduction → pruritus, percutaneous
larval eruption upon water-immersion
→ copepod larval consumption → two
molting processes → pathogenic larvae in
copepods
Pathogenesis
▪ GI symptoms
▫ Ingested copepod death in GI system
→ larval migration into stomach,
intestinal wall → entry into abdominal,
retroperitoneal space
▪ Cutaneous symptoms
▫ Larval sexual reproduction → female
survival, skin migration → limb waterimmersion → eruption
RISK FACTORS
▪ Rural Eastern-Africa residence/travel
▪ Cutaneous symptoms
▫ Painful papule (2–7cm/0.8–2.8in) →
enlarges, ↑ pain → worm emerges
through ulceration
DIAGNOSIS
DIAGNOSTIC IMAGING
X-ray
▪ Calcified dead worm in subcutaneous
tissue
▫ If worm does not emerge through skin
OTHER DIAGNOSTICS
▪ Endemic-area travel/residence history
▪ Physical examination
▫ Systemic → cutaneous symptom
development
TREATMENT
OTHER INTERVENTIONS
▪ Worm extraction; careful extraction protocol
▫ Multiple days (centimeters at a time);
keeps worm intact, prevents local
pruritic, edematous reaction
COMPLICATIONS
▪ Ectopic site migration → abscess
development
▫ Lung, eye, pericardium, spinal cord
▪ Broken worm → intensely painful,
edematous local, subcutaneous reaction
SIGNS & SYMPTOMS
▪ Systemic symptoms initially
▫ Fever, urticaria, pruritus, dizziness,
nausea/vomiting, diarrhea
Figure 82.5 A match stick being used to
extract a guinea worm from an ulcer on the
leg.
138
Prevention
▪ Community surveillance, transmission
methods education
▪ Safe water precautions
▫ Nylon filters for water filtration,
insecticides in drinking water courses,
water source covering (prevents
infected body-part immersion)
▪ Occlusive dressings applied to papules
LOA LOA (EYE WORM)
osms.it/loa-loa
PATHOLOGY & CAUSES
▪ Vector-borne filarial nematode endemic in
Africa
▫ Characterization: transient swelling
episodes, subconjunctival adult worm
migration
▪ AKA African eye worm
▪ Allergic reaction
▪ Subconjunctival infiltration
▫ Associated with conjunctivitis, eyelid
swelling
Life cycle/transmission
▪ Vector
▫ Chrysops (biting deer fly; AKA tabanid
fly)
▫ Breed in rainforest canopies, lay eggs
in muddy swamps; bite humans during
daytime
▪ Tabanid fly human bite → filarial larvae
transmission → three month maturation
process → microfilarial production →
bloodstream release ( ↑ release during
daytime) → tabanid blood meal (infected
individual) → filarial maturation in tabanid
fly
COMPLICATIONS
▪ Onchocerciasis co-infection
▪ Encephalitis (coincident headache,
insomnia, coma may result)
▪ Cardiomyopathy (endomyocardial fibrosis)
▪ Nephropathy
▪ Arthritis
▪ Lymphadenitis
▪ Calabar swelling → entrapment neuropathy
SIGNS & SYMPTOMS
Allergic reactions
▪ Calabar swellings
▫ 5–20cm/2–7.9in non-erythematous
lesions
▫ Transient, local subcutaneous swelling;
face, extremities most common;
localized pain, itching prior to swelling
episodes
▫ Urticaria, pruritus, asthma
Subconjunctival eye infection
▪ Commonly non-painful infiltration of eye
subconjunctival area; conjunctivitis may
occur (with eyelid swelling)
▪ Worm may be visible
RISK FACTORS
▪ Endemic-area residence/travel
▫ Eastern, Central Africa
139
Chapter 82 Nematodes (Roundworms)
DIAGNOSIS
LAB RESULTS
▪
▪
▪
▪
Hypergammaglobulinemia
↑ IgE level
Worm’s presence (blood smear)
Serology
▫ IgG antibodies against L. loa antigens
OTHER DIAGNOSTICS
▪ History, physical examination
▫ Worm detection in subcutaneous tissue,
conjunctiva
▫ Onchocerciasis co-infection
contraindicated (DEC provokes severe
inflammatory skin, eye response;
Mazzotti reaction)
▫ Eradication may require multiple
treatment rounds
▪ Albendazole
▫ Sterilizes mature worms without
microfilarial activity
▪ Antihistamine/corticosteroids
▫ Limits post-treatment immune reactions
(e.g. Calabar swelling)
SURGERY
▪ Large worm removal
TREATMENT
MEDICATIONS
▪ Diethylcarbamazine (DEC)
▫ Active against L. loa microfilariae,
microfilariae (adult worms)
OTHER INTERVENTIONS
Prevention
▪ Weekly DEC prophylaxis
▪ Only considered for long-term endemicarea exposure
ONCHOCERCA VOLVULUS
(RIVER BLINDNESS)
osms.it/onchocerca-volvulus
PATHOLOGY & CAUSES
▪ Filarial nematode transmitted by blackflies
▪ Leading preventable blindness cause in
sub-Saharan Africa
Life cycle/transmission
▪ Simulium blackfly human bite → larvae
skin deposited → adult parasite maturation
(microfilariae) → subcutaneous/deeper
intramuscular tissue migration →
fibrous capsule/nodule development →
reproduction → microfilarial (immature
worm) production, subcutaneous tissue
migration → human blackfly bite →
microfilarial development into infective
larvae (in blackfly)
Ocular onchocerciasis
▪ Common manifestation: West African
savanna
▪ Commonly affects anterior eye chamber
▫ Iridocyclitis, glaucoma, uveitis
▪ Posterior chamber may also be affected
▫ Onchochorioretinits, optic atrophy
▪ Ocular involvement degree correlates with
symbiotic Wolbachia bacteria quantity
Onchocercal skin disease
▪ AKA Leopard/lizard/elephant skin
(especially when depigmentation present);
common manifestation in African forest
areas
▪ Classified by chronicity
▫ Acute/chronic papular onchodermatitis
140
▪ Lichenified onchodermatitis (AKA sowda/
black/dark); epidermal atrophy, elastic fiber
breakdown may occur
PATHOLOGY
Subcutaneous involvement
▪ Onchocercoma
▪ Dermal O. volvulus → inflammatory
response (prostaglandin E2, transforming
growth factor-beta-mediated) → nodule
formation (onchocercoma); nodules
predominate in bony prominence areas,
peak inflammatory response occurs upon
subcutaneous male O. volvulus death
Ocular involvement
▪ Anterior chamber disease
▫ O. volvulus infiltration → immune
response, O. volvulus death →
Wolbachia release → innate immune
response → corneal damage
▪ Posterior chamber disease
▫ O. volvulus infiltration → immune
response → cross-reactivity of O.
volvulus antigen with retinal pigment
epithelial protein → persistent
immunologic response → inflammatory
limbus, iris, choroid damage
RISK FACTORS
▪ West Africa, Eastern South America travel/
residence
▫ Especially savanna, forest
SIGNS & SYMPTOMS
Cutaneous
▪ Pruritus
▪ Nodule development (lymphadenopathy
may develop, persist depending on
infection duration)
▪ Focal darkening/depigmentation
▪ Epidermal atrophy, hyperpigmentation,
hyperkeratosis may be present
Ocular
▪ Punctate keratitis → fluffy corneal
opacities → eosinophilic infiltrate →
sclerosing keratitis → corneal opacification;
progressive (eventually irreversible) vision
deficit
DIAGNOSIS
DIAGNOSTIC IMAGING
Ultrasound
▪ Deep ochoceroma detection
LAB RESULTS
▪ Eosinophilia
▪ Hypergammaglobulinemia
▪ PCR assay
▫ O. volvulus
OTHER DIAGNOSTICS
▪ Rheumatologic/dermatologic onchocercoma
evaluation
Skin snips
▪ Corneoscleral punch biopsy (ocular
involvement)/ disposable razor blade for
epidermal sample (cutaneous disease)
▪ 2+ snips taken in areas likely to harbor
highest microfilariae concentration
▪ Positive only within 9–15 months postinfection (mature microfilariae produce
microfilariae)
Patch test
▪ Topical DEC application → local skin
reaction assessment
▪ Akin to Mazzotti reaction
Fundoscopic and corneal evaluation
▪ Slit-lamp examination reveals wriggling
microfilariae
▪ Individuals sit forward for two minutes prior
to examination (↑ microfilarial visualization
likelihood on chamber examination)
TREATMENT
MEDICATIONS
▪ Ivermectin, doxycycline
▫ Ivermectin: kills immature worms only,
adult worm repopulate months after
treatment
141
Chapter 82 Nematodes (Roundworms)
▫ Doxycycline: kills Wolbachia (symbiotic
bacteria needed for O. volvulus fertility)
for 24 months; block reproduction
OTHER INTERVENTIONS
Prevention
▪ Protective clothing, insect repellent
(especially when blackflies most active—
morning/evening)
STRONGYLOIDES STERCORALIS
osms.it/strongyloides-stercoralis
PATHOLOGY & CAUSES
▪ Filarial disease endemic in tropical areas
with characteristic pulmonary infection
route, complications include septic shock in
immunocompromised individuals
▪ Mild GI, cutaneous, pulmonary
inflammation, disease
▫ Dermatitis, urticaria, duodenitis,
enterocolitis, pneumonitis
▫ May persist for years
Life cycle
▪ Larvae live in fecally-contaminated ground
soil → enter human host through broken
skin → hematologic spread → pulmonary
alveolar sac infiltration, penetration →
ascend tracheobronchial tree → swallowed
→ larval maturation in duodenum, jejunum
→ larval reproduction, fecal excretion
▪ Autoinfection (single-host life-cycle
completion) may occur via larval perianal
skin entry
PATHOLOGY
▪ Larvae contaminate skin through breakage
→ lung migration → inflammation infiltrate
→ intestinal wall migration → maturation,
replication → larval intestinal mucosa
penetration → autoinfection
RISK FACTORS
▪ Constipation, diverticula, steroid-use, ↓
bowel motility → ↑ autoinfection likelihood
▪ Hypogammaglobulinemia
▪ Anti-tumor necrosis factor receptor therapy
▪ Organ transplantation, immune suppression
COMPLICATIONS
▪ Hyperinfection (uncontrolled autoinfection
→ high worm-burden disease)
▫ Immunocompromised individuals
▫ Hematologic parasite spread includes
CNS, heart, liver, lungs, endocrine
glands
▫ May manifest as septic shock
SIGNS & SYMPTOMS
▪ Immunocompetent individuals
▫ Bloating, diarrhea
▪ Cutaneous
▫ Edema, petechiae, serpiginous/urticarial
tracking
▪ Gastrointestinal
▫ Anorexia, nausea, vomiting, epigastric
pain (duodenal ulceration cases),
malabsorption (chronic enterocolitis)
▪ Pulmonary
▫ Dry cough, throat irritation, dyspnea,
wheezing, hemoptysis
142
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Pulmonary disease
▫ Foci of hemorrhage, pneumonitis,
pulmonary edema
LAB RESULTS
Organism identification
▪ Stool examination
▫ Low sensitivity due to intermittent larval
release
▫ Repeat testing ↑ finding’s reliability
▪ ELISA
▫ Immunocompetent individuals
TREATMENT
MEDICATIONS
▪ Ivermectin
▪ Albendazole
▫ May combine with ivermectin in
hyperinfection/disseminated disease
states
OTHER INTERVENTIONS
Prevention
▪ Proper shoe wearing
▫ Prevents broken-skin exposure
▪ Serologic evaluation
▫ Solid organ transplant donors
OTHER DIAGNOSTICS
▪ Endemic-area travel history
Endoscopy
▪ Histopathological (biopsy-driven) diagnosis
▫ Stomach, duodenum, colon evaluation of
mucosa appearance
Figure 82.6 A duodenal biopsy containing
an entire Strongyloides worm, likely an adult
female.
143
Chapter 82 Nematodes (Roundworms)
TOXOCARA CANIS
(VISCERAL LARVA MIGRANS)
osms.it/toxocara-canis
PATHOLOGY & CAUSES
▪ Human roundworm disease: dog, cat, pig
vectors characterized by cutaneous/ocular
disease
▫ Dogs: toxocara canis
▫ Cats: toxocara cati
▫ Pigs: Ascaris suum
Life cycle/transmission
▪ Definitive hosts: cats, dogs, pigs
▪ Accidental hosts: humans
▪ Egg-laden stool → soil contamination
→ definitive host ingestion → GI tract
reproduction → larval gut wall penetration
→ pulmonary migration, penetration →
ascend tracheobronchial tree → swallowed
→ small intestine worm maturation →
reproduction, egg excretion in stool
▪ Paratenic host: non-canine, small
mammals; egg ingestion → larval gut
wall penetration → tissue migration →
cyst formation → human paratenic host
ingestion → infection
Disease
▪ Ocular involvement: inflammatory
granuloma of posterior pole, diffuse
endophthalmitis, solitary peripheral retinal
granuloma
RISK FACTORS
▪ Raw liver/undercooked meat ingestion (e.g.
rabbit, chicken, cattle, pork)
▪ Children
▫ Playground, sandbox use
COMPLICATIONS
▪ Hepatitis
▪ Pneumonitis
▪ Cardiac
▫ Pericarditis, myocarditis
▪ CNS involvement
▫ Myelitis, meningoencephalitis, seizure,
cerebral vasculitis
▪ Ocular
▫ Retinal detachment, blindness
SIGNS & SYMPTOMS
▪ Visceral larva migrans
▫ Pruritic urticaria, fever, anorexia,
malaise, irritability
▪ Ocular larva migrans
▫ Unilateral vision impairment, uveitis,
papillitis, endophthalmitis
▪ Hepatomegaly
▪ Respiratory distress
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Bilateral peribronchial infiltrates
CT scan
▪ Chest: multifocal subpleural nodules,
“ground-glass” opacities, ill-defined
margins
▪ Abdomen: multiple ill-defined lesions in
liver parenchyma
Ultrasound
▪ Abdomen: may also identify lesions
LAB RESULTS
▪ Leukocytosis
▪ Hypogammaglobulinemia (↑ IgG, IgE)
▪ ELISA
144
AfraTafreeh.com exclusive
▫ IgG antibodies against Toxocara
excretory/secretory antigens
▫ Does not differentiate Toxocara canis
from cati
OTHER DIAGNOSTICS
▪ Endemic-area travel history
▪ Physical examination
TREATMENT
MEDICATIONS
▪ Moderate → severe symptoms
▫ Albendazole
▪ Severe inflammatory disease (myocarditis,
pneumonitis, CNS involvement)
▫ Prednisone
▪ Ocular larva migrans
▫ Corticosteroids, albendazole
OTHER INTERVENTIONS
Prevention
▪ Hand hygiene practice
▪ Pet feces disposal, deworming
▪ Undercooked meat (especially liver)
consumption risk education
TRICHINELLA SPIRALIS
osms.it/trichinella-spiralis
PATHOLOGY & CAUSES
→ female worm release larvae → striated
muscle encystation
▪ Roundworm infection (prevalent
worldwide)
▫ Raw/undercooked meat with encysted
organism consumption
▪ Trichinella: nine species, twelve genotypes
▫ Animal intermediate host-specific
▫ T. spiralis: most common worldwide;
variety of carnivorous, omnivorous
animals
Disease
▪ Severity correlates with multiple factors
▫ Number of ingested larvae directly
correlates with ingested meat cooking
temperature
▫ Trichinella species
▫ Incubation period (shorter correlates
with ↑ disease severity)
▪ Gastrointestinal involvement
▫ Maturing larvae burrow in intestinal
mucosa
▫ Diarrheal illness, nausea, vomiting
▪ Muscular involvement
▫ Adult worm dissemination into skeletal
muscle
▫ Symptom resolution occurs with
complete worm encystment
▫ Mild subclinical fatigue, post-exercise
weakness persists months–years
Life cycle/transmission
▪ Domestic cycle: involves pigs, rodents
▪ Sylvatic cycle: broad animal range
▫ Bear, moose, wild boar most common
human infection sources
▪ Trichinella larvae/mature worm ingestion
by intermediate host → GI tract maturation
→ larvae release, migrate from female
worms to striated muscles → encyst →
intermediate host consumption (inadequate
cooking temperature) → larval ingestion
→ larvae release in stomach (upon gastric
acid, pepsin exposure) → small bowel
mucosa invasion → adult worm maturation
RISK FACTORS
▪ Undercooked/raw meat consumption
145
Chapter 82 Nematodes (Roundworms)
COMPLICATIONS
▪ Myositis, myocarditis
▫ Eosinophil-enriched inflammatory
response → life-threatening arrhythmia
development
▪ Encephalitis, pneumonia
▫ Direct larval parenchyma infiltration,
respiratory muscle disease
SIGNS & SYMPTOMS
▪ Gastrointestinal
▫ Nausea/vomiting, abdominal pain,
diarrhea
▪ Muscular
▫ Most severe symptoms
▫ Pain (activity-dependent; ↑ muscle use/
strain correlates with ↑ ↑ pain)
▫ Tenderness, swelling, weakness
▪ Other
▫ High fever, periorbital edema, chemosis,
visual disturbance
▫ Retinal hemorrhage on fundoscopic
examination
DIAGNOSIS
DIAGNOSTIC IMAGING
CT scan/MRI
▪ In severe neurologic-involvement cases
▪ Multifocal cerebral cortex, white matter
lesions
LAB RESULTS
▪ Serology
▫ ELISA, western blot,
immunofluorescence assays
▫ Antibodies only detectable after 2-3
weeks of disease
▫ Eosinophilia
▫ Leukocytosis
▫ ↑ Muscle enzymes, ↑ creatinine kinase, ↑
lactate dehydrogenase
▪ Muscle biopsy
▫ Near tendinous insertion → highest
yield obtained
Figure 82.7 A muscle biopsy containing a
Trchinella spp. larva.
TREATMENT
MEDICATIONS
▪ Mild disease
▫ Self-resolving
▫ Analgesia, antipyretics
▪ Systemic symptoms
▫ Antiparasitic therapy (assists larval
burrowing into intestinal mucosa
treatment)
▫ Corticosteroids
OTHER INTERVENTIONS
Prevention
▪ Postexposure prophylaxis in suspected
cases
▫ Mebendazole within six days of
exposure
▪ Undercooked/raw meat consumption risk
education, safe cooking practices
146
TRICHURIS TRICHIURA
(WHIPWORM)
osms.it/trichuris-trichiura
PATHOLOGY & CAUSES
▪ Human intestinal parasite (AKA whipworm)
▪ T. Trichiura causative trichuriasis agent
Morphology
▪ Adult
▫ 4 cm/1.6 in in length; made of anterior
whip-like esophageal portion, posterior
intestine/reproductive organ portion
▪ Egg
▫ Prolate spheroids, polar plugs at each
end
Life cycle
▪ Female T. Trichiura lays 3000–20,000 eggs
daily 60–70 days post-infection
▪ Infectious form maturation
▫ Unembryonated egg passage in stool
→ soil deposition → eggs embryonate,
become infectious in 15–30 days
▪ Human stages
▫ Egg ingestion → eggs hatch into larvae
in small intestine → larvae mature
into adult worms, embed in cecum,
ascending colon
▪ Life-span: 1-3 years
RISK FACTORS
▪ Poor hygiene, sanitary conditions;
inadequate human fecal disposal;
uncooked/contaminated vegetable
ingestion
COMPLICATIONS
▪ Heavy infection → rectal prolapse
▫ Inflammation, edema caused by high
embedded worm quantity in rectum
(usually small children)
▪ Persistent blood loss → iron-deficiency
anemia
▪ Children
▫ Impaired growth, cognitive development
SIGNS & SYMPTOMS
▪ Mostly asymptomatic
▪ Heavier infection
▫ Abdominal pain, distension, diarrhea,
fecal blood/mucus, nocturnal stooling,
tenesmus
DIAGNOSIS
OTHER DIAGNOSTICS
▪ Kato–Katz thick-smear technique
▫ Eggs in stool visualization
TREATMENT
MEDICATIONS
▪ Antihelminthic treatment
147
Chapter 82 Nematodes (Roundworms)
WUCHERERIA BANCROFTI
(LYMPHATIC FILARIASIS)
osms.it/wuchereria-bancrofti
PATHOLOGY & CAUSES
▪ Mosquito-borne nematode infection
▫ Characterization: lymphatic,
subcutaneous involvement →
disfigurement, disability
▪ Mosquito species vector geographicspecific (e.g. Culex, Aedes)
Life cycle/transmission
▪ Filarial larvae introduction into human skin
during mosquito bloodmeal → lymphatic
spread → maturation into adult worm →
reproduction, microfilarial hematologic
release (commonly nocturnal release)
→ further transmission after infected
individual’s mosquito bite
Disease
▪ Adenolymphangitis, tropical pulmonary
eosinophilia, hydrocele, chronic lymphatic
disease
▪ Chyluria
▫ Intestinal lymphatic discharge into pelvis
▫ May → nutritional deficiency (including
anemia, hypoproteinemia)
SIGNS & SYMPTOMS
▪ Fever, hydrocele
Lymphatic disease
▪ Acute adenolymphangitis
▪ Painful lymphadenopathy, retrograde
lymphangitis; self-resolving (4–7 days),
may recur multiple times per year
▪ Dilation
▪ Lymphangiectasia
▪ Lymphedema
▫ Pitting edema → non-pitting edema →
limb hardening
▫ Hyperpigmentation, hyperkeratosis may
occur late in disease
Pathogenesis
▪ Filarial antigens → TH2-type immune
response → cytokine production (IL-5, IgE)
▪ Adult worm → mechanical lymphatic
disruption → lymphangiectasia, lymphatic
dilation
▪ Endosymbiotic bacteria Wolbachia →
immune response potentiation
DIAGNOSTIC IMAGING
RISK FACTORS
LAB RESULTS
▪ Sub-Saharan africa, Southeast Africa, India,
Pacific island travel/residence
COMPLICATIONS
▪ Lymphedema (AKA elephantiasis)
▫ Chronic lymphatic vessel inflammation
→ limb swelling
DIAGNOSIS
Ultrasound
▪ Lymphatic dilation, lymphangiectasia, adult
worms may be visualized
▪ Circulating filarial antigen assays
▫ ELISA: Og4C3 antigen detection
▫ ↑ IgG4 levels indicate active infection
▪ Blood smear
▫ Nocturnal blood draw important to ↑
microfilariae concentration
148
OTHER DIAGNOSTICS
▪ Physical examination
▫ Wriggling adult filariae in lymphatic
vessels
TREATMENT
MEDICATIONS
▪ DEC
▫ Contraindication: onchocerciasis, severe
Loa Loa infection, pregnancy
▫ Doxycycline may be added as
combination agent for direct larvicidal
activity
▫ ↓ lymphedema effectiveness
▫ Proper skin care, topical antimicrobial
administration (antifungal, antibacterial
agents), extremity elevation, exercise
may ↓ edema over time
SURGERY
▪ Hydrocele complications
Figure 82.8 A histological section of a lymph
node which contains a filarial worm. The
normal lymph node architecture has been
replaced with an inflammatory infiltrate
composed largely of eosinophils.
OTHER INTERVENTIONS
Prevention
▪ Mass drug administration programs
▪ Insecticide-treated bed nets
▪ Repellant, protective clothing in mosquitoendemic regions
149
Chapter 82 Nematodes (Roundworms)
150
NOTES
NOTES
NON TUBERCULOUS
MYCOBACTERIUM
MICROBE OVERVIEW
▪ Pleomorphic acid-fast bacillus; usually rodshaped
▪ Thick waxy coating
▪ Obligate intracellular microorganism
▪ Optimal growing conditions: cool
temperatures (27–33ºC/80.6–91.4°F),
aerobic environment
▪ Proliferates slowly; cannot be cultivated in
vitro
▪ Appearance: red; Ziehl–Neelsen stain
MYCOBACTERIUM LEPRAE
osms.it/mycobacterium-leprae
PATHOLOGY & CAUSES
▪ Primarily infects skin, superficial nerves,
upper respiratory tract mucosa, eyes
▪ Chronic infection: leprosy; AKA Hansen’s
disease
▪ Targets Schwann cells → nerve damage
→ sensation loss → repeated injuries,
infections → gradual destruction of
extremities
▪ Infiltration of skin, cutaneous nerves →
hypopigmented skin lesions
Ridley-Jopling classification
▪ Tuberculoid
▫ ↑ cell-mediated immunity response
▫ AKA paucibacillary (↓ mycobacteria)
▪ Lepromatous
▫ ↓ cell-mediated immunity response
▫ AKA multibacillary (↑ mycobacteria)
▪ Broad disease spectrum
▫ Borderline tuberculoid, mid-borderline,
borderline lepromatous, indeterminate
RISK FACTORS
▪ Close contact with infected individuals (esp.
in areas of poverty), armadillos (enzootic in
Dasypus novemcinctus)
▪ Older age, genetic factors,
immunosuppression
COMPLICATIONS
▪ Lifelong neuropathy → severe
disfigurement, disability
▪ Severe ophthalmic injury → vision loss
▪ Social stigma (falsely believed contagious)
SIGNS & SYMPTOMS
▪ Can be asymptomatic for years
▪ ↓ sensation
▫ Glove, stocking pattern → repeated
painless injuries
▪ Hypopigmented/reddish skin lesions, heal
spontaneously
▫ Tuberculoid: rare, well-demarcated
▫ Lepromatous: numerous, poorly
demarcated
151
Chapter 83 Non Tuberculous Mycobacterium
Nodular swelling (face, earlobes)
Body hair loss (esp. eyebrows, eyelashes)
Tender, thickened peripheral nerves
Ocular involvement
▫ Chronic uveitis (common)
▫ Facial nerve paralysis (lagophthalmos)
→ corneal exposure, dry eye → corneal
ulceration
▪ Late stages
▫ Claw fingers, toes
▫ Foot droop (inability to lift front of foot);
peroneal nerve infiltration
▫ Nasal septum destruction → nose
collapse (saddle nose)
▪
▪
▪
▪
Figure 83.1 A skin lesion on the scalp of an
individual with leprosy.
DIAGNOSIS
LAB RESULTS
▪ Skin biopsy of active lesion
▫ Mycobacteria in cutaneous nerve
▪ Polymerase chain reaction (PCR)
▫ M. leprae DNA in tissues
OTHER DIAGNOSTICS
▪ Clinical examination
Figure 83.2 The hands of an individual with
leprosy. The distal portions of almost all the
digits, aside from one, have been lost.
TREATMENT
MEDICATIONS
▪ Multidrug therapy to prevent resistance
▫ Tuberculoid: dapsone, rifampicin; six
months
▫ Lepromatous: dapsone, rifampicin,
clofazimine; 12 months
OTHER INTERVENTIONS
Figure 83.3 Skin changes on the chest of
an individual infected with Mycobacterium
leprae.
▪ Bacillus Calmette–Guérin (BCG) vaccination
▫ Administered at birth in regions with
increased leprosy rates (e.g. Brazil, India,
Indonesia)
152
NOTES
NOTES
OPPORTUNISTIC FUNGAL
INFECTIONS
GENERALLY, WHAT ARE THEY?
PATHOLOGY & CAUSES
▪ Range of infections caused by fungi; take
advantage of weakened immunity (e.g. HIV/
AIDS, malignancy, immunosuppression),
altered microbiota, breached integumentary
barriers
▪ Present in environment worldwide →
immunocompetent, healthy individuals can
be exposed without resulting in disease
RISK FACTORS
▪ Immunosuppression (e.g. HIV/AIDS,
neutropenia, chemotherapy, hematologic
malignancies, transplant recipients)
SIGNS & SYMPTOMS
▪ Primary local cutaneous, pulmonary
infection to dissemination to various
internal organs
DIAGNOSIS
LAB RESULTS
▪ Direct microscopy with staining, culture,
tissue biopsy, bronchoalveolar lavage
(sputum sample if pulmonary in origin),
polymerase chain reaction (PCR)
TREATMENT
MEDICATIONS
▪ Antifungals
ASPERGILLUS FUMIGATUS
osms.it/aspergillus-fumigatus
PATHOLOGY & CAUSES
▪ Saprophytic fungi species responsible for
majority of invasive, chronic aspergillosis
▪ Found in soil, compost
▪ Asexual reproduction → production of
green pigmented asexual conidia (spores)
→ aerosolized → individuals inhale
everyday → macrophages attempt to
clear conidia → secondary inflammation
after conidia germinate into hyphal forms
→ neutrophil recruitment → activation of
cellular immunity to kill hyphae
▪ Histopathologically, invasive aspergillosis
characterized by progression across tissue
planes
▫ Hallmark: vascular invasion → infarction
+ tissue necrosis
▪ Characteristics of Aspergillus as successful
opportunistic pathogen
▫ Ability to grow at 37ºC/98.6°F
▫ Small conidial (2.5–3 micrometers) →
buoyant in air for prolonged periods of
time → inhaled deeply into lung alveoli
153
Chapter 84 Opportunistic Fungal Infections
▪ Aspergillus hyphae angioinvasive
▫ Thrombose arteries → hemorrhagic
infarcts → abscesses
▪ Suspect in immunocompromised
individuals with respiratory distress, fever
(despite broad-spectrum antibiotics)
▪ Second most common cause of invasive
fungal infections in neutropenic individuals
(after Candida species)
▪ Specifically affects pulmonary, sinus,
central nervous system (CNS)
Diseases
▪ Necrotizing otitis externa
▫ More common in advanced HIV cases
▪ Acute pulmonary aspergillosis
▫ Inhaled conidia
▫ Most common cause of death in persons
with chronic granulomatous disease
(CGD)
▫ Can spread locally to involve pleura,
chest wall, vertebrae → dissemination
to other organs
Figure 84.1 Bronchial washing stained with
Grocott methenamine silver stain from and
individual with pulmonary aspergillosis. The
hyphae are uniform, narrow and branch at
acute angles.
▪ Cerebral aspergillosis
▫ Occurs in approx. 40% of individuals
with invasive aspergillosis
▫ Hematogenous dissemination from
extracranial focus (e.g. lung)/direct
extension from sinus
▫ Most common brain abscess in stem cell
transplant individuals
▪ Pulmonary aspergilloma
▫ Nonsaphrophytic (noninvasive)
▫ Colonization of pre-existing cavities
(e.g. tuberculosis, sarcoidosis, bullous
emphysema, bronchiectasis)
▫ Occurs in 15–25% of persons
with cavitating lung disease from
tuberculosis
▫ Lesion impinges on major vessel/airway
→ massive hemorrhage → hemoptysis
▫ “Fungus ball”
▪ Allergic bronchopulmonary aspergillosis
▫ Exposure to allergens of A. fumigatus →
saprophytical growth → colonization of
bronchial lumen → persistent bronchial
inflammation → IgE-mediated allergic
response in airways → hypersensitivity
lung disease → bronchial obstruction
▫ Affects those with asthma (1–2%)/cystic
fibrosis (1–15%)
RISK FACTORS
▪ Decreased immunity
▫ Malignancy, chemotherapy, transplant
(esp. from HLA-mismatched donor),
HIV/AIDS, immunosuppressant therapy,
neutropenia, prolonged high dose
corticosteroid use
▪ Prior pulmonary damage/disease
▪ ↑ age
▪ History of tuberculosis, histoplasmosis,
sarcoidosis, bronchiectasis
▪ Cystic fibrosis, asthma (allergic
bronchopulmonary aspergillosis)
COMPLICATIONS
▪ Hemorrhage → massive hemoptysis
▪ Widespread bronchiectasis + fibrosis →
respiratory failure, death
SIGNS & SYMPTOMS
▪ Acute pulmonary aspergillosis
▫ Unremitting fever in high-risk cases
(most common), dry cough, chest pain,
dyspnea (more common in persons
with diffuse disease), ↑ erythrocyte
sedimentation rate (ESR)
154
▪ Invasive sinusitis
▫ Ear/facial pain, discharge, swelling; nasal
septum/turbinate pallor; epistaxis; orbital
swelling, headache; localized areas of
frank crusting, ulceration, blackened
necrotic areas
▪ Cerebral infection
▫ Headache, nausea, vomiting; altered
mental status, confusion, cranial nerve
palsies, hemiparesis
▪ Pulmonary aspergilloma
▫ May be asymptomatic; persistent,
productive chronic cough, hemoptysis,
weight loss
▪ Allergic bronchopulmonary aspergillosis
▫ Manifestations due to immune system
response to A. fumigatus antigens;
asthma-like symptoms (e.g. wheezing),
eosinophilia
▪ Invasive aspergillosis
▫ Acute onset of fever, cough, respiratory
distress, diffuse bilateral pulmonary
infiltrates
DIAGNOSIS
DIAGNOSTIC IMAGING
CT scan
▪ Increased sensitivity for radiological
diagnosis
▪ Halo sign
▫ Neutropenic individuals (hemorrhagic
nodule due to angioinvasion); rim of
ground glass opacity surrounding
nodule
▪ Air crescent sign
▫ Can develop from halo sign; cavitation
→ sloughed lung tissue encircled with
rim of air
MRI
▪ Target sign
▫ Nodule with lower central signal, higher
contrast-enhancing signal on periphery;
late stage disease
▪ For diagnosis of cerebral aspergillosis;
multiple lesions in basal ganglia
X-ray
▪ Unilateral infiltrates (interstitial, alveolar,
mixed), cavitary lesions, multiple
nondefined 1–3cm.0.39–1.18in peripheral
nodules coalesce into larger masses
LAB RESULTS
Tissue biopsy
▪ Not utilized frequently due to invasive
nature; ↑ risk of bleeding or secondary
infection in immunosuppressed individuals
Cultures
▪ Respiratory tract, sputum cultures
commonly negative; rarely diagnosed by
blood
Bronchoalveolar lavage
▪ Approx. 40% diagnostic yield
Serology
▪ Useful for diagnosis of aspergilloma,
allergic bronchopulmonary aspergillosis in
immunocompetent individual; not useful in
immunocompromised
Galactomannan antigen testing
▪ Enzyme immunosorbent assay recognizes
side chains of galactomannan molecule
▫ Positive: invasive disease
▫ High false-positive rate in neutropenic
cases
Allergic bronchopulmonary aspergillosis
▪ Eosinophilia, ↑ serum IgE
Figure 84.2 A tissue section containing
Aspergillus hyphae and fruiting heads.
155
Chapter 84 Opportunistic Fungal Infections
TREATMENT
MEDICATIONS
▪ Invasive aspergillosis
▫ Voriconazole (preferred over
amphotericin B)/caspofungin; azoleresistance developing
▪ Local pulmonary aspergilloma
▫ Percutaneous intracavitary instillation of
antifungals
▪ Allergic bronchopulmonary aspergillosis
▫ Corticosteroids (attenuate immune
system response)
▫ Antifungal therapy: itraconazole
(decrease fungal burden, antigen load)
▫ Preventative long term antifungal
therapy in immunocompromised
individuals
SURGERY
▪ Local pulmonary aspergilloma
▫ Surgical removal (e.g. lobectomy in
massive hemoptysis)
CANDIDA
osms.it/candida
PATHOLOGY & CAUSES
▪ Oval, budding yeast; forms hyphae,
pseudohyphae
▪ Nonpathological colonization of humans →
overgrowth leads to pathology
▪ Most common cause of invasive fungal
infections in immunocompromised
individuals (e.g. neutropenic cases)
▫ C. albicans species most common cause
of candidiasis
▫ Increasing proportion of fungal
infections caused by nonalbicans
Candida species (e.g. C. tropicalis, C.
parapsilosis, C. kruseim, C. glabrata)
Chronic mucocutaneous candidiasis
▪ Persistent infection of mucous membranes,
skin, nails
▪ More commonly affects those with
defective T-cell mediated immunity
Vulvovaginal candidiasis (VVC)
▪ Originates from spread from GI tract, sexual
transmission
▪ Occurs in 75% of healthy individuals who
are biologically female
▫ 80–90% caused by C. albicans
▫ 10–20% of those with VVC have
severe, recurrent infections; usually from
nonalbicans species
Candida esophagitis
▪ Most common in severely
immunocompromised individuals (e.g. HIV
individuals)
▪ May occur in absence of thrush
▪ In individuals with AIDS, can occur
simultaneously with cytomegalovirus,
herpes simplex infection (HSV)
Disseminated/invasive candidiasis
▪ Rare in immunocompetent individuals
▪ Development of invasive disease due
to interaction between Candida species
virulence factors, colonization burden, host
immunological status
▪ Candidemia
▫ Isolation of Candida from blood culture
▪ Candida species exhibit tissue tropism →
deep organ involvement (e.g. liver, spleen,
brain, bone) in absence of prolonged
candidemia
156
RISK FACTORS
▪ Antibiotic therapy, diabetes mellitus (poorly
controlled), immunocompromised state (e.g.
immunosuppressive therapy, neutropenia,
hematologic malignancy, chemotherapy,
transplant), chronic granulomatous disease,
Job syndrome, impaired cell-mediated
immunity, pregnancy, contraceptive use
(hormonal/intravaginal, intrauterine devices)
COMPLICATIONS
▪
▪
▪
▪
hypertension, flank mass, pyelonephritis,
acute urinary obstruction from fungal
mycetoma → hydronephrosis
CNS: altered mental status, characteristic
symptoms of meningitis
Optic: chorioretinal infections, lens abscess
Hepatosplenic: right upper quadrant pain;
nausea, vomiting; hepatosplenomegaly
Other: endocarditis (may be from central
vascular catheters), bone/joint infections
▪ Meningoencephalitis
▫ Obstructive hydrocephalus,
calcifications, thrombosis
▪ Renal system
▫ Pyelonephritis
▪ Abscesses in multiple organs
▪ Sepsis, septic shock
SIGNS & SYMPTOMS
Mucocutaneous growth (most common)
▪ Mouth, oropharynx
▫ AKA thrush
▫ Thick, pearly white, curd-like plaques on
oral mucosa
▫ Painful → dysphagia/odynophagia
▫ Otherwise unexplained → suspect HIV
infection
▪ Vagina
▫ Thick, cottage-cheese-like, white
vaginal discharge
▫ Painless, pruritic
▫ Dysuria possible
▪ Cutaneous candidiasis
▫ Erythematous pruritic patches + satellite
lesions
▫ Individuals who are obese, diabetic
▫ Skin folds, underneath breasts
▪ GI tract
▫ May be asymptomatic
▫ Esophagus → odynophagia
Disseminated/invasive candidiasis
▪ Candidemia: nonspecific (hard to
distinguish from bacteremia); most
commonly manifests as persistent fever
despite antibiotic therapy
▪ Renal system: candiduria, rising creatinine,
Figure 84.3 Oral candidiasis on the tongue
of a child who had recently taken oral
antibiotics.
DIAGNOSIS
LAB RESULTS
▪ Microscopic examination
▫ KOH preparation; visualization of
hyphae, pseudohyphae, blastospores
▪ Invasive candidiasis
▫ Blood/tissue culture
▪ PCR
▪ Esophagitis
▫ Tissue biopsy (definitive)
157
Chapter 84 Opportunistic Fungal Infections
OTHER DIAGNOSTICS
▪ Clinical presentation, history
TREATMENT
MEDICATIONS
▪ Oropharyngeal candidiasis
▫ Oral nystatin suspension; clotrimazole
troches (dissolves in mouth)
▪ Candida dermatitis
▫ Topical nystatin/miconazole
▪ Vulvovaginal candidiasis
▫ Local miconazole/clotrimazole creams;
oral fluconazole
▪ Invasive, systemic candidiasis
▫ Echinocandins; voriconazole,
caspofungin (preferred over
amphotericin/fluconazole)
▪ Individuals with HIV
▫ Prophylactic antifungals (e.g. oral
nystatin, fluconazole)
OTHER INTERVENTIONS
▪ Candida dermatitis
▫ Keep skin dry
▪ Invasive, systemic candidiasis
▫ Immediate removal of all central lines,
catheters (Candida can develop rapid
biofilms)
CRYPTOCOCCUS NEOFORMANS
osms.it/cryptococcus-neoformans
PATHOLOGY & CAUSES
▪ Heavily encapsulated, nondimorphic, yeastlike fungus, urease positive
▪ Virulence factors
▫ Grows well in 37ºC/98.6°F environment
▫ Produces polysaccharide capsule,
melanin (neurotropism)
▪ Most common cause of fungal meningitis in
immunocompromised adults
▪ Found in bird droppings, soil → inhalation
of airborne fungi → evident/nonevident
pulmonary infection → spreads
lymphohematogenously (can affect any
organ) → meninges
▪ CNS infection associated with high
mortality
COMPLICATIONS
▪ Increased intracranial pressure →
herniation → death
▪ May be due to buildup of cryptococcal
polysaccharide at arachnoid villi →
disruption in cerebrospinal fluid (CSF)
reabsorption
RISK FACTORS
▪ Impaired cell-mediated immunity, highdose corticosteroid treatment, hematologic
malignancies (e.g. leukemia, lymphoma)
▪ HIV/AIDS (most common
immunocompromising state): < 100 cells/
mm3 CD4+ count
Figure 84.4 A histology photomicrograph of
the lung.
158
SIGNS & SYMPTOMS
▪ Pulmonary manifestations
▫ Asymptomatic in 1/3 of
immunocompetent individuals; fever,
cough, pleuritic chest pain, dyspnea,
weight loss, hemoptysis
▪ Neurologic manifestations (most common)
▫ Acute/insidious; headache, fever,
vomiting, nuchal rigidity, mental
status changes/seizures, cryptococcal
abscesses (e.g. cryptococcomas, not
common)
▪ Skin manifestations (10–15%)
▫ Result of direct hematogenous spread/
extension from bone lesion; single/
multiple pustules/papules → ulcer/
abscess
▪ Bone infection (5–10%)
▫ Pain, swelling; joint involvement; often
found incidentally
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Focal/diffuse interstitial infiltrates, hilar
lymphadenopathy
LAB RESULTS
▪ CSF
▫ Lymphocytic pleocytosis; ↓ glucose; ↑
protein, opening pressure; results may
be unchanged in individuals with AIDS
▪ Direct microscopic analysis of CSF/other
body secretions
▫ India ink: capsule visualized with clear
halo
▫ Mucicarmine: visualization of red inner
capsule
▫ Visualize budding
▪ Culture
▫ Sabouraud glucose agar, incubation up
to two weeks
▪ Cryptococcal capsular antigen (serum, CSF,
urine, bronchoalveolar lavage)
▫ Most reliable nonculture-based method
▪ Latex agglutination test
▫ Detects polysaccharide capsular
antigen, ↑ specificity
Figure 84.5 Bronchial washings from
an immunocompromised individual with
pulmonary cryptococcosis. The cryptococcus
spores have a characteristically thick capsule.
TREATMENT
MEDICATIONS
▪ Amphotericin B, flucytosine
▪ Prevention in HIV cases with CD4+ cell
counts < 100 cells/mm3—fluconazole
OTHER INTERVENTIONS
▪ ↑ intracranial pressure complication
treatment
▫ Repeat CSF drainage (most important
factor in reducing mortality)
159
Chapter 84 Opportunistic Fungal Infections
MUCORMYCOSIS
osms.it/mucormycosis
PATHOLOGY & CAUSES
▪ Several genera of family Mucoraceae
causing rapidly progressive, invasive
mucormycosis in various body systems
▪ Subphylum Mucoromycotina, order
Mucorales
▫ Most human infections from members of
family Mucoraceae
▫ Six genera: Rhizopus, Mucor,
Actinomucor, Rhizomucor,
Apophysomyces
▪ Present in decomposing organic matter
(e.g. spoiled food items, soil)
▪ Forms broad, aseptate hyphae branching
at right angles by sexual reproduction with
formation of zygospores
▪ Inhalation of spores → sinuses, lungs
primary location of infection
▪ Immunocompetent host → lung
macrophages ingest, kill spores →
neutrophils kill hyphae
▪ Immunosuppressed individuals →
macrophages fail to stop spore germination
▪ Severe neutropenia/diabetic ketoacidotic
individuals → abnormal neutrophil function
→ increased risk of invasive infection
▪ Angioinvasive mold → results in
thrombosis, infarction, necrosis of
surrounding tissues
Diseases
▪ Rhinocerebral mucormycosis
▫ Necrotic lesion in paranasal sinus →
orbit, face, palate, brain
▫ Most common in diabetic ketoacidosis
▫ Progresses rapidly
▪ Pulmonary mucormycosis
▫ Most common in severe neutropenic
individuals
▪ Gastrointestinal mucormycosis
▫ Rare, occurs in severely malnourished
children
▫ Can affect all segments of GI tract
(esp. stomach, small/large intestine,
esophagus)
▪ Cutaneous mucormycosis
▫ Due to traumatic implantation of spores
from soil (e.g. site of surgical incisions,
burn wounds)
▫ Extensive necrotic infection →
necrotizing cellulitis
▫ Most common in immunocompetent
individuals
▪ Disseminated mucormycosis (involves
CNS)
▫ Most common in severe neutropenic
individuals following pulmonary
infection
▫ Brain (most commonly affected),
metastatic necrotic lesions can occur in
any organ
RISK FACTORS
▪ DM
▪ Diabetic ketoacidosis state
▫ ↑ risk for rhinocerebral mucormycosis
▪ Leukemia, neutropenia, chemotherapy
▫ ↑ risk of rhinocerebral, pulmonary,
disseminated disease
▪ Severe malnutrition
▫ GI mucormycosis
▪ Deferoxamine treatment for iron overload
state
▪ Recipient of bone marrow transplant
▪ Prolonged use of corticosteroids/other
immunosuppressive therapies
▪ Prolonged use of broad-spectrum
antibiotics
▪ IV drug use
COMPLICATIONS
▪ Cavernous sinus thrombosis, cranial
nerve palsies (e.g. proptosis, ptosis,
dilation, fixation of pupil), vision loss,
160
brain abscess, necrosis of frontal lobes, GI
tract perforation, perirenal abscess, renal
infarction
SIGNS & SYMPTOMS
▪ Typical course: rapid onset of necrotic
lesion → fulminant course requiring
aggressive therapy
▪ Rhinocerebral
▫ Black, necrotic lesion on nasal/palatine
mucosa; nasal/sinus congestion, pain;
epistaxis; fever; edema, induration,
necrosis of perinasal, periorbital tissue
▪ Pulmonary
▫ Nonspecific, pneumonia-like; may
involve pleuritic chest pain, cough, fever,
hemoptysis
▪ GI
▫ Abdominal pain, bleeding (e.g.
hematemesis)
▪ Cutaneous
▫ Painful edema, erythema → raised,
indurated lesions with black, necrotic
center
▪ Disseminated
▫ Altered mental status (e.g. lethargy,
obtunded state, confused)
DIAGNOSIS
DIAGNOSTIC IMAGING
Figure 84.6 Bronchial washings stained with
Papanicolaou stain from an individual with
pulmonary mucormycosis. The mucor hyphae
contain no septa, are of variable width and
branch at a wide angle.
TREATMENT
MEDICATIONS
▪ Aggressive antifungal therapy
▫ Amphotericin B
SURGERY
▪ Infected necrotic tissue
▫ Extensive surgical debridement
OTHER INTERVENTIONS
▪ Adjunctive therapy
▫ Hyperbaric oxygen, immune modulation,
white blood cell infusion
CT scan, MRI
▪ Thoracic: nodular lesions, cavitations
▪ Head: extension of infection in sinuses →
affecting brain tissue
LAB RESULTS
▪ Histopathological identification
▫ Distinct hyphae; broad irregularly
branched with rare septations
161
Chapter 84 Opportunistic Fungal Infections
PNEUMOCYSTIS JIROVECII
(PNEUMOCYSTIS PNEUMONIA)
osms.it/pneumocystis-jirovecii
PATHOLOGY & CAUSES
▪ Opportunistic yeast-like fungi (originally
classified as protozoan) responsible for
pneumocystis pneumonia
▪ Formerly known as Pneumocystis carinii
▪ Airborne transmission route; human-tohuman route occurs early in life
▪ Immunocompetent individuals may act as
asymptomatic reservoirs
▪ 5–7 micrometer cysts contain up to eight
pleomorphic intracystic sporozoites →
become excysted → form trophozoites
▪ Reside in alveoli
▪ Disease: pneumocystis pneumonia
▫ Occurs exclusively in
immunocompromised individuals
▫ Remains localized in lungs
▫ Clinical manifestations due to
inflammatory reaction in alveoli lumen/
septum
▫ Fatal if left untreated
RISK FACTORS
▪ Defects in cell-mediated immunity, HIV/
AIDS, severe combined immunodeficiency
syndrome, hematological malignancies,
transplant recipients, hyper IgM syndrome
SIGNS & SYMPTOMS
▪ Most infections asymptomatic in
immunocompetent individuals
▪ Abrupt onset of tachypnea, fever, cough
▪ Respiratory distress
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray/CT scan
▪ Diffuse, bilateral ground glass opacities
▪ First appearing in perihilar area →
progresses peripherally, apical regions
spared
LAB RESULTS
▪ Microscopic examination with silver stain
▫ Disc-shaped yeast
▪ Open lung biopsy
▪ Bronchoalveolar lavage
▪ PCR of sputum/lavage samples
▪ ↓ PaO2 (reflects severity of disease)
▪ Unchanged WBC count
TREATMENT
Figure 84.7 A foamy alveolar cast in a
bronchial washing taken from an individual
with Pneumocystis pneumonia.
MEDICATIONS
▪ Trimethoprim-sulfamethoxazole
▪ Alternatives
▫ Pentamide, dapsone + trimethoprim,
atovaquone
162
▪ Prophylactic trimethoprimsulfamethoxazole/pentamide
▫ Individuals with HIV, < 200 CD4+ cells/
mm3
OTHER INTERVENTIONS
▪ Respiratory support
Figure 84.8 A bronchial wash stained with
Grocott’s methenamine silver highlighting
Pneumocystis spores.
SPOROTHRIX SCHENCKII
osms.it/sporothrix-schenckii
PATHOLOGY & CAUSES
▪ Chronic subcutaneous thermally dimorphic
fungus → sporotrichosis
▪ Found in soil, decomposing vegetation,
plant materials (e.g. moss, hay, wood, rose
bushes)
▪ Found worldwide, mostly in temperate/
tropical regions (16–22ºC/60.8–71.6°F)
▪ Outside human body grows as filamentous
mold; in tissue grows as small budding
yeast cells
Diseases
▪ Lymphocutaneous sporotrichosis
▫ Follows traumatic inoculation of skin/
subcutaneous tissue (e.g. minor insult
from thorns or splinters) → incubation
1–4 weeks → papule develops at site
of inoculation → ulceration of primary
lesion → nonpurulent, odorless drainage
→ similar lesions occur along lymphatic
channel proximal to primary lesion
▪ Pulmonary sporotrichosis
▫ Following inhalation of Sporothrix
conidia
▫ May progress to disseminated disease
▪ Osteoarticular sporotrichosis
▫ Most commonly affected joints: knee,
elbow, wrist, ankle
▫ Chronic infection with progressive
decreased range of motion, pain,
swelling
▪ Meningeal sporotrichosis
▫ Rare, mostly in individuals with cellular
immune defects (e.g. lymphoma, AIDS)
▫ Chronic course
▪ Disseminated cutaneous sporotrichosis
▫ Rare (< 1%)
▫ Numerous small papules/vesicles →
necrotic, ulcerated nodules on trunk,
limbs
▫ Follows lymphatic spread
RISK FACTORS
▪ Lymphocutaneous
▫ Exposure due to skin trauma (e.g. living
in homes with dirt floors)
▫ Individual with outdoor preoccupation
▫ Contact with cats
▪ Pulmonary
▫ Chronic obstructive pulmonary disease
(COPD)
▪ Excessive alcohol use
163
Chapter 84 Opportunistic Fungal Infections
SIGNS & SYMPTOMS
▪ Lymphocutaneous sporotrichosis
▫ Primary papule → ulcerated lesion;
similar lesions visualized along
lymphatic channel proximally; chronic,
fixed cutaneous lesion
▪ Pulmonary
▫ Fever, night sweats, weight loss, fatigue;
dyspnea, cough; purulent sputum;
hemoptysis
▪ Osteoarticular sporotrichosis; progressive
decreased range of motion, pain, swelling
in joints
▪ Meningeal sporotrichosis
▫ Chronic (weeks to months) fever,
headache
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Unilateral/bilateral upper lobe cavities
▪ Variable amount of fibrosis
▪ Scattered nodular lesions
LAB RESULTS
▪ Culture (most sensitive)
▫ Tissue biopsy, sputum, body fluid
▫ Sabouraud’s agar at room temperature
▫ Characteristic arrangement of conidia on
hyphae
▪ Direct microscopy
▫ Typical oval/cigar-shaped cells
▫ Asteroid bodies of S. Schenckii
▪ CSF analysis
▫ Lymphocytic pleocytosis, ↑ protein, ↓
glucose
OTHER DIAGNOSTICS
▪ Clinical examination, history
Figure 84.10 Sporothrix fungi forming
conidia.
Figure 84.9 Partially healed skin lesions of
sporotrichosis in a typical lymphcutaneous
distribution.
TREATMENT
MEDICATIONS
▪ Localized: itraconazole
▪ Severe: amphotericin B
164
NOTES
NOTES
ORTHOMYXOVIRUSES
GENERALLY, WHAT ARE THEY?
PATHOLOGY & CAUSES
▪ Associated clinical syndromes: influenza
(“the flu”), pneumonia
▪ RNA virus family; causes acute respiratory
disease
Genetic material
▪ Negative-sense, single-stranded RNA
Taxonomy
▪ Genera
▫ Influenza A, Influenza B, Influenza C:
infect humans
▫ Influenza D, Isavirus, Thogotovirus,
Quaranjavirus
▪ Classified by surface protein
▫ Influenza A, B: hemagglutinin (H)
(glycoprotein, allows progeny release);
neuraminidase (N) (lectin; binds to host
cell through sialic acid residues)
▫ Influenza C: hemagglutinin esterase
fusion (F) (binds to host cell)
Morphology
▪ Enveloped (outer lipid membrane)
▪ Spherical/filamentous
▪ Size: 50–120nm
SIGNS & SYMPTOMS
▪ Fever, malaise, myalgia, sore throat, nonproductive cough
DIAGNOSIS
▪ Clinical examination
DIAGNOSTIC IMAGING
X-ray
▪ Chest
CT scan
LAB RESULTS
▪ Molecular assays
▪ Viral culture
TREATMENT
MEDICATIONS
▪ Antiviral therapy
165
Chapter 85 Orthomyxoviruses
INFLUENZA VIRUS
osms.it/influenza
PATHOLOGY & CAUSES
▪ Single-stranded RNA virus; causes acute
respiratory disease
▪ AKA “the flu”
▪ Incubation: 1–4 days
Pathogenesis
▪ Influenza virus penetrates upper respiratory
tract → hemagglutinin binds to epithelial
cell sialic acid residue → endocytosis →
viral replication → neuraminidase releases
progeny → viral infection spreads
▪ Viral shedding (progeny release)
▫ Duration: average 4–8 days
▫ Magnitude: ↑ symptoms = ↑ shedding
▪ Influenza A antigen variations → immune
evasion, reinfection
▫ Antigenic shift: major changes in
H/N proteins (two different influenza
virus genome segments reassort) →
epidemic/pandemic
▫ Antigenic drift: minor changes in H/N
proteins (mutation in H/N gene) →
outbreak
Taxonomy
▪ Nomenclature: [type] / [original host] /
[location of first identification] / [strain
number] / [year of origin] ([subtype])
▫ Host type, subtype included in influenza
A viruses only
▫ E.g. H1N1 type A flu virus of duck
origin, found in Alberta, Canada,
35th strain, found in 1976 → A/duck/
Alberta/35/76(H1N1)
Pandemics, associated strains
▪ 1918 “Spanish flu” → H1N1
▪ 1957 “Asian flu” → H2N2
▪ 1968 “Hong Kong flu” → H3N2
▪ 2009 (worldwide) → H1N1
RISK FACTORS
▪ Immunosuppression, age ≥ 65 years, age
< six months, nursing/chronic care facility
resident, pregnancy, chronic disease,
morbid obesity
COMPLICATIONS
▪ Secondary bacterial infection (e.g.
pneumonia, sinusitis, otitis media,
bronchiolitis), acute respiratory distress
syndrome, myositis, rhabdomyolysis,
myocarditis, pericarditis, encephalitis
▪ Secondary bacterial infections generally
due to Streptococcus pneumoniae
SIGNS & SYMPTOMS
Uncomplicated influenza
▪ Systemic: fever, malaise, myalgia,
headaches, weakness, dizziness
▪ Respiratory: non-productive cough, sore
throat, nasal secretion
▪ Mild cervical adenopathy
Complicated influenza
▪ Primary influenza pneumonia: fever,
dyspnea, cyanosis
▪ Secondary bacterial pneumonia: fever,
cough, purulent sputum
Transmission
▪ Direct contact, airborne droplets, fomites
Outbreak
▪ Abrupt
▪ Winter; year-round in tropical regions
▪ Duration: 2–3 months
166
DIAGNOSIS
▪ Clinical examination, during outbreak
DIAGNOSTIC IMAGING
X-ray
▪ chest
CT scan
▪ Primary influenza pneumonia: bilateral
reticular/reticulonodular opacities,
sometimes consolidation
▪ Secondary bacterial influenza: pulmonary
infiltrates
LAB RESULTS
▪ Respiratory tract specimen molecular
assay: reverse-transcriptase polymerase
chain reaction (RT-PCR)
▪ Rapid antigen test: immunoassay
▪ Respiratory tract specimen viral culture
TREATMENT
MEDICATION
▪ Severe illness/risk factors → antiviral
therapy
▫ Neuraminidase inhibitors; H1N1
commonly resistant
▫ M2 proton channel inhibitors; active
against influenza A only
▪ Symptomatic treatment: acetaminophen,
non-steroidal anti-inflammatory drugs
(NSAID)
▪ Secondary bacterial infection: antibiotics
OTHER INTERVENTIONS
▪ Hydration
Prevention
▪ Vaccine
▫ Inactivated (intramuscular/intradermal)/
live attenuated (intranasal); trivalent/
quadrivalent (2 influenza A antigens
+ 1/2 influenza B antigens); annual,
single-dose application (before winter);
age ≥ six years
▪ Antiviral prophylaxis (high-risk individual)
▫ Neuraminidase inhibitors
▪ Infection control
▫ E.g. hand hygiene, face mask
167
NOTES
NOTES
PAPILLOMAVIRUS
HUMAN PAPILLOMAVIRUS
osms.it/human-papillomavirus
PATHOLOGY & CAUSES
▪ Human papillomavirus (HPV): virus that
causes cutaneous and mucosal infections,
resulting in warts (verrucae)
▪ > 200 known types; > 40 transmitted
through sexual contact
▪ Humans: only host
Genetic material
▪ Small, double-stranded, circular DNA virus
Taxonomy
▪ Papillomaviridae family
Morphology
▪ Non-enveloped, capsid virus
CAUSES
▪ Transmission
▫ Skin-to-skin contact via breaks in
epithelium (autoinoculation causes local
spread)
▫ Vaginal, anal, oral sexual intercourse
▫ Vertical transmission (congenital
infection)
▪ Cutaneous HPV
▫ Infection of the basal stem cells of
keratinized skin → viral genome
replicates within proliferating cells →
infected cell eventually reaches the
upper epithelial layers → hyperkeratotic
growth
▪ Mucosal HPV
▫ → infection of epithelium → integration
into the host genome → flat, papular, or
pedunculated growths → high-grade
lesions and cancer may develop
RISK FACTORS
▪ Epithelial trauma
▪ Walking barefoot
▪ Occupational
▫ Meat, poultry, and fish handlers
▪ Use of communal showers
▪ Smoking
▪ Early age of first sexual intercourse
▪ Multiple sexual partners
▪ Uncircumcised males
▪ Immunocompromised state; esp. HIV/AIDS
COMPLICATIONS
▪ Some types have oncogenic potential
SIGNS & SYMPTOMS
▪ Common warts: verruca vulgaris (HPV
types 2, 4)
▫ Cauliflower-like raised surface
located on hands, feet, elbows, knees,
subungual/periungual (under, around
fingernail/on cuticle; may be painful)
▫ Common in children, adolescents
▪ Plantar: verruca plantaris (HPV type 1)
▫ Located on soles of feet
▫ May induce pain when walking
▪ Flat: verruca plana (HPV types 3, 20, 28)
▫ Flat warts found on arms, face, forehead
▫ Common in children, adolescents
▪ Anogenital: condyloma acuminatum
(HPV types 6, 11 low risk; 15 types have
oncogenic potential; types 16 and 18 cause
90% of all genital warts)
168
▫ Cervical cancer: types 16 and 18 70%
of all cases
▫ Anal cancer: type 16 is the usual cause
▫ Other cancers; e.g. oropharyngeal,
vaginal, vulvar, penile cancers usually
caused by type 16
▪ Laryngeal papillomatosis (HPV types 6, 11)
▫ Larynx, respiratory tract
TREATMENT
MEDICATIONS
▪ Cutaneous warts
▫ Topical salicylic acid, fluorouracil 5%
▪ Anogenital warts
▫ External warts: podofilox solution or
gel, sinecatechins ointment, imiquimod
cream
SURGERY
▪ Anogenital warts
▫ Surgical removal
OTHER INTERVENTIONS
▪ Cutaneous warts
▫ May resolve spontaneously
▫ Cryotherapy (liquid nitrogen)
▪ Anogenital warts
▫ External or internal (vaginal, cervical,
intra-anal): cryotherapy, trichloroacetic
acid, bichloroacetic acid
Figure 86.1 A giant anal condyloma in an
immunocompromised male.
DIAGNOSIS
LAB RESULTS
▪ Genetic testing: in situ hybridization/
polymerase chain reaction (PCR)
▫ Real-time PCR (detect HPV viral load)
▪ Immunohistochemistry: biomarker
detection
▫ E6, E7 mRNA
▫ p16 cell-cycle protein
▪ Cervical lesions: cytology
▫ If positive for abnormal cells: colposcopy
and biopsy
Prevention
▪ HPV vaccines
▫ Gardasil (quadrivalent), Cervarix
(bivalent), 9-valent
▫ Both vaccines protect against HPV 16,
18; Gardasil also protects against HPV
6, 11
▫ Administered before primary infection
occurs (9–13 years old)
▪ Cervical cancer screening
▫ Pap smear
▪ Effective barrier contraception
▫ Condoms (reduces risk; less protection
compared to other STIs)
▪ Decrease number of sexual partners
169
Chapter 86 Papillomavirus
Figure 86.2 The histological appearance
of a condyloma. There is hyperkeratosis
and parakeratosis. Numerous keratinocytes
demonstrate perinuclear clearing known as
koilocytosis.
170
NOTES
NOTES
PARAMYXOVIRUSES
MICROBE OVERVIEW
▪ Paramyxoviruses: negative-sense singlestranded RNA virus family
▪ Natural hosts: humans, vertebrates, birds
▪ Replication: occurs in cytoplasm; exits by
budding
▪ Transmission: air borne particles
▪ Viral structure: enveloped, linear genomes,
spherical/pleomorphic
▪ Pathogenic paramyxoviruses: human
parainfluenza virus (HPIV), measles,
mumps, respiratory syncytial virus (RSV)
HUMAN PARAINFLUENZA VIRUSES
(HPIV)
osms.it/human-parainfluenza-viruses
PATHOLOGY & CAUSES
▪ Croup (laryngotracheobronchitis): infection
usually caused by HPIV
▪ Four distinct HPIV serotypes
▫ HPIV-1: croup
▫ HPIV-2: croup; upper, lower respiratory
tract illnesses
▫ HPIV-3: bronchiolitis, pneumonia
▫ HPIV-4: infrequently detected
▪ Common respiratory distress cause
(children)
▪ Viral infection → infiltration of histiocytes,
lymphocytes, other white blood cells →
airway inflammation, edema → upperairway obstruction → ↑ breathing work,
barky cough, inspiratory stridor (turbulent,
noisy airflow), vocal hoarseness
RISK FACTORS
▪ Age
▫ Six months to three years
▪ Biologically-male to biologically-female
ratio of 1.4:1
▪ Previous intubation
▪ Hyperactive airways
▪ Congenital airway narrowing
▪ Late autumn (peak case onset)
COMPLICATIONS
▪ Respiratory failure
▪ Bacterial superinfection
▫ Pneumonia, bacterial tracheitis
SIGNS & SYMPTOMS
▪ Prodrome
▫ Upper respiratory tract infection
symptoms (coryza, cough, mild fever)
▪ Acute onset: “barking” cough
▪ Inspiratory stridor; biphasic stridor (severe
obstruction sign)
▪ Hoarseness
171
Chapter 87 Paramyxoviruses
▪ Respiratory distress, ↑ breathing work
(e.g. suprasternal, intercostal, subcostal
retractions)
▪ Agitation
▪ Symptoms worse at night
▪ Asynchronous chest movement
▪ Severe: fatigue, hypoxia, hypercarbia
DIAGNOSIS
TREATMENT
MEDICATIONS
▪ Corticosteroids; dexamethasone for antiinflammatory effects
▪ Nebulized epinephrine in moderate, severe
croup; temporary airway obstruction relief
OTHER INTERVENTIONS
▪ Provide comfort, avoid child’s further
distress
OTHER DIAGNOSTICS
Westley score
▪ Severity classification
▪ Calculated on five factors
▫ Level of consciousness, cyanosis, stridor,
air entry, retractions
▫ Score between 0–17 classifies case as
mild, moderate, severe croup; impending
respiratory failure
MEASLES VIRUS
osms.it/measles
PATHOLOGY & CAUSES
▪ A paramyxovirus that causes measles, a
highly infectious illness
▫ Fever, cough, coryza, conjunctivitis,
followed by exanthem
▪ Transmitted via person-to person contact,
droplets → infects upper respiratory tract
epithelial cells
Clinical stages (four)
▪ Incubation
▫ 6–21 days
▫ Virus infects respiratory mucosa/
conjunctiva → local replication →
lymphatic tissue spread → disseminates
via blood circulation → first virema
(infection of endothelial, epithelial,
monocyte, macrophage cells)
▫ Usually asymptomatic
▪ Prodrome
▫ 2–4 days
▫ Onset of fever, malaise, anorexia,
conjunctivitis, coryza, cough
▪ Exanthem
▫ Onset 2–4 days after fever
▫ Erythematous, maculopapular,
blanching rash
▫ Begins on face → trunk → extremities
▪ Recovery
▫ Cough persists 1–2 weeks
▫ Immunity thought to be lifelong
RISK FACTORS
▪
▪
▪
▪
▪
Measles virus exposure
Travel to measles-endemic areas
No prior measles immunization
Failed measles vaccine response
Immunocompromised individuals: AIDS,
lymphoma/other malignancy, T cellsuppressive medication
172
COMPLICATIONS
▪ Secondary infection
▪ Diarrhea (most common)
▪ Pneumonia (most common children’s death
cause)
▪ Otitis media (younger individuals)
▪ Encephalitis, acute disseminated
encephalomyelitis, subacute sclerosing
panencephalitis
▪ Subacute sclerosing panencephalitis
SIGNS & SYMPTOMS
▪ Prodrome
▫ Fever onset, malaise, anorexia,
conjunctivitis, coryza, cough
▪ Koplik’s spots on buccal mucosa (1–2 days
before rash onset)
▫ Red spots on erythematous buccal
mucosa
▫ Measles pathognomonic
▪ Maculopapular, blanching, erythematous
rash (approx. 14 days after initial infection)
▫ Head → trunk → extremities
▪ Persistent cough after resolution
▪ Modified measles
▫ Measles infection in individual with
existing measles immunity
▫ Milder symptoms
▪ Atypical measles
▫ Measles virus infection in individuals
immunized with killed virus vaccine
▫ Higher, prolonged fever
▫ Dry cough, pleuritic chest pain may
present
DIAGNOSIS
▪ Individual presenting with febrile rash,
cough, coryza, conjunctivitis
LAB RESULTS
▪ Measles detection; one of following
▫ Enzyme-linked immunosorbent assay
(ELISA): positive measles-specific IgM
serology (most common)
▫ Measles IgG antibody: ↑ (between
acute, convalescent titers)
▫ Reverse transcription polymerase chain
reaction (PCR): measles virus RNA
detection
▫ In culture: Measles virus isolation
TREATMENT
MEDICATIONS
▪ Antipyretics, bacterial superinfection
treatment
OTHER INTERVENTIONS
Figure 87.1 Koplik spots on the oral mucosa
of an individual infected with the measles
virus.
▪ Respiratory support, fluids
▪ Vitamin A supplementation
▫ Vitamin A deficiency plays role in
delayed recovery, complications
▪ Prevention
▫ No specific antiviral therapy
▫ MMR (measles, mumps, rubella) vaccine
▫ Infection control (airborne transmission
precautions for four days after rash
onset)
173
Chapter 87 Paramyxoviruses
Figure 87.2 The histological appearance
of the lungs of an individual with measles
pneumonia. There are numerous giant cells,
the nuclei of which display inclusions.
MUMPS VIRUS
osms.it/mumps
PATHOLOGY & CAUSES
▪ Causes mumps; largely preventable by
vaccination
▫ Fever, headache, malaise, myalgia,
anorexia; followed by parotitis
▪ Transmission
▫ Highly contagious
▫ Transmission via respiratory droplets,
direct contact, contaminated fomites
▫ Viral shedding begins before symptoms
onset
▪ Incubation period: 14–18 days
▪ Outbreaks: schools, military posts, camps,
healthcare settings, workplaces
▪ Replication: occurs in upper respiratory
tract epithelium → spread via lymphatics →
viremia
▪ Lifelong post-infection immunity
RISK FACTORS
COMPLICATIONS
▪ Orchitis/oophoritis, meningitis, encephalitis,
pancreatitis, myocardial involvement,
arthritis, deafness
SIGNS & SYMPTOMS
▪ Prodrome
▫ Fever, malaise, headache, myalgias,
anorexia
▪ Parotitis
▫ Swelling, inflammation, tenderness of
parotid gland(s)
▫ May obscure mandible angle
▫ Unilateral/bilateral
▫ Usually 48 hours after prodrome onset
▫ Commonly children 2–9 years old
▫ Stensen duct orifice: may be
erythematous, enlarged
▪ Mastitis
▪ Unvaccinated status, international travel,
vaccine failure, immunosuppressed
individuals, healthcare workers, closecontact
174
DIAGNOSIS
▪ Diagnosis suspected in individuals with
parotitis/other salivary gland swelling,
orchitis/oophoritis with prodrome, mumps
virus exposure
LAB RESULTS
▪ In parotitis setting, diagnosis established by
detection of
▫ Mump virus RNA: reverse-transcription
PCR (buccal/oral swab)
▫ Serum mumps IgM (may not be
detectable until 5 days after symptom
onset)
▪ Full blood count
▫ Leukocytosis may be seen
▪ Lumbar puncture indicated in suspected
meningitis/encephalitis
Figure 87.3 Mumps virus causes parotitis,
which presents as swelling at the angle of
the jaw, widening the facial outline.
TREATMENT
▪ No specific antiviral therapy
MEDICATIONS
▪ Analgesics, antipyretics, non-steroidal
inflammatory agents (orchitis/oophoritis)
OTHER INTERVENTIONS
▪ Prevention
▫ Measles, mumps, rubella (MMR) vaccine
▫ Infection control (isolation with droplet
precaution until parotid swelling
resolved)
175
Chapter 87 Paramyxoviruses
RESPIRATORY SYNCYTIAL VIRUS
(RSV)
osms.it/respiratory-syncytial-virus
PATHOLOGY & CAUSES
SIGNS & SYMPTOMS
▪ Most common cause of bronchiolitis: viral
infection of the lower respiratory tract,
blockage of small airways (bronchioles)
▫ May also be caused by rhinovirus,
influenza virus
▪ Terminal bronchiolar epithelial cell viral
infection → lung epithelial cell damage/
destruction → small bronchi/bronchioles
inflammation → edema, mucus production,
inflammation → small airways/atelectasis
obstruction
▪ Commonly: children < two years old
▪ Often preceded by upper respiratory tract
infection symptoms; rhinorrhea, headache,
mild fever
▪ Prodrome
▫ Upper respiratory tract infection (rhinitis,
fever)
▪ Cough; tachypnea; expiratory wheeze; ↑
breathing work (nasal flaring, grunting,
retractions); crackles heard on auscultation;
cyanosis
RISK FACTORS
▪ Infants < 12 weeks old, November–May,
prematurity, bronchopulmonary dysplasia/
other chronic lung disease history, tobacco
smoke exposure, daycare attendance,
impaired airway clearance/function (e.g.
cystic fibrosis), congenital heart disease,
immunodeficiency
COMPLICATIONS
▪ Bacterial pneumonia, apnea, respiratory
failure, dehydration, aspiration pneumonia,
asthma
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ If differential diagnosis includes pneumonia
OTHER DIAGNOSTICS
Pulse oximetry
▪ ↓ oxygen saturation
TREATMENT
MEDICATIONS
▪ Oral corticosteroids: prior wheeze history
OTHER INTERVENTIONS
▪ Supplemental oxygen, hydration,
mechanical ventilation
▫ Respiratory symptoms peak on days
3–5, begin to resolve
176
Chapter 88 Parvoviruses
Parvovirus B1 is the smallest known DNA animal virus, coming in at an itty bitty 18 to 28 nanometers in
diameter.
In comparison, the average size of a single human red blood cell is a whopping 7200 nanometers!
While it's mostly known for causing fifth disease, or “slapped cheek syndrome,” in children, parvovirus B19
can also affect adults and it can cause serious illness in individuals with pre-existing conditions like sickle-cell
anemia and HIV.
Parvovirus B19 is part of the parvoviridae family.
It’s a single-stranded DNA virus surrounded by an icosahedral capsid, which is a spherical protein shell made
up of 20 equilateral triangular faces.
And it’s “naked” because the capsid isn’t covered by a lipid membrane.
Parvovirus B19 is primarily transmitted by respiratory droplets when someone coughs or sneezes.
You can also catch it via an infected blood transfusion and a pregnant female can also transmit it through the
placenta to her unborn child.
Now, although the virus first enters cells of the respiratory tract by binding to receptors on host cells, it doesn’t
replicate in them.
Instead it keeps travelling through cells and into the circulatory system until it reaches bone marrow, where
red blood cells are made, a process called erythropoiesis.
Once there, parvovirus B19 uses receptor-mediated endocytosis to enter erythroid progenitor cells, also called
proerythroblasts, the early cells that eventually become red blood cells.
It then uses these cells’ DNA replication machinery in the nucleus to replicate its DNA and assemble new
copies of the virus.
Why not simply replicate in cells of the respiratory system?
Well it turns out that Parvovirus B19 needs two things: it prefers to bind to a specific receptor, the P antigen,
which is found in large numbers on proerythroblasts’ cell membrane and it needs cells that pass through the S
phase of the cell cycle, which is the phase where cell DNA is replicated.
Since the body is constantly producing new red blood cells, there are always proerythroblasts going through
the S phase at any given time.
As the virus replicates and matures, it produces a protein called non-structural protein 1 or NS1, which is toxic
to human cells and causes apoptosis, or cell death.
This means that erythropoiesis breaks down, and fewer new red blood cells go into circulation as a result of
parvovirus B19 infection.
But thankfully this is only temporary.
When the cell dies, it bursts open, releasing copies of the virus into the blood, also called viremia.
177
Our immune system detects the virus and starts producing specific immunoglobulin M and immunoglobulin G
antibodies to fight the infection by forming immune complexes with the parvovirus B19 antigen.
For individuals with a functioning immune system, this typically happens between 10 and 14 days after first
becoming infected with the virus.
Parvovirus B19 is most common in young children and those who live or work with them, like parents,
siblings, and daycare workers.
Fetuses are at risk of parvovirus B19 if their pregnant mother has never had the virus in the past.
Immunocompromised individuals are also particularly at risk of chronic parvovirus B19 infection, since their
immune system cannot mount an appropriate response to the virus.
The incubation period for parvovirus B19 - basically the period before viremia starts - is between 4 and 14
days, after which symptoms develop.
Flu-like symptoms - like a mild fever, headache, and aching muscles - are most common during viremia.
Once the immune response begins and the viremia ends, these symptoms go away and some individuals will
then develop a rash and/or joint pain.
The rash appears as uniform redness of the cheeks, but not the area around the mouth, giving the classic fifth
disease “slapped cheek” appearance.
A lace-like rash might also appear on the trunk and the limbs.
Joint pain and inflammation, or arthralgia and arthritis, linked to parvovirus B19 infection usually affects the
small joints of the hands, wrists, feet, and knees, and are often symmetrical, meaning that the same joints on
both sides of the body will be affected.
Children tend to get the rash whereas adults are more likely to develop joint pain, but it’s not exclusive to
either group.
There are a few complications caused by parvovirus B19 infection.
The decreased red blood cell production can cause transient aplastic crisis in individuals who have underlying
conditions like sickle cell anemia, hereditary spherocytosis, and thalassemia.
Because they already have fewer red blood cells, the breakdown of erythropoiesis results in severe anemia,
with symptoms like pale skin, fatigue, and weakness.
Parvovirus B19 in a pregnant female can cause anemia in her fetus.
Because there are fewer red blood cells to carry oxygen, the heart will pump a larger volume of blood to give
the growing fetus all the oxygen it needs.
This raises the pressure inside blood vessels and fluid can start to leak out of capillaries as a result.
This can result in hydrops fetalis, or the abnormal accumulation of fluid in soft tissues.
178
Chapter 88 Parvoviruses
Fetal anemia is also linked to fetal loss, particularly if the parvovirus B19 infection is in the first half of the
pregnancy.
The good news is that there no fetal defects associated with parvovirus B19 for those fetuses that survive the
infection.
Lastly, immunocompromised individuals, such as organ transplant recipients and people with HIV, can develop
a serious complication from parvovirus B19 called pure red blood cell aplasia.
This is a form of chronic, severe anemia where there are very few immature red blood cells in circulation in
blood vessels or erythroid progenitor cells in the bone marrow.
Symptoms of pure red blood cell aplasia are similar to other forms of anemia, such as lethargy and malaise.
Parvovirus B19 infection is usually diagnosed by clinical examination.
Blood tests looking for antibodies to the virus - specifically immunoglobulin M and immunoglobulin G - are
reserved for atypical presentations of the virus and for pregnant individuals.
Another option is polymerase chain reaction, or PCR, which looks for viral DNA.
It’s the preferred diagnostic method of parvovirus B19 infection in immunocompromised individuals, who
typically do not have high IgM or IgG levels in response to an infection.
PCR can also be used on amniotic fluid to diagnose infection in a fetus.
When it comes to treatment for parvovirus B19 infection, it varies based on the symptoms.
Fifth disease is usually mild and gets better on its own, while a transient aplastic crisis often requires
transfusion of blood products.
Arthralgia and arthritis are treated with NSAIDs and usually resolve on their own.
Chronic infections in immunocompromised individuals are treated with immune globulin intravenous therapy
or IVIG, which involves giving antibodies - mostly immunoglobulin G - taken from donor blood plasma.
Severe anemia in a fetus between 18 and 35 weeks of gestation can be treated with an intrauterine blood
transfusion.
Before 18 weeks, the procedure is too difficult technically.
After 35 weeks, risks of the transfusion are high compared to simply delivering the baby.
Finally, there is no vaccine for parvovirus B19.
Preventive measures include proper hand washing with antiseptic soap and water and sanitizing surfaces that
would’ve come into contact with respiratory droplets, since the virus can survive on surfaces.
179
NOTES
NOTES
PICORNAVIRUSES
MICROBE OVERVIEW
▪ Have small, cytoplasmic, single-stranded,
linear, positive-polarity RNA
Taxonomy
▪ Family Picornaviridae; 35 genera (e.g.
Enterovirus, Apthovirus, Cardiovirus,
Rhinovirus, Hepatovirus)
Morphology
▪ Non-enveloped
▪ Icosahedral capsid
▪ Diameter
▫ 27–30nm(smallest of RNA viruses)
▪ Genome length
▫ About 2500nm
Transmission
▪ Hosts: humans, birds, vertebrates
180
COXSACKIEVIRUS
osms.it/coxsackievirus
PATHOLOGY & CAUSES
SIGNS & SYMPTOMS
HFMD (hand, foot and mouth disease)
▪ Clinical syndrome characterized by oral
enanthem; maculopapular/vesicular rash of
hands, feet
▫ Common exanthem in children and
adults
▫ Most commonly caused by coxsackie A
virus
HFMD
▪ Mouth, throat pain
▫ Young children may refuse to eat
▪ Mild fever
▪ Lethargy
▪ Oral enanthem: on tongue, buccal mucosa
▫ Erythematous macules → vesicles with
halo of erythema
▪ Exanthem: macular, maculopapular,
vesicular; nonpruritic, usually not painful
▫ Involves hands (including palms), feet
(including soles), buttocks, legs, arms
▫ Palmer and plantar desquamation 1–3
weeks after presentation
Herpangina
▪ Benign clinical syndrome characterized
by fever, papulo-vesiculo-ulcerative oral
enanthem
▫ Most commonly caused by coxsackie A
virus
▪ Transmission person to person via oralfecal route or respiratory aerosols →
incubation for 3–5 days → virus replicates
in the submucosal lymphoid tissue of
pharynx or lower intestine → spread to
regional lymph nodes (minor viremia) →
dissemination throughout the body →
major viremia
RISK FACTORS
▪ Age
▫ Most common in children <10 years
▪ Season
▫ Spring, summer, autumn
▪ Poor hygiene
Herpangina
▪ Acute onset with high fever; may present
with febrile seizure
▪ Anorexia, emesis, irritability common in
young children
▪ Malaise, headache, sore throat, dysphagia,
abdominal pain
▪ Papulovesicular lesions in throat
▪ Additional findings
▫ Neck stiffness, positive Kernig sign,
cervical adenitis
COMPLICATIONS
▪
▪
▪
▪
▪
▪
Reduced oral intake → dehydration
Rhombencephalitis
Acute flaccid paralysis
Aseptic meningitis
Myocarditis
Pancreatitis
Figure 89.1 Characteristic mouth lesions of
hand, foot and mouth disease.
181
Chapter 89 Picornaviruses
DIAGNOSIS
LAB RESULTS
▪ Throat, stool, vesicular fluid samples for cell
culture/polymerase chain reaction (PCR)
TREATMENT
MEDICATIONS
▪ Analgesia, fluids
▪ No specific antiviral therapy
OTHER INTERVENTIONS
▪ Treat complications (e.g. neurological,
cardiovascular)
Prevention
▪ Infection control practices
▫ Hand hygiene; isolate hospitalized
individuals; contact precautions
Figure 89.2 Characteristic mouth lesions of
hand, foot and mouth disease.
HEPATITIS A VIRUS
osms.it/hepatitis-a-virus
PATHOLOGY & CAUSES
▪ Hepatitis A virus (HAV) → hepatitis A
infection
▪ Reservoir
▫ Humans
▪ Transmission
▫ Oral-fecal route
▪ Infection of hepatocyte by HAV →
replication hepatocyte cytoplasm→
hepatocellular damage → HAV-specific
CD8+ T lymphocytes, natural killer cells
destroy infected hepatocytes → host’s
immune response to HAV → hepatic injury
▪ Infection usually self-limiting (does not
become chronic); results in life-long
immunity (IgG antibodies)
RISK FACTORS
▪ Poor sanitation
▪ Limited access to clean water
▪ Travel to low resource countries
COMPLICATIONS
▪ Cholestatic hepatitis: characterized by
prolonged jaundice
▪ Relapsing hepatitis: symptoms relapse
after acute illness
▪ Autoimmune hepatitis: chronic hepatitis,
characterized by hyperglobulinemia
SIGNS & SYMPTOMS
▪ Usually self-limiting; fulminant hepatic
failure rare
▪ Incubation period
▫ 28 days (average)
▪ Symptoms
▫ Nausea, vomiting, anorexia, fever,
malaise, abdominal pain
182
▪ Signs
▫ Dark urine, pale stools, jaundice (peaks
within two weeks of infection), pruritus,
scleral icterus, hepatosplenomegaly
DIAGNOSIS
LAB RESULTS
▪ Elevated serum aminotransferase levels
may indicate diagnosis
▪ Established diagnosis by detection of
serum IgM anti-HAV antibodies
▫ Serum IgG antibodies in absence
of anti-HAV IgM indicates previous
infection/vaccination
TREATMENT
Prevention
▪ Hepatitis A vaccination
▪ Immune globulin; passive immunization
indicated for
▫ Immunocompromised individuals
(unable to amount immune response to
HAV vaccine)
▫ < 12 months, > 40 years
▫ Individuals with chronic liver disease
▫ Allergy to hepatitis A vaccine
OTHER INTERVENTIONS
Prevention
▪ Infection control practices
▫ Handwashing; avoid tap water, raw
foods in poorly-sanitized areas
MEDICATIONS
▪ Medications known to cause liver damage
should be avoided/used with caution
POLIOMYELITIS (POLIO)
osms.it/poliomyelitis
PATHOLOGY & CAUSES
▪ Infectious disease caused by poliovirus
▪ Characterized by (rare but devastating)
cases of muscle weakness, permanent
paralysis
▫ Most infections remain asymptomatic
▫ Some experience minor symptoms (e.g.
fever, sore throat, headache)
▫ Some may recover from muscle
paralysis
▪ Natural host
▫ Humans
▪ Transmitted by fecal-oral route; less
commonly via respiratory droplets
▫ Asymptomatic, infected persons may
shed virus
▪ Pathogenesis
▫ Oral entry → poliovirus infects cells
of mouth, nose, throat → spread to
lymphatics → primary replication in
tissue of gastrointestinal tract and
oropharynx → primary (minor) viremia
→ invasion of the central nervous
system → replication in motor neurons
of spinal cord, brain stem, or motor
cortex → destruction of motor neurons
→ secondary (major) viremia and
paralysis
RISK FACTORS
▪ Unvaccinated status
▪ Travel to countries endemic for poliovirus
COMPLICATIONS
▪ Post-polio syndrome
▫ Slowly developing muscle weakness
similar to initial infection
▪ Bulbar poliomyelitis
183
Chapter 89 Picornaviruses
▪
▪
▪
▪
▪
▪
▪
▫ Infection of brain stem; may lead
to drooling, aspiration pneumonia,
respiratory muscle paralysis
Skeletal malformations due to muscle
paresis, paralysis
Equinus foot (club foot)
Stunted growth
Osteoporosis, bone fractures
Urinary tract infections, kidney stones
Paralytic ileusv
Myocarditis, cor pulmonale
Acute flaccid paralysis
▪ Complete paralysis; spinal, bulbar,
bulbospinal
▪ Quadriplegia/respiratory failure
▪ Reflexes absent
▪ Sensation intact
SIGNS & SYMPTOMS
Minor illness/minor viremia
▪ Abortive poliomyelitis
▫ Nausea
▫ Vomiting
▫ Abdominal pain
▫ Constipation
▫ Diarrhea
▫ Sore throat
▫ Mild fever
▫ Coryza
Major illness/major viremia
▪ Involvement of central nervous system
(CNS)
▪ Nonparalytic aseptic meningitis
▪ Headache
▪ Neck, back, abdominal, extremity pain
▪ Fever
▪ Vomiting
▪ Lethargy
▪ Irritability
Paralytic disease
▪ Varies from one muscle to muscle group
▪ Reduced tone; often asymmetric
▪ Affects proximal muscles > distal muscles
▪ Affects legs > arms
▪ Worsens over 2–3 days
Figure 89.3 An individual with atrophy of the
mucles of the right leg caused by polio.
DIAGNOSIS
▪ Acute-onset flaccid paralysis
LAB RESULTS
▪ PCR detection of poliovirus RNA from
cerebrospinal fluid; cerebrospinal fluid (CSF)
may also show ↑ leukocytes, ↑ protein
▪ Alternative
▫ Detection by poliovirus isolation, culture
(from throat secretions); comparison of
viral titers in acute, convalescent sera
184
AfraTafreeh.com exclusive
TREATMENT
MEDICATIONS
Antiviral therapy
▪ Role remains uncertain
Prevention
▪ Passive immunization: gamma globulin;
reduces infected individuals’ disease
severity
▪ Inactivated poliovirus vaccine
▫ Given in high-income countries
▫ Cannot revert to paralytic form
▪ Live attenuated oral poliovirus vaccine
▫ Inexpensive, easy to administer; given in
countries where virus endemic
▫ Risk: attenuated virus reverting to
paralysis-causing form
OTHER INTERVENTIONS
▪ Physical therapy; respiratory failure →
mechanical ventilation
▪ No effective treatment for restoring motor
neuron function
Figure 89.4 An individual with polio inside
an iron lung, which provided mechanical
ventilation by creating negative pressure.
RHINOVIRUS
osms.it/rhinovirus
→ host inflammatory response to virus
→ elaboration of inflammatory mediators,
recruitment of polymorphonuclear
leukocytes → symptoms → illness lasts
about 1–2 weeks
PATHOLOGY & CAUSES
▪ The causative agent of most common colds
▪ > 100 serotypes
▪ Most frequent human infectious disease,
preferentially infecting the upper respiratory
tract
▪ Usually causes mild, self-limiting disease,
with increased incidence in early autumn
(September–November) and in Spring
(March–May)
▪ Predisposes to other infections
▫ Otitis media in children, community
acquired pneumonia
▪ Potential to infect the lower respiratory
tract → exacerbation of asthma, chronic
bronchitis
▪ Inoculation of the nose or conjunctiva →
intracellular adhesion molecule-1 (ICAM1) attachment → incubation (2–4 days)
Transmission
▪ Airborne droplet nuclei/aerosols (from
sneezing, coughing)
▪ Droplet transmission
▪ Direct contact (e.g. hand contact → rubbing
eyes, nasal mucosa)
▪ Fomites
RISK FACTORS
▪
▪
▪
▪
▪
Fatigue (insufficient sleep)
Psychological stress
Work in daycare/schools
Smoking
Underlying chronic disease
185
Chapter 89 Picornaviruses
COMPLICATIONS
▪
▪
▪
▪
Sinusitis
Lower respiratory tract disease
Asthma exacerbations
Acute otitis media
SIGNS & SYMPTOMS
▪
▪
▪
▪
▪
▪
▪
▪
▪
Cough
Coryza
Rhinorrhea (clear/purulent)
Sore throat
Myalgia
Fatigue, malaise
Headache
Anorexia
Fever
DIAGNOSIS
DIAGNOSTIC IMAGING
▪ Lower respiratory tract infection → chest
radiograph
OTHER DIAGNOSTICS
▪ Nasal cavity examination: swollen,
erythematous nasal turbinates
TREATMENT
MEDICATIONS
▪ Analgesics
▫ Relieve headache, ear pain, myalgia
▫ E.g. acetaminophen, nonsteroidal antiinflammatory drugs
▪ Antihistamine/decongestant combinations
▪ Cough suppressants
OTHER INTERVENTIONS
▪ Zinc supplements at initial infection may
reduce duration
186
NOTES
NOTES
POLYOMAVIRUS
MICROBE OVERVIEW
▪ Small DNA virus → central nervous
system, urinary system, skin infection
▪ Poly = multiple, oma = tumor →
relationship with multiple tumors
▪ Viral DNA replication, virion assembly
occurs inside cell nucleus
▪ Primary infection usually asymptomatic →
persistent state
Taxonomy
▪ Human polyomavirus species
▫ BK virus
▫ JC polyomavirus
▫ Wu polyomavirus
▫ KI polyomavirus
▫ Merkel cell polyomavirus (MCV)
▫ Trichodysplasia spinulosa virus (TSV)
Morphology
▪ Structure
▫ Double-stranded circular DNA
▫ Icosahedral capsid composed of few
proteins: VP1 (cell surface binding),
VP2, VP3
▫ Nonenveloped
▪ Non-structural proteins: large, small T
antigens
▫ Initiate viral DNA replication
▫ Oncogenic potential
Transmission
▪ Probable transmission routes
▫ Fecal-oral, oral, respiratory
RISK FACTORS
▪ Immunodeficiency
BK VIRUS
(HEMORRHAGIC CYSTITIS)
osms.it/bk-virus
PATHOLOGY & CAUSES
▪ Causes hemorrhagic cystitis
▪ BK virus in hematopoietic stem cell
transplant recipients → bladder
inflammation → bladder mucosa bleeding
▪ Primary infection usually asymptomatic/
mild respiratory infection → urinary tract
infection → virus in bladder mucosal lining,
kidneys
▪ Weak immune system → BK viral
replication → cells large, round → separate
from basal membrane, cell lysis
CAUSES
▪
▪
▪
▪
Radiation
Chemotherapy
Immunosuppressive drugs
Urinary tract infection (e.g. BK virus,
adenovirus)
187
Chapter 90 Polyomavirus
TREATMENT
SIGNS & SYMPTOMS
▪
▪
▪
▪
MEDICATIONS
Frequent urination
Dysuria
Hematuria
Suprapubic pain
▪ Mesna, chemotherapy co-administered
▫ ↓ hemorrhagic cystitis risk
SURGERY
▪ Cystectomy (severe case)
DIAGNOSIS
OTHER INTERVENTIONS
LAB RESULTS
▪ Polymerase chain reaction (PCR) →
infection agent fragments in urine
▪ Urinalysis, cytology
▪ Cystoscopy
▪ Bladder irrigation
▫ Water/sodium chloride solution
▪ Pain management
▪ Hyperhydratation
▪ Hyperbaric oxygen, prostaglandins (↓
efficacy)
JC VIRUS
(PROGRESSIVE MULTIFOCAL
LEUKENCEPHALOPATHY)
osms.it/jc-virus
PATHOLOGY & CAUSES
▪ Causes progressive multifocal
leukoencephalopathy (PML)
▪ Immunocompromised individuals → JC
virus → PML (rare demyelinating disease)
▪ Childhood primary asymptomatic infection
→ latent JC virus in lymphoid organs,
kidneys
▪ Immunodeficiency → virus spreads via
blood → infects brain oligodendrocytes,
astrocytes → viral replication → cell lysis →
myelin sheath loss
▪ Triggers
▫ Immunosuppressive drugs (e.g.
Natalizumab)
▫ Immune system disorders (e.g. HIV/
AIDS)
SIGNS & SYMPTOMS
▪
▪
▪
▪
▪
Cognitive, mental dysfunction
Gait, coordination problem
Hemiparesis, monoparesis
Double/blurred vision
Seizure
DIAGNOSIS
DIAGNOSTIC IMAGING
CT scan
▪ Hypodense lesions affect white matter
MRI
▪ T1 image decreased signal
▪ T2 image increased signal
188
▪ Affects subcortical, periventricular white
matter (usually)
▪ Absent contrast enhancement
LAB RESULTS
Brain biopsy
▪ Large oligodendrocyte nuclei with inclusion
bodies
▪ Myelin sheath destruction
▪ Abnormal, enlarged astrocytes
▪ Macrophages engulfing myelin
▪ Immunohistochemistry for JC proteins
Cerebrospinal fluid (CSF) analysis
▪ PCR detects viral DNA
▪ ↑ white blood cell count (pleocytosis)
▪ ↑ protein level
Figure 90.1 An MRI scan in the coronal place
of the head of an individual with progressive
mutlifocal leucoencephalopathy.
TREATMENT
▪ No specific treatment; high mortality
MEDICATIONS
▪ HIV-infected individuals
▫ Start/optimize antiretroviral therapy
(ART)
▪ No HIV infection
▫ Stop immunosuppressive therapy
▪ Potentially beneficial drugs
▫ Interleukin-2, cytarabine,
chlorpromazine, mefloquine
OTHER INTERVENTIONS
▪ Natalizumab-caused PML
▫ Plasma exchange
Figure 90.2 Immunohistochemical
staining for JC virus protein in the brain of
an individual with progressive multifocal
leukoencephalopathy. The protein,
stained brown, has accumulated in the
oligodendrocytes.
189
Chapter 2 Acyanotic Defects
NOTES
POXVIRIDAE
MICROBE OVERVIEW
Genetic material
▪ Linear double-stranded DNA
Replication
▪ In host cell cytoplasm
Taxonomy
▪ Poxviridae: family of double-stranded DNA
viruses
Associated clinical syndromes
▪ Febrile rash illnesses: smallpox
(eradicated), monkeypox
▪ Skin lesions: vesicles, pustules, papules,
skin thickening
Morphology
▪ Brick-shaped/ovoid
▪ Enveloped (outer lipid membrane)
▪ Size: 220–450nm
MOLLUSCUM CONTAGIOSUM
osms.it/molluscum-contagiosum
PATHOLOGY & CAUSES
▪ Molluscum contagiosum virus (MCV):
poxvirus; causes papular skin disease
▪ Four subtypes; genotype 1 causes most
U.S. cases
▪ Common in children, adolescents
▪ Skin penetration → stratum spinosum
replication within keratinocytes →
epidermal hypertrophy → papules
▪ Incubation period: 2–6 weeks
COMPLICATIONS
▪ Widespread/refractory lesions in
immunosuppressed individuals
CAUSES
▪ Physical contact, autoinoculation, fomites
RISK FACTORS
▪ Contact sports; sexual intercourse with
infected individuals; immunosuppression;
atopic dermatitis
Figure 91.1 The wart-like lesions caused by
molluscum contagiosum infection.
190
SIGNS & SYMPTOMS
▪ Dome-shaped, shiny, umbilicated papules
(2–5 mm); sometimes polypoid
▪ Lesion distribution: trunk, genitals,
intertriginous areas (e.g. axilla, antecubital
folds); not on palms, soles
▪ Sometimes pruritus, inflammation, swelling
▪ Molluscum dermatitis: eczematous
patches/plaques around papules
▪ Eyelid lesions → keratoconjunctivitis
DIAGNOSIS
Figure 91.2 A histological section through
a molluscum wart at low power. There is
marked acanthosis and marked hyperplasia
causing inversion.
LAB RESULTS
▪ Histologic examination: keratinocyte
eosinophilic inclusion bodies (Henderson–
Paterson bodies)
OTHER DIAGNOSTICS
▪ Clinical examination: dermoscopy;
polylobular, amorphous structures with
central umbilication, peripheral blood
vessels
TREATMENT
▪ Optional; lesions resolve spontaneously
in 6–18 months in immunocompetent
individuals
MEDICATIONS
Chemical disruption
▪ Topical blistering agent: cantharidin
▪ Antimitotic agent: podophyllotoxin
▪ Topical immunomodulator: imiquimod
▪ Potassium hydroxide (KOH)
▪ Keratinolytic agent: salicylic acid
Antiviral treatment
▪ Cidofovir
SURGERY
Lesion removal
▪ Cryotherapy, curettage, laser
OTHER INTERVENTIONS
Figure 91.3 A histological section through
a molluscum wart at high power. The
stratum spinosum and granulosum contain
eosinophilic inclusions, known as molluscum
bodies.
Prevention
▪ Avoid sharing towels/clothing
▪ Cover lesions with bandage/clothing
▪ Barrier contraception (e.g. condoms)
191
Chapter 2 Acyanotic Defects
NOTES
REOVIRUSES
MICROBE OVERVIEW
▪ Rotavirus: most important Reovirus in
clinical practice
Genetic Material
▪ RNA viruses with segmented double
stranded linear RNA
Morphology
▪ Encapsulated (icosahedral capsid)
▪ Nonenveloped
Replication/multiplication
▪ Replicates in cytoplasm
ROTAVIRUS
osms.it/rotavirus
PATHOLOGY & CAUSES
▪ A major cause of acute diarrhea, especially
in children
▫ Most important cause of severe
gastroenteritis in infants, young children
worldwide
▪ Short incubation period
▫ ↓ activity of brush-border enzymes
(such as maltase, lactase) →
malabsorption of nutrients → presence
of reducing substances in stools →
osmotic diarrhea
▫ Direct effects of enterotoxin nonstructural protein 4 (NSP4) on
gastrointestinal mucosa (apoptosis of
enterocytes)
▫ Activation of enteric nervous system →
↓ absorption of Na+, loss of K+
CAUSES
▪ Transmission by fecal-oral route (ingestion
of water/food contaminated by stools)
▫ Spreads easily; minimal infective dose
(10 viral particles)
RISK FACTORS
▪ Age
▫ Usually affects children between 6–24
months of age
▪ Cooler months in temperate climates
▪ Hospitalization, long term care facilities,
day care centers, kindergartens, college
dormitories
▪ Immunodeficiency
▪ Non-immunized status
COMPLICATIONS
▪ Secondary lactase deficiency
▪ Severe dehydration
▫ Shock, multisystem failure
▪ Central nervous system complications
▫ Seizures, encephalopathy
▪ Persistent gastroparesis, diarrhea
▪ Necrotizing enterocolitis
▪ Intussusception
▪ Biliary atresia
▪ Can be fatal
192
SIGNS & SYMPTOMS
▪ Average duration: 8 days
▪ Children: watery diarrhea, vomiting, fever
▪ Adults: less severe symptoms
DIAGNOSIS
LAB RESULTS
Blood tests
▪ ↑ blood urea nitrogen (BUN)
▪ Hyperchloremic acidosis
▪ ↓ serum calcium
Stool analysis
▪ Immune based assays
▫ Enzyme linked immunosorbent assay
(ELISA), latex agglutination tests (best
diagnostic tests); PCR, culture
TREATMENT
OTHER INTERVENTIONS
▪ Most cases are self-limited, do not require
pharmacotherapy
▫ Antidiarrheal medications not
recommended (they delay elimination of
infectious agent from intestines)
▪ ↑ fluid intake; oral rehydration solutions
▪ Good nutrition
▪ If infection is severe
▫ Hospitalization for IV fluids
Prevention
▪ Live attenuated vaccine is indicated
routinely for all infants, except for those
with history of intussusception/severe
combined immunodeficiency (SCID)
193
Chapter 2 Acyanotic Defects
NOTES
RETROVIRUSES
MICROBE OVERVIEW
▪ RNA viruses
▫ Require DNA generation, integration
into host DNA → produce viral progeny
▪ Large viral family
▫ Seven genera
▪ Target cells determined by viral
glycoprotein spikes on cell membrane
→ recognized cell surface receptors,
coreceptors → viral entry → provirus
creation
▫ Viral reverse transcriptase takes singlestranded viral RNA genome → creates
linear, double-stranded DNA virus →
provirus
▪ Provirus integration (via integrase) into
host-cell DNA → viral particle production
▫ Integration stability of provirus,
transmission to host-cell progeny
determines retroviral infection
persistence in host organism
Morphology
▪ Lipid-enveloped particles 80–100nm
diameter
▪ Protein core
▫ Two linear, ⊕-sense, single-stranded
RNA genomes (7–11 kb)
▫ Enzymes (gene locus) needed for viral
replication (protease—pol gene, p10
locus; reverse transcriptase—pol gene,
p66 locus; integrase—pol gene, p32
locus)
TYPES
Human endogenous retroviruses
▪ Proviral DNA/partial genomic sequences
integrated into host-genome
▪ Constitutes up to 8% of human DNA
▪ Vertical transmission via germline cells
▪ Translated DNA does not lead to infectious
viruses; may → functional proteins
Human exogenous retroviruses
▪ Passed horizontally via exposure to blood,
sexual secretions, breast milk
HUMAN IMMUNOFEDICIENCY VIRUS
osms.it/HIV-(AIDS)
PATHOLOGY & CAUSES
▪ HIV: human immunodeficiency virus
▪ Member disease of Lentivirus genus of
Retroviridae family
▫ Characterized by immune cell targeting,
immunodeficiency
Two pathogenicity targets
▪ Immune system
▫ Mucosal HIV virion infiltration →
bloodstream spread → infection of
T cells, dendritic cells, macrophages
→ latency (may be chronic,
indolent) → active replication →
symptom progression/emergence
→ further replication→ severe
194
immunocompromise, progression to
AIDS (acquired immune deficiency
syndrome)
▫ HIV infection → targeting, infection of
CD4+ cells (e.g. CD4+ T-lymphocytes,
monocytes, macrophage) →
replication, spread → ↓ CD4+ cells →
immunodeficiency
▫ Co-receptor CCR5 important in early
infection; persistence
▪ Central nervous system (CNS)
▫ HIV infection → macrophage/microglia
infection → abnormal CNS cytokine
milieu → neuronal cell death
Structure
▪ 110nm diameter spherical virion
▪ Core surrounded by lipid bilayer envelope
▪ Lipid envelope
▫ External glycoprotein gp-120, anchored
by gp-41
▫ Binding sites for host-cell CD4+
receptor (co-receptors—chemokine
receptors; especially CCR5, CXCR4)
▪ Core
▫ Two single-stranded RNA copies
▫ Two transfer RNA primers (host-cell
origin)
▫ Multiple enzyme copies: reversetranscriptase (RT), integrase,
ribonuclease H (RNAse H)
▫ Other proteins: vpr, vif, nef (important in
early virion life-cycle)
Genome
▪ 9kb
▪ Similar to other retroviruses (six genes—tat,
rev, vpr, vpu, vif, nef)
▫ HIV-2 specific: Vpx gene (homologous
to gene in simian immunodeficiency
virus (SIV); not present in other
lentiviruses)
Life cycle
▪ Cell contact, fusion: gp-120, CD4+
receptor fusion → gp-120 conformational
change → opened gp-120 recognition site
→ gp-120 binding to host-cell co-receptor
(CCR5/CXCR4) → gp-41 conformational
change → gp-41 hydrophobic domain
exposed → virion lipid bilayer insertion into
cell membrane → virus-host-cell membrane
fusion → viral entry
▪ Viral genome replication: viral RNA
transcribed by viral RT in host-cell
cytoplasm → HIV provirus production
(double-stranded complementary DNA
transcript) → transport to nucleus →
integrase insertion in cell DNA → viral DNA
transcription → viral RNA, protein
▫ Host-cell activation (naive T cells)
important factor in determining active
vs. latent HIV replication
▫ Antigen/cytokine-mediated T
cell activation → NF-KB (nuclear
transcription factor) cell stress activation
→ ↑ NF-KB promoter-associated gene
transcription (in HIV proviral DNA) →
↑ viral replication → viral RNA, protein
aggregation in cytoplasm → viral protein
cleavage (e.g. gag) → viral content
assembly → ↑ virion production (↑↑
virion budding → cell death)
▪ Host-cell death: ↑ virion budding → ↑
host-cell plasma membrane permeability; ↑
viral replication → native protein synthesis
interference → dysregulated cellular protein
concentrations → cell death
▫ Non-cytopathic host-cell HIV infection:
HIV infection → inflammasome pathway
activation → pyroptosis (inflammatory
cytokine, cellular content release) →
immune cell recruitment → viral spread
(likely plays large role in HIV infection
spread)
Transmission
▪ HIV-1, HIV-2
▪ Three transmission modes
▫ Sexual: USA—biologically-male
individuals engaging in same-sex sexual
contact (MSM) particularly important
transmission mode (highest disease
incidence → homosexual biologicallymale individuals); global—majority of
sexual transmission via heterosexual
intercourse; co-existent sexuallytransmitted disease (genital ulceration
especially) → ↑ transmission risk during
intercourse
▫ Parenteral: non-iatrogenic, intravenous
(IV) drug users (shared needles,
syringes, other paraphernalia
contaminated with HIV ⊕ blood);
iatrogenic (hemophiliacs who received
195
Chapter 93 Retroviruses
HIV-contaminated factor VII, IX
concentrates; HIV-contaminated
blood transfusion recipients; health
personnel—e.g. needle-stick injuries)
▫ Vertical, mother-to-infant: in utero,
transplacental spread; infected birth
canal → delivery → neonatal infection;
neonatal HIV ⊕ breast milk ingestion
▪ HIV-2 only
▫ Believed to be zoonosis resulting from
SIV cross-speciation infection
▫ Endemic areas correlate with those of
sooty mangabey
▫ Less transmissible than HIV-1
Disease
▪ Acute retroviral syndrome
▪ AIDS
AIDS
▪ Persistent fever (> one week); fatigue;
weight loss; diarrhea; generalized
lymphadenopathy (LAD); serious,
opportunistic infection
▪ Secondary neoplasms
▪ Neuropsychiatric disease
▫ Delirium; major depression; mania;
schizophrenia; post traumatic stress
disorder; substance abuse, addiction
(commonly HIV infection risk factor)
▫ Dementia (AKA AIDS dementia
complex): cytomegalovirus
encephalitis, progressive multifocal
leukoencephalopathy, cerebral
toxoplasmosis, cryptococcal meningitis,
CNS lymphoma
TYPES
▪ Likely related to SIV
▪ Two human virus types
▫ HIV-1, HIV-2
RISK FACTORS
▪ West Africa residence (HIV-2), homosexual/
bisexual biologically-male individuals
(USA), IV drug users, hemophiliacs, blood
(or component) transfusion recipients,
maternal HIV infection
COMPLICATIONS
▪ Opportunistic infections, secondary
malignancies, AIDS, neuropsychiatric
disease
Figure 93.1 An esophaeal biopsy composed
of squamous mucosa with candida hyphae.
Esophageal candidiasis is a common
opportunitic infection in individual with AIDS.
SIGNS & SYMPTOMS
Acute retroviral syndrome
▪ Self-resolving, flu-like syndrome; sore
throat; myalgias; fever; weight loss; fatigue
Chronic infection
▪ Variable
▫ Asymptomatic → minor infection
▫ Oral/vaginal candidiasis, herpes zoster,
mycobacterial tuberculosis (especially
Sub-Saharan Africa)
Figure 93.2 The histological appearance of
mycobacterium avium intracellulare. There
are numerous organisms within the cell
cytoplasm.
196
197
Chapter 93 Retroviruses
198
DIAGNOSIS
TREATMENT
LAB RESULTS
MEDICATIONS
Serology: enzyme-linked immunoassay
(ELISA)
▪ IgG, IgM, p24 antibody testing
▫ Time to positivity: 15–45 days (P24
(viral core protein) earliest positive
marker)
▫ May be combined for HIV-1, HIV-2, p24
immunoassay in diagnostic, screening
purposes
Highly active antiretroviral therapy
(HAART)
▪ Antiretroviral drug combination regimen
▫ Early initiation → ↓ morbidity, ↓
mortality, ↓ transmission risk (regardless
of CD4+ count)
▪ Six distinct drug classes
▫ Combination of three drug types used
for effective viral load management,
combat particular drug class resistance
development
▪ Protease inhibitor preferred initial agent
▫ Resistance testing results → narrow
therapy
▪ Complications
▫ Immune reconstitution inflammatory
syndrome
▫ ↓ in clinical state (symptom return
experienced during active viremic
phase), despite ↑ CD4+ levels
▫ Believed to be due to reinvigorated host
response to high antigenic burden of
persistent microbes, remaining viral load
▪ ⊕ Viral RNA level test (> 100,000 copies/
mL)
▫ May be used in indeterminate HIV-1/
HIV-2/ p24 immunoassay
▪ Leukopenia
▫ ↓↓ CD4+ count
▫ CD4+:CD8 ratio < 1
OTHER DIAGNOSTICS
▪
▪
▪
▪
Flu-like illness
Opportunistic infection
Needlestick injury with HIV ⊕ individual
Unprotected sex with partner of unknown/
HIV ⊕ status
Pre-exposure prophylaxis
▪ Daily tenofovir use can very effectively ↓
transmission
199
Chapter 93 Retroviruses
200
OTHER INTERVENTIONS
Monitoring
▪ HIV RNA testing two, four, eight weeks
after ART initiation
▫ Continue testing every two weeks until
levels below detection limits
▫ Drug resistance testing at 24 weeks if ↑
in RNA viral level/no ↓ in RNA levels
▪ Once viral suppression achieved → repeat
testing for RNA levels 3–6 months
Screening
▪ One-time for individuals 13–75 years old
▪ Pregnant individuals (even in negative prior
pregnancy screening)
▪ Annual/more frequent for high-risk
individuals
▫ MSM (USA)
▫ IV drug users
▫ Sex partners of HIV unknown status,
HIV ⊕, bisexual, individuals who inject
drugs
▪ Blood factor screening
▫ Plasma/recombinant factor hemophiliac
recipients
Opportunistic infection prophylaxis
▪ Determined by CD4+ count
▪ Situational avoidance advised (e.g. cat litter,
Toxoplasma exposure)
Pre-exposure prophylaxis
▪ Indications
▫ High-risk sexual behavior/drug use
▫ Reliable individual to adhere to daily
medication regimen
Saito Says Hi
201
Chapter 93 Retroviruses
HUMAN T-LYMPHOTROPIC VIRUS
osms.it/human_t-lymphotropic_virus
PATHOLOGY & CAUSES
▪ Oncogenic retrovirus endemic to certain
areas of world → T cell leukemia/lymphoma
▫ AKA HTLV-1
▫ Genus: Deltaretrovirus
▪ Structure
▫ Enveloped, single-stranded RNA virus
▫ Only human pathogen of oncovirus
subfamily
▪ Genome
▫ Contains retrovirus-consistent gag, pol,
env, long terminal repeat (LTR)
▫ HTLV-1 specific: tax; encodes protein
essential for viral replication; viral RNA
transcription stimulation from 5’ LTR
▪ Transmission
▫ Usually via infected T cells (vs. virion
particle)
▫ Breastmilk, sexual transmission, blood
transfusion, tissue donation, IV drug
use, zoonotic transmission (nonhuman
primate source)
▪ Disease
▫ Adult T cell leukemia/lymphoma
▫ Myelopathy/tropical spastic paraparesis
Targets CD4+ cells
▪ Unlike HIV life cycle, pathogenesis; largely
unknown targeting, infection, replication
mechanisms
▫ Tumor initiating cell appear to be CD4+
memory T cell with stem-cell-like
properties
▪ Distinct from HIV
▫ HTLV-1 → proliferation of T cell
population rather than killing of cells
▪ Integrated provirus → mitotic cell division
→ host cell replication
▫ Low replication rate
▫ Host cell DNA polymerase ensure high
transcription fidelity (HTLV-1 genetically
stable)
Tax-specific oncogenic hallmarks
▪ Inr pro-growth signaling, cell survival
▫ Stimulates AKT (via PI3K), NF-KB,
cyclin D2, ↓ CDK inhibitors → ↑ prosurvival, cell growth → polyclonal T cell
expansion
▪ Inc genomic instability
▫ Interference with DNA-repair function
▫ Inhibition of cell cycle checkpoints
(activated by DNA damage)
RISK FACTORS
▪ Travel/residence in Japan, Caribbean basin,
South America, Africa
▫ Sporadic incidence in USA
COMPLICATIONS
▪ Mycosis fungoides
▫ Cutaneous T cell leukemia/lymphoma
manifestation
▪ Uveitis
▪ Gastric cancer
Rheumatologic, pulmonary disorders
▪ Chronic inflammatory arthropathy
▫ Shoulder, wrists, knees
▪ Sjögren syndrome
▪ Immune thrombocytopenia
Infectious replication
▪ Integrated provirus re-expression →
intracellular virion
202
SIGNS & SYMPTOMS
Adult T cell lymphotropic leukemia/lymphoma
▪ Skin lesions
▪ Generalized lymphadenopathy
▪ Hepatosplenomegaly
Myelopathy/spastic paraparesis
▪ Insidious-onset lower extremity weakness,
spasticity
▪ Hyperreflexia, ankle clonus present
▪ ⊕ extensor plantar responses
▪ Lumbar pain
▪ Others
▫ Back pain
▫ Detrusor instability → nocturia, urinary
frequency, incontinence
▫ Minor sensory change: paresthesias, ↓
vibrational sense
DIAGNOSIS
DIAGNOSTIC IMAGING
▪ Lytic bone lesions
MRI
▪ Tropical spastic paraparesis
▫ Cervical/thoracic cord atrophy
▫ Spinal cord white matter disease
LAB RESULTS
▪ Leukopenia
▪ Hypercalcemia
▪ Histology is variable, with characteristic
circulating tumor cells
▫ Medium-sized lymphocytes with
condensed chromatin, bizarre
hyperlobated nuclei (AKA clover leaf/
flower cells)
▪ ELISA
▫ Antibody against HTLV-1 virus
detection
▪ Western blot
▫ Confirmatory testing for ELISA
▪ Polymerase chain reaction (PCR)
▫ Available for HTLV-1
▫ Useful with ↑↑ suspicion (ELISA
negative)
▪ Flow cytometry
▫ ↑ FoxP3 expression in leukocytes (↑
regulatory T cell-indicative)
▫ Otherwise immunophenotypically
mature T lymphocytes
OTHER DIAGNOSTICS
▪ Travel/residence in endemic area
TREATMENT
▪ No therapy proven to benefit affected
individuals
MEDICATIONS
▪ Corticosteroid therapy may slow disease
course
▪ Antiviral therapy
▫ NRTI zidovudine, IFN-alpha proven
beneficial
OTHER INTERVENTIONS
Prevention
▪ Endemic area-avoidance when
breastfeeding
▪ Blood donor screening
▪ Safe-sex practices
▪ Discourage needle sharing
203
Chapter 2 Acyanotic Defects
NOTES
RHABDOVIRUSES
MICROBE OVERVIEW
▪ Diseases
▫ Rabies encephalitis, vesicular diseases
▪ Genera: Lyssavirus, Vesiculovirus,
Sigmavirus, Varicosavirus, Spirivivirus, etc.
Genetic material
▪ Rod-shaped, single-stranded RNA virus
Morphology
▪ Enveloped, bullet-shaped, with helical
nucleocapsids, linear genomes
Taxonomy
▪ Order: Mononegavirales
▪ Family: Rhabdoviridae
Transmission
▪ Via bite (infected host’s saliva)
RABIES VIRUS
osms.it/rabies-virus
PATHOLOGY & CAUSES
▪ Serious central nervous system (CNS) viral
zoonotic infection
▪ Virus spreads via nerves (retrograde
axoplasmic transport)
▪ Muscle tissue inoculation → incubation
(1–3 months), local multiplication →
acetylcholine receptor binding → nerve
entry → travel via spinal cord axons →
brain infection (found in cerebellum Purkinje
cells, hippocampal neurons) → encephalitis
Advanced-stage
▪ CNS → other organs (salivary glands,
cornea, skin, gastrointestinal, etc.) via
parasympathetic nervous system
CAUSES
▪ Caused by genus Lyssavirus (multiple
species, Rhabdoviridae family)
Transmission
▪ Saliva in virus-infected host bite (dogs,
bats, cats, raccoon, foxes, skunks, monkeys,
etc.)
▪ Rabies-infected organ/tissue
transplantation (rare)
▪ Aerosol transmission (e.g. bat caves)
possible
RISK FACTORS
Animal exposure (bite risk)
Travel (rabies-endemic Asian/African areas)
Age < 15 years
Deep bite
Head wound (virus → brain transmission
risk ↑ )
▪ No post-exposure prophylaxis
▪ Occupational (laboratory, veterinarian)
▪ Recreational (spelunking → ↑ bat exposure
risk)
▪
▪
▪
▪
▪
204
COMPLICATIONS
▪
▪
▪
▪
▪
Encephalopathy
Increased intracranial pressure
Coma
Permanent neurological deficits
Often fatal
SIGNS & SYMPTOMS
Prodromal stage
▪ Non-specific symptoms (first week)
▫ Headache; low grade fever, chills;
myalgia, weakness, fatigue; malaise,
anorexia; nausea/vomiting; sore throat;
photophobia (sometimes)
▪ Wound site pain/tenderness/paresthesia/
tingling/itching
Encephalitic rabies
▪ AKA furious rabies
▪ Most common form
▪ Involuntary pharyngeal spasms →
hydrophobia (fear of water); aerophobia
▪ Fever
▪ Muscle spasms → opisthotonus position
▪ Seizure
▪ ↑ autonomic stimulation
▫ Excess salivation; lacrimation; sweating;
mydriasis; impaired temperature
homeostasis; tachycardia
▪ Dysphagia
▪ Aggressiveness, agitation, hallucination,
confusion
▪ Respiratory distress → coma → respiratory
arrest → death
Paralytic stage
▪ AKA dumb rabies
▪ Ascending flaccid paralysis
▪ Sphincter atony
▪ Hydrophobia (rare)
▪ Neck stiffness
▪ Cranial nerves palsy
▪ Fasciculations/deep tendon reflex loss
▪ Pharyngeal, diaphragm muscle paralysis →
death
DIAGNOSIS
▪ Clinical Presentation
▫ History of rabid animal bite, rabies
infection symptoms/signs
DIAGNOSTIC IMAGING
CT scan
▪ Cerebral edema
LAB RESULTS
Reverse transcription PCR (RT-PCR)
▪ Saliva
▫ Detects rabies virus RNA
Skin punch biopsy
▪ RT-PCR; immunofluorescence staining for
viral antigen
Cerebrospinal fluid (CSF)
▪ Indirect immunofluorescence, virus
neutralization assay
▪ CSF analysis
▫ Pleocytosis, ↑ protein
Serum
▪ Anti-rabies virus antibodies in serum
appear after first week (if individual not
immunized)
OTHER DIAGNOSTICS
Post mortem
▪ Brain tissue/other neural tissue examination
▫ Negri bodies, eosinophilic cytoplasmic
inclusions in nerve cell cytoplasm (often)
TREATMENT
MEDICATIONS
▪ Antivirals
▪ Rabies vaccine
▫ Both post-, pre-exposure prophylaxis
▫ Immunocompetent people: four
injections (day 0, 3, 7, 14)
▫ Immunocompromised people: fifth
injection (day 28)
205
Chapter 94 Rhabdoviruses
▫ Previously immunized people: two
injections (day 0, 3)
▪ Human rabies immune globulin (HRIG)
▫ Single dose (20 units/kg) injected in,
around wound
▫ Remainder administered intramuscular
at distant site (e.g. other deltoid)
OTHER INTERVENTIONS
Post-exposure prophylaxis (rapid)
▪ Wound cleaning
▫ Water/soap, povidone iodine
▪ Antibiotics/tetanus prophylaxis
Figure 94.1 Multiple Negri bodies in the brain
of an individual infected with the rabies virus.
The negri bodies form in the Purkinje cells of
the cerebellum.
Management
▪ Respiratory (supplemental oxygen,
mechanical ventilation), cardiovascular
support (fluids)
Prevention
▪ Exposed population may receive preexposure prophylaxis
▪ Domestic animal vaccination (especially
dogs, cats)
206
NOTES
NOTES
RICKETTSIAL DISEASES
GENERALLY, WHAT ARE THEY?
PATHOLOGY & CAUSES
▪ Vector-borne obligate intracellular bacteria;
poor Gram staining, rod-shaped structure
▪ Common tropism for endothelial cells →
variable amount of hemorrhage, edema,
organ dysfunction
SIGNS & SYMPTOMS
▪ Disease-specific; fairly consistent
integument manifestations (e.g. rash)
DIAGNOSIS
LAB RESULTS
▪ Isolation of Rickettsiae
▫ Inoculation of animal/via cell culture
▪ Serology
▫ Enzyme-linked immunosorbent
assay (ELISA), western blot,
microimmunofluorescent antibody test
(detection of bacterial-specific antigens)
▪ Immunologic detection in tissue
▫ Isolation in epithelial tissue (from
integument involvement)
▫ Requires sophisticated laboratory
capability (not common in endemic
areas of disease)
▪ Polymerase chain reaction (PCR)
▫ Detection of rickettsial DNA
OTHER DIAGNOSTICS
▪ Clinical presentation (disease-specific)
TREATMENT
MEDICATIONS
▪ Prompt antimicrobial therapy
▫ Doxycycline (preferred)
207
Chapter 95 Rickettsial Diseases
ANAPLASMA
osms.it/anaplasma
PATHOLOGY & CAUSES
▪ Tick-borne, obligate intracellular bacteria,
endemic to wooded areas in North America
→ self-resolving disease, AKA human
granulocytic anaplasmosis (HGA)
Vector
▪ Anaplasma phagocytophilum: Ixodes tick
▫ Ixodes scapularis: also transmits
Borrelia burgdorferi, Babesia spp. in
eastern United States (US)
▫ Ixodes pacificus: main vector in western
US
▫ Ixodes ricinus: implicated in European
disease
Life cycle and transmission
▪ Reservoir hosts: deer, white-footed
mice (source of disease, not affected by
pathogen)
▪ Vector: Ixodes tick (connects organism from
reservoir to target)
▪ Human transmission
Pathogenesis
▪ Tick bite → blood circulation → leukocyte
infection, membrane attachment →
phagocytosis → replication (in early
endosome of leukocyte) → dysfunctional
vacuolization, immature lysosomal micelles
→ microcolony (AKA morulae) development
→ release into extracellular space after cell
lysis/exocytosis
▫ P-selectin glycoprotein (identified
binding domain for A. phagocytophilum)
▫ Ligand: P-selectin glycoprotein ligand-1
(PSGL-1) required on granulocytes for
internalization
Disease: HGA
▪ Direct leukocyte cell death
▪ Inflammatory response → perivascular
inflammatory infiltrates in multiple organ
systems (without organ failure/endothelial
damage)
RISK FACTORS
▪ Residence in/travel to wooded areas in
North America
▫ Especially during peak tick activity (e.g.
spring, summer)
▪ Direct contact with slaughtered deer
▪ Occupational exposure (e.g. military)
COMPLICATIONS
▪ Co-infection with Borrelia burgdorferi,
Babesia spp.
▪ Respiratory insufficiency, renal failure,
septic shock
▪ Neurological
▫ Demyelinating polyneuropathy, brachial
plexopathy
▪ Serious, fatal opportunistic infections
▫ Herpes simplex esophagitis, invasive
aspergillosis,
SIGNS & SYMPTOMS
▪ Onset 1–2 weeks after identified tick bite
▪ Fever, malaise, headache
▪ Rash
▫ Typically trunk (sparing hands, feet),
maculopapular (more evident in
children)
▪ Gastrointestinal (GI) symptoms infrequent
▪ Neurological (rare)
▫ Mental status change, meningismus,
clonus
DIAGNOSIS
LAB RESULTS
▪ Leukopenia
▫ Specific to disease (neutropenia)
▪ ↑ hepatic enzymes, lactate dehydrogenase
208
▪ Serology: indirect IFA
▫ Detection of IgG/IgM antibodies of
Anaplasma species
▫ If negative on acute serum testing,
repeat with convalescent serum
(confirms diagnosis if ↑ fourfold in IgG
antibody titer)
▪ Whole blood PCR
▫ Detects epank1 primers on genogroup
A. phagocytophilum
▪ Wright stain: morulae of Anaplasma in
leukocyte
▫ A. phagocytophila: peripheral blood
neutrophils; 25–75% (highest among
morulae-producing bacteria)
OTHER DIAGNOSTICS
▪ History
▫ Tick bite in endemic area
TREATMENT
Figure 95.1 Mites of the genus Ixodes act as
vectors for many rickettsial diseases.
MEDICATIONS
▪ Prompt antibacterial management
▫ Doxycycline (if pregnant, rifampin);
chloramphenicol
OTHER INTERVENTIONS
▪ Prevention
▫ Avoid tick habitats
▫ Careful inspection after outside activity
in wooded areas (esp. in spring,
summer); rapid discovery, tick removal
< 24–48 hours post bite → effective
prophylaxis
▫ Skin application of insect repellants
▫ Proper clothing for outside work/play
(light-colored, long pants tucked into
socks, long-sleeved shirts)
209
Chapter 95 Rickettsial Diseases
COXIELLA BURNETII (Q FEVER)
osms.it/coxiella-burnetii
PATHOLOGY & CAUSES
▪ Coxiella burnetii: primarily zoonotic
pathogen → febrile illness (after contact
with animal amniotic fluid/placental
contents)
Taxonomy
▪ Order: Legionellales
▪ Family: Coxiellaceae
Morphology
▪ Short, pleomorphic rod
▫ Strict intracellular bacterium
Life cycle
▪ Source of human infections
▫ Farm animals (e.g. cattle, goats, sheep)
▫ Wild animals (e.g. birds, rabbits, reptiles)
▫ Arthropods (e.g. ticks)
▪ Main reservoir
▫ Ticks
Transmission
▪ Inhalation of spores/bacteria
▫ Animal feces, milk, products of
conception
▪ Ingestion of contaminated milk
▪ Percutaneous
▫ Crushing of ticks near skin breaks
▪ Vertical spread (transplacental)
Pathogenesis
▪ Host cell
▫ Macrophage
▪ Antigenic variation (AKA phase variation)
important in virulence
▫ Lipopolysaccharide capsule
modifications underly antigenic variation
RISK FACTORS
▪ Occupation involving animal contact (e.g.
veterinarian, farmer)
▪ ↑ age
▪ Unpasteurized milk consumption
COMPLICATIONS
▪ Q fever pneumonia, chronic hepatitis,
osteomyelitis
▪ Infective endocarditis
▫ Pre-existing heart/valve disease
predisposes to endocarditis
development
▫ May have secondary, septic embolic
manifestation
SIGNS & SYMPTOMS
▪ Q fever (sudden onset)
▫ Fever, headache (often frontal), general
malaise, cough, anorexia, myalgia
▪ Pneumonia
▫ Cough, pleural effusion
▪ Hepatitis
▫ Hepatomegaly
DIAGNOSIS
LAB RESULTS
▪ ↑ serum hepatic enzymes, leukopenia/
leukocytosis, thrombocytopenia
▪ Immunofluorescent antibody assays:
detect IgM/IgG antibodies; differentiate
between acute, chronic infection
▫ IgM: detectable 4 days after symptom
onset
▫ IgG: detectable 9–14 days after
symptom onset
▫ Concentration of serum samples can
assist in diagnosis (esp. if vague clinical
presentation)
210
▪ PCR
▫ Blood/serum detection possible before
IgM serology peak
▪ Immunohistochemistry
▪ Culture
OTHER DIAGNOSTICS
▪ History
▫ Animal contact, occupation
▫ Trimethoprim-sulfamethoxazole:
pregnant individuals; treat even if
asymptomatic
▪ Chronic infection: prolonged therapy
▫ 18 months doxycycline,
hydroxychloroquine (monitor
serologic response across therapeutic
intervention; biannual ophthalmic
examinations required)
OTHER INTERVENTIONS
TREATMENT
MEDICATIONS
▪ Prompt antimicrobial treatment
▫ Doxycycline: effectiveness of
antibacterial agent, severity of
complications warrants use despite side
effects; shorter therapy for children (14
day course)
▪ Management
▫ Individuals with pre-existing
valvulopathy/cardiomyopathy;
echocardiogram
▪ Prevention
▫ Whole cell vaccine; recommended for
individuals working with farm animals
(e.g. farmers, slaughterhouse workers)
EHRLICHIA
osms.it/ehrlichia
PATHOLOGY & CAUSES
▪ Tick-borne, obligate intracellular bacteria
with leukocytic tropism, associated with
febrile disease with rare, serious neurologic
complication
▫ AKA human monocytic ehrlichiosis
(HME)
▪ Characteristics
▫ Small (0.5–1.5 micrometer) gramnegative cocci, (1.8 megabases)
genome
TYPES
▪ Ehrlichia ewingii
▫ Southeastern, central United States
▪ Ehrlichia chaffeensis
▫ Northeastern, midwestern United States
▪ Ehrlichia sennetsu
▫ Western Japan
Vector
▪ Amblyomma americanum (AKA lone star
tick)
▫ Ehrlichia ewingii, Ehrlichia chaffeensis
Life cycle
▪ Tick bite → blood circulation → leukocyte
infection, membrane attachment →
phagocytosis → replication (in early
endosome of leukocyte) → dysfunctional
vacuolization, immature lysosomal micelles
→ microcolony (AKA morulae) development
→ release into extracellular space after cell
lysis/exocytosis
Pathogenesis
▪ Direct leukocyte cell death
▪ Inflammatory response → perivascular
inflammatory infiltrates in multiple organ
system without organ failure/endothelial
damage
Disease: Sennetsu fever
211
Chapter 95 Rickettsial Diseases
RISK FACTORS
▪ Residence in/travel to western Japan
▪ Occupational exposure (e.g. military)
COMPLICATIONS
▪ Co-infection with Babesia spp./B.
burgdorferi
▪ Encephalitis, seizure
▫ Associated most with E. chaffeensis
▫ May result in persistent neurologic
deficit (rare; seen especially in children)
▪ Heart failure, respiratory insufficiency, renal
failure, shock
SIGNS & SYMPTOMS
▪ Sennetsu fever
▫ Abrupt-onset fever, chills, headache,
malaise, sore throat, myalgias,
arthralgias
▪ Atypical rash
▫ Maculopapular with occasional
petechiae; located on trunk (sparing
hands, feet)
▪ Generalized lymphadenopathy
▫ Most associated with E. sennetsu;
includes hepatosplenomegaly
▪ GI
▫ Associated with E. chaffeensis
▫ Anorexia, diarrhea, nausea, vomiting
▪ Leukopenia, thrombocytopenia, anemia,
hyponatremia
▪ Hepatic transaminitis: most common in E.
chaffeensis infection
▪ Cerebrospinal fluid (CSF) analysis:
pleocytosis (mononuclear cells with
morulae), ↑ protein
OTHER DIAGNOSTICS
▪ History
▫ Tick bite in endemic area
TREATMENT
MEDICATIONS
▪ Prompt antibacterial management:
poxycycline, chloramphenicol
OTHER INTERVENTIONS
Prevention
▪ Avoidance of tick habitats
▪ Careful inspection after outside activity
(esp. in spring, summer)
▫ Rapid discovery, removal < 24–48 hours
after bite → effective prophylaxis
▪ Proper skin application of insect repellants
DIAGNOSIS
LAB RESULTS
▪ Serology: indirect IFA
▫ Detection of IgG/IgM antibodies
(Ehrlichia species)
▫ If negative on acute serum testing,
repeat with convalescent serum
(confirms diagnosis if fourfold increase
in IgG antibody titer)
▪ Whole blood PCR: detects 16S rRNA gene
▪ Wright stain: morulae (Ehrlichia) in
leukocyte
▫ E. ewingii: in peripheral blood
granulocyte
▫ E. chaffeensis: in peripheral blood
monocyte
212
RICKETTSIA RICKETTSII (ROCKY
MOUNTAIN SPOTTED FEVER)
osms.it/rickettsia-rickettsii
PATHOLOGY & CAUSES
▪ Tick-borne, obligate intracellular, Gramnegative bacteria endemic to parts of North
America → potentially lethal febrile disease
▪ Characteristics
▫ Weakly gram-negative, nonmotile
coccobacillus; 0.7–2.0 micrometers:
cannot be visualized by traditional
staining methods/direct fluorescent
antibody techniques
▫ Bacterial contents: ribosome; single,
circular chromosome; microcapsule
surrounding cell wall (may be important
in pathogenicity)
Vectors
▪ Dermacentor variabilis (American dog tick)
▫ Eastern, South-central US
▪ Dermacentor andersoni (Rocky Mountain
wood tick)
▫ West of Mississippi River
▪ Rhipicephalus sanguineus (common brown
dog tick)
▫ Southwestern US
▪ Virulence of strain depends on tick’s
feeding status
▫ ↑ feeds → ↑ incubation at high
temperatures → ↑ extracellular slime
→ ↑ virulence (AKA reactivation
phenomenon)
Life cycle
▪ Tick bite (requires 6–10 hours of feeding)
→ proliferates by binary fission
▪ Grows in nucleus, cytoplasm of host cells
Pathogenesis
▪ Tropism for endothelial cells, downstream
systemic effects as sequelae
▪ Endothelial cell entry: rickettsial outer
membrane proteins (rOmps) interact with
▪
▪
▪
▪
▪
▪
lipopolysaccharides, surface-exposed
proteins (SEPs) for entry
▫ rOmps bind Ku70 (membrane protein)
→ activate Ku70 → recruit ubiquitin
ligase → ubiquitination of Ku70 →
act upon cAMP receptors protein
kinase A, Epac (exchange protein)
→ rearrangement of host cell actin
filaments → rickettsial endocytosis
Bacterial spread: R. rickettsii express
phospholipase D, tlyC → lyse phagosomal
membrane → entry into cytosol →
polymerization of host cell monomeric
actin filaments → invagination of host cell
membranes → passage into neighboring
cells
Further bacterial spread
▫ Filopodia (from host cell membranes)
assist in intercellular movement
▫ Bloodstream, lymphatic spread assist in
more distant infection sites
Small blood vessel injury (not entirely
elucidated)
▫ Associated with phospholipase A
activity, protease activity, free radicalinduced lipid peroxidation
▫ Cell necrosis (from other infected cells)
→ CD8+ T-cell response → endothelial
cell injury → immune, phagocytic
cellular response → lymphohistiocytic
vasculitis
Sequelae of small vessel injury: ↑
fluid in interstitial space → exposes
brain, lung parenchyma to devastating
pathophysiologic consequences
Ability to spread cell-to-cell without
causing obvious damage
▫ Rarely accumulate in large numbers
inside cells
Speeds of 4.8m/min
▫ Achieves speed via rapid recruitment,
polymerization of host cell actin
filaments
213
Chapter 95 Rickettsial Diseases
RISK FACTORS
▪ Residence in/travel to endemic areas (esp.
in spring, summer)
▪ ↑ age (peak: 40–64)
▪ Individuals who are biologically male
▪ Glucose-6-phosphate dehydrogenase
(G6PD) deficiency
COMPLICATIONS
▪ Skin necrosis at sites of terminal arterial
supply (e.g. fingers, toes, nose, ears,
genitals)
▪ Interstitial pneumonitis, myocarditis,
encephalitis
SIGNS & SYMPTOMS
▪ Early infection
▫ Fever, headache, malaise, myalgias,
arthralgias, nausea (without vomiting),
edema (esp. in children)
▪ Rash development (hallmark of infection)
▫ Blanching, erythematous rash
▫ Macules (1–4mm) → petechiae
▫ Ankles, wrist → truncal spread → palms,
soles rash (characteristic of late-stage
disease)
▪ Confusion, conjunctival erythema, seizures,
focal neurologic deficit
▪ Fundoscopic examination
▫ Retinal vein engorgement, arterial
occlusion, flame hemorrhage
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Interstitial infiltrates
Echocardiogram
▪ Minimal myocardial dysfunction with
normal capillary wedge pressure
▪ Consistent with noncardiogenic nature of
pulmonary edema (commonly present)
▪ Advanced disease
▫ Hyponatremia (sign of central nervous
system involvement); transaminitis; ↑
bilirubin; azotemia (due to hypovolemia);
↑ prothrombin, partial thromboplastin
times
▪ CSF
▫ Pleocytosis (monocytic,
polymorphonuclear predominance
possible), ↑ protein
OTHER DIAGNOSTICS
▪ History
▫ Residence in/travel to endemic area;
recollection of tick bite (only 30% of
individuals recollect bite)
TREATMENT
MEDICATIONS
▪ Early treatment: prompt, empiric
antimicrobial therapy with doxycycline
(first-line), chloramphenicol (second-line)
▫ Even if mild symptoms (due to
potential lethality of bacterial strain);
recommended for pregnant individuals,
children
▫ Goal: initiate < five days after symptom
onset
▫ Duration: until three days after
resolution of febrile illness; minimum
seven days
▪ Antiemetics, antimotility agents (individuals
intolerant of doxycycline)
OTHER INTERVENTIONS
▪ Hemodynamic monitoring in ICU setting;
respiratory support; renal replacement
therapy, blood transfusions
▪ Prevention
▫ Vigilant detection, early removal of ticks,
proper clothing
LAB RESULTS
▪ Thrombocytopenia (worsens with
progression of disease)
214
AfraTafreeh.com exclusive
RICKETTSIA TYPHI
(MURINE TYPHUS)
osms.it/rickettsia-typhi
PATHOLOGY & CAUSES
▪ Rat-borne zoonotic disease transmitted to
humans via flea; flu-like illness; minority of
individuals require ICU-level care
▫ AKA murine, endemic, flea-borne
typhus
Transmission
▪ Zoonotic reservoir: Rattus typhi
▪ Vector: Xenopsylla cheopis (AKA Oriental
rat flea)
Life cycle
▪ Flea feeds on infected rodent → lifetime
infection → fecal bacterial shedding →
human contact with flea feces through
breaks in skin barrier/inhalation → human
disease
▫ Further human infection occurs via body
lice passed human-to-human
▪ Replicates in high titers in yolk sacs of
embryonated chicken eggs
Pathogenesis (not well elucidated)
▪ Perivascular infiltration with lymphocytes,
macrophages, plasma, mast cells (on
biopsy)
▪ Vasculitis (rare)
▫ Accompanied by mural/intimal thrombi;
heart, lungs, kidneys, central nervous
system (CNS)
▪ Disease
▫ Mild, flu-like illness
RISK FACTORS
▪ Warmer climates, seaports, major
commercial areas
COMPLICATIONS
▪ Shock, sepsis, myocarditis, renal/respiratory
failure, severe hemolysis (associated with
G6PD deficiency)
▪ Neurological
▫ Meningitis, meningoencephalitis,
facial paralysis, hearing loss, ocular
abnormalities
SIGNS & SYMPTOMS
▪ Fever (8–16 days after exposure), chills,
headache (commonly frontal), myalgia, rash
(concurrent with fever; maculopapular, less
commonly petechial; spares face, palms,
soles), nausea, vomiting
DIAGNOSIS
LAB RESULTS
▪ ↑ erythrocyte sedimentation rate (ESR)
▪ Left shifted leukocyte count (absolute
neutropenia/lymphopenia possible)
▪ Abnormal hepatic enzymes
▪ Electrolytes
▫ Hyponatremia, hypokalemia, ↑ serum
creatinine
TREATMENT
MEDICATIONS
▪ Prompt doxycycline antimicrobial therapy
▪ Ciprofloxacin (viable alternative)
OTHER INTERVENTIONS
Prevention
▪ Rodent, flea eradication
▪ Rat-proofing homes, food service areas
▪ Protective clothing for occupational
exposure to rats
215
Chapter 2 Acyanotic Defects
NOTES
RODS
MICROBE OVERVIEW
▪ Rod shaped bacteria (bacilli), Gramnegative, non-spore forming
BACTEROIDES FRAGILIS
osms.it/bacteroides-fragilis
PATHOLOGY & CAUSES
▪ Flora of oral cavity, GI tract (primarily large
intestine), genitourinary tract of individuals
who are biologically female; responsible for
variety of infections
▪ Obligately anaerobic, non-motile
▪ Beta lactamase positive → resistance to
beta lactam antibiotics
▪ Fastidious → difficult to isolate
▪ Grows in 20% bile
▪ Usually involved in widespread
polymicrobial anaerobic Gram-negative
bacilli infections (AGNB)
▫ Intra-abdominal, skin/soft tissue,
pulmonary
RISK FACTORS
▪ Prior antibiotic use
▪ ↓ blood supply (e.g. trauma, malignancy,
surgery, shock, vascular disease)
▪ Tissue necrosis
▪ Disruption of intestinal mucosa
▪ High risk for aspiration pneumonia (e.g.
alcoholism, coma, stroke)
▪ Bronchial obstruction → lung abscesses
SIGNS & SYMPTOMS
▪ Foul-smelling discharge, tissue necrosis,
formation of abscesses, gas production in
tissues/discharges
DIAGNOSIS
DIAGNOSTIC IMAGING
CT scan
▪ Abscesses/presence of gas in infected site
▫ Highly suggestive of anaerobic infection
LAB RESULTS
▪ Rapid enzymatic test
▪ Polymerase chain reaction (PCR) assays
▪ Direct needle aspiration
216
TREATMENT
MEDICATIONS
▪ Antimicrobials
▫ Recovered anaerobes mixed with
aerobic organisms; clindamycin,
metronidazole, carbapenems,
tigecycline, beta-lactam/beta-lactamase
inhibitors, cefoxitin
SURGERY
▪ Drain abscesses, relieve obstructions
BARTONELLA HENSELAE
osms.it/bartonella-henselae
PATHOLOGY & CAUSES
▪ Zoonotic microbe; affects skin (most
common), regional lymph nodes,
internal organs (e.g. liver, spleen); small,
pleomorphic; fastidious, slow growing;
requires specific culture conditions; grows
in lysis centrifugation tubes; detected with
Warthin–Starry (WS)/silver stain
▪ Natural reservoir
▫ Domestic cats, usually young cats
▪ Mode of transmission
▫ Infection follows cat scratch, bite,
exposure to cat feces/fleas
Commonly associated diseases
▪ Cat-scratch disease (CSD)
▫ Usually affects children
▪ Bacillary angiomatosis (BA)
▫ Rare vasoproliferative disorder; usually
occurs in severely immunocompromised
individuals (e.g. due to HIV)
COMPLICATIONS
CSD
▪ Lymph nodes necrosis; can disseminate,
cause life-threatening complications
involving visceral organs (e.g. liver, spleen),
central nervous system (CNS), eyes
BA
▪ Potentially fatal if untreated
SIGNS & SYMPTOMS
▪ May be asymptomatic
CSD
▪ Usually develops 1–3 weeks after
inoculation
▪ Regional lymphadenopathy/lymphadenitis
(most common)
▫ Usually unilateral; lymph nodes drain
site of inoculation; tender, swollen
lymph nodes
▪ Rare
▫ Low grade fever, malaise, ↓ appetite,
abdominal aches
▪ Cutaneous erythematous lesion at site of
inoculation; regresses spontaneously after
1–4 weeks
▪ Visceral organs → hepatomegaly,
splenomegaly
▪ Eyes → neuroretinitis, parinaud
oculoglandular syndrome
▪ Central nervous system (CNS) →
encephalopathy
BA
▪ Usually affects skin → red papules, nodules/
subcutaneous masses
DIAGNOSIS
LAB RESULTS
▪ Difficult to culture; incubation takes 2–4
weeks
▪ PCR assays
217
Chapter 96 Rods
Biopsy
▪ CSD: granulomatous inflammation of lymph
nodes, stellate microabscesses; organisms
visualized using WS/silver stain
▪ BA: biopsy of lesions; Bartonella
demonstrated with WS stain
Serologic test
▪ Indirect immunofluorescence antibody
(IFA)/enzyme immunoassay (EIA)
Nonspecific findings
▪ ↑ erythrocyte sedimentation rate (ESR), ↑
C-reactive protein (CRP) in CSD
OTHER DIAGNOSTICS
▪ Clinical findings suggestive of Bartonella
infection with history of contact with cat
Figure 96.1 The histological appearance of
an lymph node excised from an individual
with cat-scratch disease. There are numerous
neutrophils which form characteristic stellate,
or star-shaped, microabscesses.
TREATMENT
MEDICATIONS
CSD
▪ Usually self-limited within 2–4 months;
antimicrobial therapy with azithromycin
prevents life-threatening complications;
alternative agents include doxycycline,
rifampin
BA
▪ Erythromycin, doxycycline, tetracycline
Figure 96.2 The histopathological
appearance of bacillary angiomatosis.
There is a proliferation of small capillaries
surrounded by mixed inflammatoru cells,
histiocytes and bacterial colonies.
218
ENTEROBACTER
osms.it/enterobacter
PATHOLOGY & CAUSES
▪ Opportunistic microbe; nosocomial,
various organ system infections;
Enterobacteriaceae family; motile,
nonfastidious; fast lactose fermenter, pink
colonies on MacConkey agar (lactose
containing medium); grows in aerobic,
anaerobic conditions, rapidly on selective,
nonselective agars; expresses fimbria;
hemolysin, urease positive
▪ Commonly isolated species: E. cloacae, E.
aerogenes
▪ Natural reservoir
▫ Soil, water, intestinal flora; occasionally
respiratory tract
▪ Mode of transmission
▫ Endogenous: transfer from flora to
adjacent sterile sites
▫ Exogenous: direct/indirect contact of
mucosal surfaces with Enterobacter
Commonly associated diseases
▪ Lower respiratory tract infections
▫ Tracheobronchitis, pneumonia, lung
abscess, empyema
▪ Urinary tract infections (UTIs) in
hospitalized individuals
▫ Cystitis, pyelonephritis, prostatitis
▪ Bloodstream infections (BSI)
▪ Skin, soft tissue infections
▫ Cellulitis, fasciitis, myositis, skin
abscesses, wound infections
▪ Intra-abdominal infections
▫ Abscesses, peritonitis following
intestinal perforation/surgery
▪ Uncommon
▫ Endocarditis, septic arthritis,
osteomyelitis, CNS, ocular infections
▪ Prolonged hospitalization
▪ Invasive procedures (e.g. post-surgery
infections)
▪ Prior antibiotic therapy
▪ Invasive devices (e.g. venous
catheterization)
▪ Underlying conditions
▫ Malignancy, chronic obstructive
pulmonary disease (COPD), diabetes
mellitus (DM), burns
▪ Mechanical ventilation
▪ Immunosuppression
COMPLICATIONS
▪ Septic shock
SIGNS & SYMPTOMS
▪ Non-specific, depends upon organ system
affected
▪ UTIs
▫ Frequency, urgency of urination, dysuria
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray/CT scan
▪ Respiratory infections
Abdomen CT scan/ultrasound
▪ Abdominal infections
LAB RESULTS
▪ Urinalysis for UTIs
Microbe identification
▪ Gram staining, culture
RISK FACTORS
▪ More common in neonates, elderly
individuals
219
Chapter 96 Rods
TREATMENT
MEDICATIONS
▪ Antimicrobials
▫ Carbapenems, aminoglycosides,
fluoroquinolones, trimethoprimsulfamethoxazole, polymyxins
SURGERY
▪ Drain abscesses
OTHER INTERVENTIONS
▪ Remove invasive devices (e.g. central
venous catheters)
ESCHERICHIA COLI
osms.it/escherichia-coli
PATHOLOGY & CAUSES
▪ Frequent cause of variety of infections;
Enterobacteriaceae family; encapsulated,
motile; certain strains hemolytic (betahemolytic on blood agar); fast lactose
fermenter (pink colonies on MacConkey
agar); nonfastidious; beta-galactosidase
positive (breaks down lactose into glucose,
galactose); iron uptake system
▪ AKA E.Coli
▪ Natural reservoir
▫ Intestinal, vaginal flora
▪ Mode of transmission
▫ Person-to-person contact, contaminated
food/water, dislocation from intestinal
tract
▪ Growing conditions
▫ On selective media; aerobic, anaerobic;
eosin-methylene blue (EMB) agar for
isolation (colonies with green metallic
sheen); 15–45°C/59–113°F (some
strains more heat resistant)
▪ Virulence factors
▫ Fimbriae, K-capsule, lipopolysaccharides
(LPS) endotoxin
▪ Pathogenic factors
▫ Fimbriae → cystitis, pyelonephritis
▫ K-capsule → pneumonia, neonatal
meningitis
▫ LPS endotoxin → septic shock
Commonly associated diseases
▪ Genitourinary tract infections
▫ Leading cause of cystitis, pyelonephritis,
prostatitis
▪ Intra-abdominal infections
▫ Enteric infections, abscesses,
cholecystitis, spontaneous bacterial
peritonitis (E. coli most common cause)
▪ Pneumonia
▪ Meningitis (in neonates)
TYPES
Enteroinvasive (EIEC)
▪ Invades intestinal mucosa → necrosis,
inflammation → dysentery
Enterotoxigenic (ETEC)
▪ Heat-labile toxin (LT) → over-activates
adenylate cyclase (cAMP) → ↑ Cl- secretion
in gut, water efflux
▪ Heat-stable toxin (ST) → overactivates
guanylate cyclase (cGMP) → ↓ resorption of
NaCl, water in gut
▪ Produces LT, ST → travellers’ diarrhea
(watery), mimics Vibrio cholerae illness
Enteropathogenic (EPEC)
▪ Adheres to apical surface, flattens villi,
prevents absorption → diarrhea
Enterohemorrhagic (EHEC)
▪ Most commonly isolated serotype O157:H7
▪ Produces Shiga-like toxin → enhances
cytokine release
▫ Necrosis, inflammation → dysentery
▫ Endothelial damage → formation
of microthrombi → hemolysis
220
(microangiopathic hemolytic
anemia), platelet consumption
(thrombocytopenia), ↓ renal blood flow
(acute renal failure) → hemolytic uremic
syndrome (HUS)
▫ O157:H7 most common serotype
SIGNS & SYMPTOMS
▪ Depends on organ system affected
▪ Clinical manifestations generally
nonspecific, except in enteric infections
▫ EHEC: bloody diarrhea, malaise, fever
▫ EIEC: dysentery, mimics shigellosis;
bloody diarrhea, abdominal pain,
tenesmus
▫ ETEC: watery diarrhea (travellers’
diarrhea), nausea, abdominal pain
▫ EPEC: fever, watery diarrhea; can last >
two weeks; typically affects children
DIAGNOSIS
LAB RESULTS
▪ Culture to isolate E. coli, gram staining, ↑
fecal leukocytes (except ETEC), PCR assays
▪ Complete blood count
▫ Leukocytosis
▪ Urinalysis in UTIs
▪ Pyuria
▫ Leukocyte esterase + → evidence of
white blood cell (WBC) activity
▪ Proteinuria, bacteriuria
▪ Nitrite test + → ↓ urinary nitrates → marker
of infection
TREATMENT
MEDICATIONS
▪ Antimicrobial treatment (UTIs, pneumonia,
meningitis, intra-abdominal infections,
sepsis)
▫ Third-generation cephalosporins,
quinolones, doxycycline, trimethoprim/
sulfamethoxazole (TMP/SMZ)
▪ Antidiarrheal medications
▫ Contraindicated in children, EIEC
infections
▪ EHEC
▫ Antibiotics contraindicated; can
increase risk of HUS, thrombotic
thrombocytopenic purpura (TTP)
complications
▪ ETEC
▫ Antibiotics useful; reduce diarrhea
duration
OTHER INTERVENTIONS
▪ Most enteric infections self-limited,
managed conservatively
▫ Fluid, electrolytes correction
221
Chapter 96 Rods
KLEBSIELLA PNEUMONIAE
osms.it/klebsiella-pneumoniae
PATHOLOGY & CAUSES
▪ Causative agent of infections, usually occur
in immunocompromised individuals; major
cause of hospital-acquired infections;
Enterobacteriaceae family, facultative
anaerobe, nonmotile; possesses prominent
polysaccharide capsule; beta-lactamase
positive (resistant to ampicillin, amoxicillin;
susceptible to cephalosporins); urease
positive, lactose fermenter, iron uptake
system
▪ Natural reservoir
▫ Flora of human mouth, intestine; soil,
water
Commonly associated diseases
▪ Pulmonary infections
▫ Pneumonia, empyema, lung abscesses,
bacteremia
▪ UTIs
▫ Cystitis, pyelonephritis, prostatitis,
abscesses
▪ Pyogenic liver/splenic abscesses
▪ Infective endocarditis
▪ Spontaneous bacterial peritonitis
▪ Endophthalmitis
▪ CNS infections
▪ Meningitis, brain abscesses
▪ Deep neck infections
▪ Skin, soft tissue infections
COMPLICATIONS
▪ Pneumonia can be fatal regardless of
treatment
SIGNS & SYMPTOMS
▪ Pneumonia presents as
bronchopneumonia/bronchitis (acute onset
of symptoms)
▫ Productive cough (e.g. mucoid; bloody
sputum, AKA “currant jelly sputum”),
high grade fever, chills, chest pain,
dyspnea, tachypnea, crackles
▪ UTIS (frequency, urgency, dysuria)
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Findings suggestive of pneumonia
LAB RESULTS
▪ Complete blood count
▫ Leukocytosis
▪ Specimen culture
▫ E.g. blood, sputum, urine
▫ Gram stain: Gram-negative, rod-shaped
capsule appears as clear space
TREATMENT
RISK FACTORS
▪ Prolonged hospitalization
▪ Prior use of antibiotics
▪ Prolonged use of invasive devices (e.g.
catheters)
▪ Underlying medical conditions
▫ Alcoholism, DM, underlying malignancy,
hepatobiliary disease, COPD, renal
failure
▪ Use of glucocorticoids
MEDICATIONS
▪ Antimicrobial therapy
▫ Beta-lactams with beta-lactamase
inhibitors, cephalosporins,
fluoroquinolones, aminoglycosides,
carbapenems
SURGERY
▪ Drain abscesses
▪ Debride necrotic issues
222
LEGIONELLA PNEUMOPHILA
osms.it/legionella-pneumophila
PATHOLOGY & CAUSES
SIGNS & SYMPTOMS
▪ Gram-negative, facultative intracellular
(stain poorly with Gram stain); best
visualized by silver stain; appears in
specimens as short rod; longer, filamentous
in culture; grows best on charcoal yeast
extract medium with cysteine, iron;
serogroup 1 most commonly associated
with human infections
▪ Natural reservoir
▫ Water sources (e.g. air conditioning
systems, tubing systems of domestic
water supplies, water-cooling towers)
▪ Mode of transmission
▫ Water aerosol inhalation
Legionnaires’ disease
▪ Incubation period
▫ 2–10 days
▪ Atypical pneumonia
▫ Mild cough (may cough bloody mucus),
high grade fever, chills, dyspnea, chest
pain, rales
▪ GI
▫ Diarrhea, nausea, vomiting, abdominal
pain
▪ CNS
▫ Confusion, lethargy, headache, focal
neurologic signs, hallucinations
Commonly associated diseases
▪ Legionnaires’ disease
▫ Atypical form of pneumonia
▪ Pontiac fever
▪ Rare self-limited upper respiratory tract
infection
▪ Extrapulmonary disease (rare)
▫ Heart most common extrapulmonary
site (e.g. myocarditis, endocarditis)
Pontiac fever
▪ Acute onset
▪ Mild, febrile, flu-like syndrome
RISK FACTORS
▪ ↑ age, immunosuppression, smoking,
chronic lung disease, organ transplant,
renal failure, cardiac disease
COMPLICATIONS
▪ Pneumonia
▫ Progresses rapidly, can be fatal (esp.
immunosuppressed individuals)
▪ Renal failure
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray/CT scan
▪ Legionnaires’ disease
▫ Unilateral
▪ Diffuse, patchy inflammation of interstitium
→ consolidation involving ≥ one lobe
LAB RESULTS
▪ Nonspecific findings
▫ Hyponatremia, thrombocytopenia,
leukocytosis, hypophosphatemia
▪ Culture of respiratory specimens
▫ Many neutrophils, few microorganisms
▪ Urinary antigen test using enzyme-linked
immunosorbent assay (test of choice along
with culture)
▫ Rapid; sensitive, specific; legionella
persists in urine for weeks
223
Chapter 96 Rods
▪ Fluorescent antibody test on sputum
specimens
▪ Serological tests
▫ Serum antibodies develop after 8–10
days; fourfold rise in titre/single high titre
▪ PCR using sputum/other specimens (less
sensitive than culture)
TREATMENT
MEDICATIONS
▪ Atypical pneumonia
▫ Macrolides, tetracyclines.
fluoroquinolones, rifampicin
OTHER INTERVENTIONS
▪ Eradication of organism from responsible
water source to control disease
NONTYPHOIDAL SALMONELLA
osms.it/nontyphoidal-salmonella
PATHOLOGY & CAUSES
▪ Causes salmonellosis (foodborne
gastroenteritis); Enterobacteriaceae
family; motile; encapsulated; facultative
intracellular, anaerobes; nonlactose
fermenter; oxidase-; possesses type III
secretion system, iron uptake system; high
infectious dose; H2S production on TSI
agar; produces endotoxin
▪ Most common human pathogen
▫ S.enteritidis
▪ Natural reservoir
▫ Humans, animals; poultry, eggs, pets,
turtles common sources
▪ Mode of transmission
▫ Food (e.g. undercooked meat, poultry,
eggs, milk), fecal-oral route
RISK FACTORS
▪ Age (usually affects young children,
older adults); season (peak incidence
in summer, fall); low gastric acidity
(atrophic gastritis, use of acid-suppressive
agents); immunodeficiency; sickle cell
disease; hemolytic anemia; alcoholism;
cardiovascular dysfunction; malignancies;
IV drug use
COMPLICATIONS
▪ Can disseminate via blood → extraintestinal
manifestations (e.g. endocarditis, vascular
infections, cholecystitis, hepatic/splenic
abscesses)
SIGNS & SYMPTOMS
▪ Present 24–48 hours after food ingestion
▪ Loose stools/diarrhea often bloody (can
persist for two weeks)
▪ Nausea, vomiting
▪ Fever (resolves within two days)
▪ Abdominal cramps
DIAGNOSIS
LAB RESULTS
▪ Stool culture, ↑ fecal leukocytes, blood
cultures to detect bacteremia; bone marrow
aspiration, culture
TREATMENT
MEDICATIONS
▪ Antimicrobial therapy
▫ Generally not required, prolongs
duration of fecal shedding of Salmonella,
doesn’t shorten duration of symptoms
224
▪ Antibiotics
▫ Only indicated in immunocompromised
individuals; antimicrobial agents (e.g.
ciprofloxacin)
PROTEUS MIRABILIS
osms.it/proteus-mirabilis
PATHOLOGY & CAUSES
▪ Common cause of community acquired,
nosocomial UTIs (e.g. primarily cystitis,
pyelonephritis)
▪ Enterobacteriaceae family; highly
motile; nonlactose fermenters; oxidase
negative; expresses mannose-resistant
hemagglutination, calcium dependent/
independent hemolysins; swarming growth
in discontinuous manner on agar media;
H2S production on TSI agar; resistance to
polymyxins, tetracyclines
▪ Urease positive → converts urea to
ammonia, CO2 → ↑ urine pH → ammonia
combines with magnesium, phosphate →
form magnesium-ammonium-phosphate
(struvite) stones
▪ Pr. mirabilis causes 90% of infections
▪ Natural reservoir
▫ Intestinal flora
▪ Virulence factors
▫ Fimbria (important for adherence to
tissue)
Commonly associated diseases
▪ Urethritis, acute prostatitis, pyogenic liver
abscesses
RISK FACTORS
▪ Hospitalization; ↑ age (most common in
elderly); multiple prior UTIs; prior use of
antibiotics; urinary tract surgery; urinary
catheterization; structural abnormalities/
obstructions of urinary tract; underlying
medical conditions (e.g. DM, chronic kidney
disease); neurogenic bladder; frequent
sexual activity
COMPLICATIONS
▪ Abscesses, sepsis, meningitis, chronic
pyelonephritis, xanthogranulomatous
pyelonephritis (chronic destructive
granulomatous process of renal
parenchyma)
SIGNS & SYMPTOMS
▪ Nonspecific, disease-dependent:
symptoms commonly associated with
urethritis, acute prostatitis, pyogenic liver
abscesses
DIAGNOSIS
DIAGNOSTIC IMAGING
Ultrasound/CT scan
▪ Kidneys; indicated if therapy fails
X-ray
▪ Struvite stones appear radiopaque
LAB RESULTS
▪ Urine culture to isolate Proteus
▪ Complete blood count
▫ Leukocytosis
▪ Urinalysis
▫ Alkaline urine pH (> 7.0), pyuria,
bacteriuria
225
Chapter 96 Rods
TREATMENT
MEDICATIONS
▪ Uncomplicated UTIs
▫ Cystitis: TMP/SMX, nitrofurantoin,
quinolones, fosfomycin
▫ Pyelonephritis: quinolone/ampicillin,
gentamicin/third-generation
cephalosporin
▪ Complicated UTIs
▫ E.g. diabetes, nephrolithiasis, pregnancy,
anatomic abnormalities of urinary tract
▫ Therapy should be prolonged
SURGERY
▪ Drainage of collections (e.g. perinephric
abscesses)
▪ Removal of struvite renal calculus
Figure 96.3 Proteus species spread in a
swarming fashion on an agar plate.
PSEUDOMONAS AERUGINOSA
osms.it/pseudomonas-aeruginosa
PATHOLOGY & CAUSES
▪ One of most common causes of hospitalacquired infections in immunocompromised
individuals; motile; aerobic; nonlactose
fermenter (derives energy from
carbohydrates by oxidation); nonfastidious;
oxidase, catalase, elastase, leukocidin,
hemolysin positive; resistant to many
antibiotics
▪ Grows on variety of culture media
▫ Colonies greenish-blue due to
production of procyanin (blue),
pyoverdin (yellow-green); fruity, grapelike odor
▪ Natural reservoir
▫ Water, soil sources (e.g. rivers, ponds),
animals, plants, hospital equipment;
hospitalized individuals asymptomatic
carriers
▪ Mode of transmission
▫ Direct contact with contaminated
materials/infected individuals
▪ Virulence factors
▫ Exotoxin A → inactivates elongation
factor (EF-2) → inhibition of protein
synthesis → death of host cells
▫ Phospholipase C → degrades
membranes
▫ Endotoxin → fever, shock
▫ Mucoid exopolysaccharide → biofilm
formation → protection from immune
system
Commonly associated diseases
▪ Pneumonia; sepsis; genitourinary tract
infections; skin, soft tissue infections; ear
infections (e.g. external otitis, chronic otitis
media); eye infections (e.g. keratitis); GI
infections; bone, joint infections (usually
affects vertebral column); infective
endocarditis
RISK FACTORS
▪ Prolonged hospitalization, catheterization,
IV drug use, severe burns, contact lenses,
eye trauma
226
▪ Mechanical ventilation/endotracheal
intubation
▫ P. aeruginosa second most common
cause of ventilator-associated
pneumonia
▪ Cystic fibrosis (CF) → chronic infection with
P. aeruginosa
▪ Immune system deficits (e.g. neutropenia,
diabetes)
COMPLICATIONS
▪ Sepsis/necrotizing pneumonia can be fatal
in immunocompromised individuals
▪ Chronic infections in CF → bronchiectasis,
pulmonary fibrosis → pulmonary failure
▪ Disseminated intravascular coagulation due
to sepsis
▪ Eye infections → vision loss
SIGNS & SYMPTOMS
▪ Generally nonspecific, depend on organ
system
▪ Eye infections (esp. cornea) extremely
painful, rapidly destructive
▪ Ecthyma gangrenosum
▫ Due to sepsis, typically appears in
immunocompromised individuals
▫ Well-demarcated, black, necrotic
cutaneous lesion; rapidly progressive
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Pneumonia
▫ Bilateral bronchopneumonia with
nodular infiltrates with/without pleural
effusion
LAB RESULTS
▪ Culture (blood/other specimens) on
selective media
▫ Enhances production of procyanin
▪ Oxidase test
▪ PCR assays
▪ Complete blood count
▫ Leukocytosis
▪ Serological tests
▫ Only individuals with CF/COPD
TREATMENT
MEDICATIONS
▪ Antimicrobial therapy
▫ Extended-spectrum penicillins, betalactamase inhibitors, aminoglycosides,
carbapenems, monobactams,
polymyxins, fluoroquinolones, third/
fourth-generation cephalosporins
Figure 96.4 Pseudomonas species will
adopt a greenish hue when cultured on
cetrimide agar.
227
Chapter 96 Rods
SERRATIA MARCESCENS
osms.it/serratia-marcescens
PATHOLOGY & CAUSES
▪ Opportunistic microbe; uncommon cause
of variety of hospital-acquired infections;
Enterobacteriaceae family; motile; aerobic;
slow, weak lactose fermenter; urease
positive (struvite stones in individuals with
recurrent UTIs); catalase positive; grows on
media at 30–37°C/86–99°F, utilize most
carbohydrates with production of acid, gas;
produces red pigment (prodigiosin) colonies
on agar; often resistant to commonly used
antibiotics (e.g. ampicillin, macrolides, firstgeneration cephalosporins)
▪ Natural reservoir
▫ Water, soil sources
▪ Virulence factors
▫ Fimbria, cell surface components, longchain LPS, hemolysin
Commonly associated diseases
▪ Respiratory infections (e.g. pneumonia),
UTIs, wound infections, CNS infections
(e.g. meningitis, cerebral abscesses),
intra-abdominal infections, septicemia,
endocarditis, otitis externa
RISK FACTORS
▪ Neonates, infants
▪ More common in individuals who are
biologically male
▪ Prolonged hospitalization; catheterization
(e.g. intravenous, urinary catheters);
mechanical ventilation; recent surgery/
obstruction of urinary tract; cardiac
valve replacement; IV drug use; immune
system deficits (e.g. neutropenia, chronic
granulomatous disease, DM); head trauma/
brain surgery; contact lenses
COMPLICATIONS
▪ Septic shock, stroke (due to endocarditis)
SIGNS & SYMPTOMS
▪ Nonspecific, depend upon disease
▪ Symptoms associated with respiratory
infections, UTIs, wound infections, CNS
infections, intra-abdominal infections,
septicemia, endocarditis, otitis externa
DIAGNOSIS
DIAGNOSTICS IMAGING
Chest X-ray
▪ Pneumonia
Abdominal ultrasound/CT scan
▪ Abnormalities of urinary tract, intraabdominal infections, abscesses
Echocardiography
▪ Valvular vegetations
Brain CT scan
▪ Follow with lumbar puncture in individuals
with suspected meningitis
Figure 96.5 Serratia marcessens grows red
colonies when cultured on agar.
228
LAB RESULTS
▪ Complete blood count
▫ Leukocytosis, ↑ neutrophils with left
shift, possible anemia
TREATMENT
MEDICATIONS
▪ Antibiotic therapy
▫ Aminoglycosides with antipseudomonal
beta-lactam
▫ If infection persists, fluoroquinolones/
carbapenems
SURGERY
▪ Drainage of collections (e.g. abscesses)
SHIGELLA
osms.it/shigella
PATHOLOGY & CAUSES
▪ Causes acute infectious diarrhea
(shigellosis), one of most common causes
of bloody diarrhea; Enterobacteriaceae
family; facultative anaerobe; nonmotile;
nonlactose fermenter; urease, oxidase
negative; resistance to low pH of gastric
acids; highly virulent; possesses virulence
plasmids; invades colonic mucosa via
microfold (M) cells of Peyer patches
▪ Natural reservoir
▫ Human only; part of intestinal tract flora
▪ Mode of transmission
▫ Usually fecal-oral route, contaminated
water, food; easy person-to-person
spread
TYPES
▪ S. dysenteriae: serogroup A
▫ Releases Shiga toxin, causes most
severe form of shigellosis
▪ S. flexneri: serogroup B
▪ S. boydii: serogroup C
▪ S. sonnei: serogroup D
COMPLICATIONS
hypokalemia)
▪ Reiter’s syndrome
▫ Reactive arthritis, urethritis,
conjunctivitis
▫ Affects HLA-B27+ individuals
▪ Hemolytic-uremic syndrome
▫ Hemolytic anemia, thrombocytopenia,
acute renal failure
▫ Usually in infections due to S.
dysenteriae
▪ Toxic megacolon
▪ Intestinal obstruction
SIGNS & SYMPTOMS
▪ Incubation period
▫ 2–3 days/one week
▪ Sudden onset
▫ Abdominal pain
▫ Diarrhea that is initially watery,
progresses to bloody (bacillary
dysentery) in 50% of cases
▫ Tenesmus
▫ Fever, malaise, headache, anorexia
▪ Less common
▫ Nausea, vomiting
▪ Dehydration (due to fluid loss)
▪ Electrolyte imbalance (e.g. hyponatremia,
229
Chapter 96 Rods
▪ Subacute presentation in minority of
individuals
▫ Several weeks of waxing/waning
diarrhea
DIAGNOSIS
DIAGNOSTIC IMAGING
Colonoscopy
▪ Hemorrhagic, ulcerated mucosa
▪ Most prominent in left colon, ileum also
involved
▪ Pseudomembranes
TREATMENT
MEDICATIONS
▪ Antimicrobial therapy
▫ Shorten duration of symptoms, reduce
fecal shedding of organisms, risk of
complications; azithromycin orally, thirdgeneration cephalosporin parenterally
▪ Antidiarrheal medications contraindicated
▫ Delay fecal shedding of Shigella
OTHER INTERVENTIONS
▪ Most cases self limited (resolve within one
week), do not require antibiotics
LAB RESULTS
▪ Stool culture
▪ ↑ leukocytes, blood in feces
▪ PCR testing of stools
YERSINIA MARCESCENS
osms.it/yersinia-marcescens
PATHOLOGY & CAUSES
▪ Enterobacteriaceae family; facultative
anaerobe, intracellular; motility depends
on temperature (motile at 25°C/77°F,
nonmotile at 37°C/99°F); nonlactose
fermenter; oxidase negative
▪ Important human pathogens
▫ Y. pestis, Y.enterocolitica
▪ Natural reservoir
▫ Y. pestis: ground squirrels, gerbils, voles,
rats
▫ Y. enterocolitica: cattle, deer, pigs, birds,
pigs
▪ Virulence factors
▫ Adhesins to bind to host cell beta
1 integrins, endotoxin, coagulase,
fibrinolysin, iron uptake system
(mediates iron capture, transport)
TYPES
Yersinia enterocolitica
▪ Causes yersiniosis
▫ Gastrointestinal infections (e.g. ileum,
appendix, right colon)
▪ Common types
▫ Enterocolitis, terminal ileitis, mesenteric
lymphadenitis, pseudoappendicitis
▪ Mode of transmission
▫ Pet feces, inadequately cooked foods,
contaminated pork, milk, water
Yersinia pestis
▪ Causes human plague
▫ Zoonotic infection, highly fatal if
untreated
▪ Subtypes
▫ Wild (sylvatic), urban plague (typically
transmitted from rats, more severe)
230
▪ Clinical forms
▫ Bubonic (more common), septicaemic,
pneumonic (least common, high
mortality rate)
▪ Mode of transmission
▫ Fleas from rats, other rodents
COMPLICATIONS
Yersinia enterocolitica
▪ Reiter’s syndrome, myocarditis, erythema
nodosum, kidney disease, mucosal
ulceration, bowel necrosis (due to
mesenteric vessel thrombosis), septicemia
(fatal if untreated)
Yersinia pestis
▪ Bubonic plague → sepsis → secondary
pneumonic plague/meningitis
▪ Systemic plague → hypotension,
disseminated intravascular coagulation,
multiorgan failure
▪ Pneumonic plague (rapidly fatal if
untreated)
SIGNS & SYMPTOMS
Yersinia enterocolitica
▪ Enterocolitis (most common)
▫ Abdominal pain, acute diarrhea, low
grade fever, nausea, vomiting
▪ Abdominal tenderness
▫ Right lower quadrant, mimics
appendicitis (pseudoappendicitis)
▪ Extraintestinal features of pharyngitis,
arthralgia, erythema nodosum (painful, red/
purple lesions, resolve spontaneously)
Yersinia pestis
▪ Incubation period
▫ 2–8 days
▪ Bubonic plague (sudden onset)
▫ fever; painful lymphadenopathy (bubos),
inguinal lymph nodes frequently
involved; chills, fatigue
▪ Septicemic plague
▫ Fever; malaise, GI symptoms (nausea,
vomiting, diarrhea)
▪ Pneumonic plague (sudden onset):
▫ Dyspnea, high grade fever, chest pain,
cough with blood-containing sputum
DIAGNOSIS
DIAGNOSTIC IMAGING
Ultrasound/CT scan
▪ Yersinia enterocolitica: exclude appendicitis
Colonoscopy
▪ Yersinia enterocolitica: aphthoid lesions in
cecum; elevations, ulcers in terminal ileum
Chest X-ray
▪ Yersinia pestis: pneumonic plague
LAB RESULTS
Yersinia enterocolitica
▪ Identification of microbe
▪ Stool culture
▫ Cefsulodin-irgasan-novobiocin (CIN)
agar
▫ In extraintestinal disease, cultures of
lymph nodes/blood may be positive
▪ PCR assays
▪ ↑ leukocytes in stool, blood
▪ Serological tests
▫ Tube agglutination, enzyme-linked
immunosorbent assay (ELISA)
Yersinia pestis
▪ Nonspecific findings
▫ Leukocytosis, thrombocytopenia
▪ Isolation of organism in blood culture
▫ Positive cultures in individuals with
bubonic, septicemic plague
▪ Peripheral blood smear
▫ Wright–Giemsa stain reveals rodshaped bacteria, Wayson stain reveals
characteristic “safety pin” appearance
▪ Rapid antigen testing in sputum/serum
▪ Serological tests
▫ Fourfold rise in serum antibody titers
between acute, convalescent phase
▪ PCR assays
231
Chapter 96 Rods
OTHER DIAGNOSTICS
Yersinia pestis
▪ Clinical findings suggestive of plague,
history of traveling to plague-endemic
areas
TREATMENT
MEDICATIONS
Yersinia enterocolitica
▪ Antimicrobial therapy
▫ TMP/SMX, aminoglycosides
▫ Alternative agents: third-generation
cephalosporins, tetracyclines,
fluoroquinolones
Figure 96.6 The black, necrotic fingers of a
man infected with Yersinia pestis.
Yersinia pestis
▪ Antimicrobial therapy
▫ Aminoglycosides (streptomycin/
gentamicin)
▫ Alternative agents: doxycycline,
tetracycline, fluoroquinolones,
chloramphenicol
OTHER INTERVENTIONS
Yersinia enterocolitica
▪ ↑ fluid intake, good nutrition
232
NOTES
NOTES
SPIROCHETES
MICROBE OVERVIEW
Morphology
▪ Thin-walled, Gram-negative flexible spiral
rods
▪ Length: 5–250μm; diameter: 0.1–0.6μm
▪ Unique double membrane separated by
periplasmic space
▪ Corkscrew-like motility via axial filament
situated lengthwise between inner and
outer membranes (endoflagella)
Replication
▪ Reproduction: sexual transverse binary
fission
BORRELIA BURGDORFERI
(LYME DISEASE)
osms.it/lyme-disease
PATHOLOGY & CAUSES
▪ An obligate parasite that causes Lyme
disease, a systemic inflammatory disease
▪ Non-spore forming
▪ Life cycle includes mammals, birds, and
ticks (genus Ixodes), principally in North
America
▪ Killed when exposed to hypotonic
or hypertonic environments, drying,
disinfectants (e.g. bleach), or temperatures
> 50°C/122°F
▪ Transmission
▫ Via tick bite → enters blood → spreads
to tissues and organs; especially joints,
heart, nervous system
▪ Infection progresses through three stages
1. Localized disease (occurs 3–30 days
after exposure)
2. Disseminated disease (days to
months after exposure; multisystem
involvement)
3. Late/chronic disease (months to years
after exposure)
RISK FACTORS
▪ Exposure to ticks in endemic areas
▫ Living in areas bordering forests
▫ Outdoor employment, recreation
▪ Most cases occur in late spring to summer
COMPLICATIONS
▪ Meningitis, cranial neuropathy
▪ Lyme carditis
▫ AV block, myopericarditis
▪ Chronic joint inflammation
▪ Post–Lyme disease syndrome
SIGNS & SYMPTOMS
Early signs/symptoms of localized disease
▪ Erythema migrans (EM) rash
▫ Non-painful, gradually expanding
“bull’s-eye” rash appearing at the site of
tick bite; feels warm to palpation; may
itch
233
Chapter 97 Spirochetes
▪ Constitutional
▫ Low grade fever, chills, headache,
fatigue, myalgia, arthralgia,
lymphadenopathy
Disseminated signs/symptoms
▪ Severe headaches and neck stiffness
▪ Formation of additional EM
▪ Joint pain and swelling; especially knees,
other large joints
▪ Facial palsy
▪ Palpitations (Lyme carditis)
▪ Episodic dizziness, dyspnea
▪ Pain
▫ Shooting pains, numbness, or tingling in
the hands or feet
▪ Short-term memory loss
Late/chronic disease
▪ Presence of nonspecific symptoms (e.g.
headache, fatigue, joint pain) that persists
after treatment for Lyme disease
DIAGNOSIS
LAB RESULTS
CDC testing criteria
▪ Two tiered testing for lyme disease
▫ First test: enzyme immunoassay (EIA) or
immunofluorescence assay (IFA)
▫ Second test (as needed): IgM and/or IgG
western blot
Other laboratory findings
▪ Blood chemistry
▫ ↑ ESR, serum creatine phosphokinase,
aspartate aminotransferase (AST) and/
or alanine aminotransferase (ALT)
▪ Blood studies
▫ Anemia, leukocytosis, thrombocytopenia
OTHER DIAGNOSTICS
▪ History of exposure to ticks in an endemic
area and characteristic clinical presentation
TREATMENT
MEDICATIONS
▪ Antibiotics
▫ Doxycycline, amoxicillin, or cefuroxime
axetil
Figure 97.1 A bulls-eye-shaped rash, known
as erythema migrans, at the site of infection
with Borellia burgdorferi, the causative agent
of Lyme disease.
234
BORRELIA SPECIES
(RELAPSING FEVER)
osms.it/borrelia-species
PATHOLOGY & CAUSES
▪ Relapsing fever: bacterial infection caused
by Borrelia spirochetes
Tick-borne relapsing fever (TBRF)
▪ Endemic
▪ Found in endemic areas
▫ Mountainous areas of North America,
plateau regions of Mexico, Central and
South America, Mediterranean, central
Asia, Africa
▪ Caused primarily by Borrelia hermsii,
Borrelia turicatae, Borrelia parkeri in North
America
▪ Spread by soft tick species Ornithodoros
parkeri and Ornithodoros turicata
▪ Risk factors
▫ Occupying rodent-infested cabins,
caves, or other dwellings
▫ Camping near rodent nests
▪ Tick attaches to human host (usually at
night during sleep) → bites and feeds on
blood → saliva and spirochete enter host’s
circulation
Louse-borne relapsing fever (LBRF)
▪ Epidemic
▪ Caused by Borrelia recurrentis
▪ Spread person-to-person via body louse
▪ Risk factors
▫ Primarily seen in low resource countries
▫ Famines, wars; causes epidemics
among refugees, migrant populations
▫ Homelessness (exposure to lice)
▫ Poor hygiene
▪ B. recurrentis grows in the body cavity
Pediculus humanus corporis → human
host crushes louse/louse feces with fingers
→ spirochete enters either through bite
site, through breaks in the skin caused by
scratching, or through conjunctiva when
fingers touch eyes
After entry into human host
▪ Spirochete divides every 6–12 hours (can
number up to 106–108 per mL) → leaves
blood and enters tissues: brain, eye, inner
ear, liver, heart, testes, and other organs
Pattern of intermittent illness
▪ Relapsing fever caused by spirochetemia
▪ Characterized by recurrent episodes of
fever and constitutional symptoms with
intermittent periods of general well-being
▫ Interval range between fevers is 4–14
days
▫ Pattern of intermittent illness caused
by outer membrane lipoproteins called
variable major proteins (VMPs)
▫ Employed as multiphasic antigens to
evade host adaptive immunity
▫ TBRF: multiple febrile periods last 1–3
days each
▫ LBRF: first episode lasts 3–6 days,
followed by a single milder episode
Complications
▪ Neurological
▫ Meningitis, subarachnoid hemorrhage,
cranial nerve neuritis, Bell’s palsy,
hearing loss
▪ Ocular
▫ Iridocyclitis, panophthalmitis, vision loss
▪ Cardiac
▫ Myocarditis, cardiac failure
▪ Respiratory
▫ Bronchopneumonia, acute respiratory
distress syndrome
▪ Hematologic
▫ Thrombocytopenia
▪ Other
▫ Hepatitis, splenic rupture
▪ During pregnancy
235
Chapter 97 Spirochetes
▫ Spontaneous abortion, prematurity,
neonatal death
▪ Jarisch–Herxheimer reaction
▫ During treatment with antibiotics →
release of proinflammatory cytokines
triggered by products released from
dead microbes
SIGNS & SYMPTOMS
▪ Characteristics of febrile episodes
▫ Sudden onset of high fever → crisis
phase: chills, ↑ ↑ temperature, ↑ HR, ↑
BP → diaphoresis, ↓ fever, ↓ BP
▫ ↑ mortality during crisis and immediate
aftermath
▪ Constitutional
▫ Sudden onset of high fever and chills;
followed by headache, myalgia,
arthralgia, nausea
▪ Neurologic
▫ Dizziness, delirium, stupor, facial palsy
▪ Cardiac
▫ Prolonged QTc interval.
▪ Respiratory
▫ Nonproductive cough, signs of
respiratory distress
▪ Hematologic
▫ Epistaxis, petechiae, ecchymoses,
blood-tinged sputum
▪ Other
▫ Hepatomegaly, abdominal pain,
photophobia, skin rash
DIAGNOSIS
LAB RESULTS
Visualization of microbe
▪ Blood: thick or thin smears
▫ Giemsa, Wright, or acridine orange stain
▫ Direct or indirect immunofluorescence
▫ Phase contrast or dark field microscopy
▪ PCR, culture, serology
▪ Tissue specimen
▫ Silver stain (e.g. Warthin-Starry,
modified Dieterle)
▫ Immunofluorescence
TREATMENT
MEDICATIONS
▪ Antibiotics
▫ Penicillin G, ceftriaxone, doxycycline
OTHER INTERVENTIONS
Prevention
▪ Avoidance and eradication of vector
236
LEPTOSPIRA
osms.it/leptospira
PATHOLOGY & CAUSES
▪ A spirochete that causes the disease
leptospirosis
▫ AKA Weil's disease, Weil–Vasiliev
disease, swineherd's disease, rice-field
fever, waterborne fever, nanukayami
fever, cane-cutter fever, swamp fever,
mud fever, Stuttgart disease, canicola
fever
▪ Infected mammal excretes microbe in urine
which remains viable in stagnant water
and wet soil → environmental exposure
by humans → microbe usually enters via
non-intact skin, mucous membranes, or
conjunctivae (rarely via contaminated food,
water, or aerosols)
▪ Effects of microbe
▫ Damages blood vessel endothelium →
organ damage
▫ Inhibits the Na+-K+-Cl- cotransporter
activity in the thick ascending limb of
Henle → hypokalemia, hyponatremia
COMPLICATIONS
Electrolyte imbalance, kidney failure
Hepatitis, hepatic hemorrhage
Aseptic meningitis
Pulmonary hemorrhage, acute respiratory
distress syndrome (ARDS)
▪ Uveitis, optic neuritis
▪ Myocarditis
▪ Rhabdomyolysis
▪
▪
▪
▪
▫ Nonproductive cough
▫ Pharyngitis
▫ Myalgias, arthralgias
▫ Conjunctival suffusion (redness without
exudate)
▫ Lymphadenopathy
▫ Jaundice
▫ Uremia, bacteremia, oliguria,
hypokalemia
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Nodular densities, consolidation; may have
a ground-glass appearance
LAB RESULTS
▪ Identification of microbe
▫ PCR, ELISA, microscopic agglutination
▪ Other laboratory studies
▫ Leukocytosis with left shift
▫ Hypokalemia, hyponatremia
▫ ↑ hepatic transaminases, ↑ direct
bilirubin
▫ Urinalysis: proteinuria, pyuria, granular
casts, hematuria, ↑ creatine kinase
▫ CSF: neutrophilic pleocytosis, ↑ protein
OTHER DIAGNOSTICS
▪ History and physical examination
SIGNS & SYMPTOMS
▪ Variable clinical course
▫ Ranges from mild disease that resolves
uneventfully to severe and potentially
fatal
▪ Common symptoms
▫ Abrupt onset of fever, chills
▫ Headache
237
Chapter 97 Spirochetes
OTHER INTERVENTIONS
TREATMENT
MEDICATIONS
▪ Antibiotics
▫ Doxycycline (mild cases), penicillin
(severe cases)
▪ Address complications
▪ Prevention
▫ Avoidance of potential infectious
sources of infection, domestic animal
vaccination
TREPONEMA PALLIDUM (SYPHILIS)
osms.it/syphilis
PATHOLOGY & CAUSES
▪ The spirochete bacterium that causes
syphilis, a localized and systemic disease
▪ Transmission
▫ Sexually: by direct contact with an
infectious lesion (primarily) → enters via
microscopic abrasions
▫ Perinatally: crosses placenta easily →
congenital syphilis
▪ Progresses through stages
▫ Primary: localized sores (chancre)
appear after about three weeks at site of
infection
▫ Secondary: 2–8 weeks after chancre
resolution, hematogenous bacterial
dissemination causes systemic
symptoms
▫ Latent: asymptomatic
▫ Tertiary (late): organ damage develops
10–30 years after initial infection
▪ Neurosyphilis and ocular syphilis can occur
at any stage
▫ Neurosyphilis begins when the microbe
invades the CSF
RISK FACTORS
Unprotected sex
Multiple sexual partners
Biologically male
Biologically-male individuals engaging in
same-sex sexual contact (MSM)
▪ IV drug use
▪ Existing sexually-transmitted disease,
especially HIV
▪
▪
▪
▪
COMPLICATIONS
▪ Cardiovascular: syphilitic aortic aneurysms,
dilated aorta, aortic valve regurgitation;
coronary artery narrowing
▪ Congenital syphilis: hemolytic anemia,
deafness, keratitis, periostitis
▪ Neurosyphilis: dementia, meningitis, brain
or spinal cord ischemia/infarction, seizures,
ischemic stroke
▪ Ocular syphilis: uveitis, vitritis, retinitis,
optic neuropathy, blindness
▪ Otosyphilis: hearing loss, tinnitus
SIGNS & SYMPTOMS
Stages
▪ Presentation will vary according to stage of
disease
▪ Primary
▫ Chancre: painless ulcers at inoculation
site (in contrast to painful lesions seen in
genital herpes or chancroid)
▫ Single or multiple; usually firm, round,
painless
▫ Heals with or without treatment;
treatment prevents progression to
secondary stage
▪ Secondary
▫ May be asymptomatic
▫ Rash: diffuse rough, reddish-brown
maculopapular on extremities, palms
of hands and/or soles of feet, back;
raised, gray-whitish lesions on mucous
membranes
▫ Condylomata lata
238
▫ Myalgia
▫ Fatigue
▫ Lymphadenopathy
▫ Fever
▫ “Moth-eaten” alopecia
▫ Resolves without treatment; treatment
prevents progression to latent and
tertiary stages
▪ Latent
▫ Positive serology; asymptomatic
▪ Tertiary (late)
▫ Gummas: non-cancerous,
granulomatous growths on internal
organs, bones, skin; more common in
HIV-infected individuals
▫ Evidence of organ involvement; charcot
joints, aoritis (due to destruction of vasa
vasorum)
Complications
▪ Neurosyphilis
▫ Meningitis: headache, nausea and
vomiting, stiff neck
▫ Tabes dorsalis: muscle weakness,
locomotor ataxia, ↓ proprioception,
incoordination
▫ General paresis (paralytic dementia)
▫ Facial and limb hypotonia, intention
tremors
▫ Forgetfulness, personality changes
▪ Ocular syphilis
▫ ↓ visual acuity; loss of vision
▫ Argyll Robertson pupil: small pupils that
constrict poorly in response to direct
light
▪ Serum treponemal tests
▫ Fluorescent treponemal antibody
absorption (FTA-ABS)
▫ Treponema pallidum particle
agglutination assay (TPPA)
▫ Syphilis enzyme immunoassays (EIAs)
▪ If neurologic symptoms, lumbar puncture
and CSF examination
▫ ↑ lymphocytes, ↑ protein
▫ CSF-VDRL reactivity
OTHER DIAGNOSTICS
▪ History and physical examination
Figure 97.2 A painless penile chancre, seen
here, is the clinical manifestation of primary
syphilis.
Congenital syphilis
▪ Presents with vesicular/bullous rash,
rhinitis, hepatosplenomegaly, jaundice, and
pseudoparalysis
DIAGNOSIS
LAB RESULTS
▪ Identification of microbe
▪ Serum nontreponemal tests (may be
nonreactive in late neurosyphilis)
▫ Venereal disease research laboratory
(VDRL)
▫ Rapid plasma reagin (RPR)
Figure 97.3 Condylomata lata, seen here,
are the clinical manifestation of secondary
syphilis.
239
Chapter 97 Spirochetes
TREATMENT
MEDICATIONS
▪ Parenteral (IM/IV) penicillin G
▫ Doxycycline or tetracycline (PO);
ceftriaxone (IM, IV) if penicillin allergy
OTHER INTERVENTIONS
▪ Treat partners
▪ Screening during first prenatal visit (VDRL
or RPR)
240
NOTES
NOTES
STAPHYLOCOCCUS
MICROBE OVERVIEW
▪ Staphylococcus: genus of Gram-positive
bacteria responsible for many diseases
▪ Aerobic, facultative anaerobic
▪ Frequent skin colonization: up to half of
population
Genetic material
▪ All staphylococci are catalase-positive
Taxonomy
▪ Staphylo-: cluster; -coccus: berry
Morphology
▪ Organized in grape-like clusters
▪ Gram stain: round, Gram-positive (purple)
bacteria; thick peptidoglycan cell wall
Antibiotic resistance mechanisms
▪ β-lactamase: hydrolysis of β-lactam
antibiotics; some β-lactam antibiotics are
resistant to the enzyme (e.g. oxacillin,
nafcillin, flucloxacillin)
▪ mecA gene → penicillin binding protein
(PBP2a) → reduced affinity for β-lactam
antibiotics; present in methicillin-resistant
strains
▪ vanA gene → modifies cell wall
peptidoglycans → vancomycin cannot bind
to bacteria
STAPHYLOCOCCUS AUREUS
osms.it/staphylococcus-aureus
PATHOLOGY & CAUSES
▪ Staphylococcus aureus: common
bacterium, causes infections in most organ
systems
▪ Aureus: golden colonies in mannitol salt
agar, due to mannitol fermentation
▪ Skin colonization: approx. 30% of
population
▫ Primary colonization site: nostrils
▪ Pathogen transmission: direct contact,
fomites
Virulence factors
▪ Cytolysins: alpha-toxin, Panton–Valentine
leukocidin (PVL); both destroy neutrophils
▫ PVL related to severe skin, lung
infections
▪ Hemolysins: form pores in host cells (e.g.
erythrocytes, macrophages, lymphocytes)
▪ Superantigens: enterotoxins, toxic shock
syndrome toxin-1 (TSST-1)
▫ Superantigens stimulate activation
of excessive amount of T-cells →
increased production of cytokines →
uncontrolled inflammation
▪ Epidermolytic toxins A and B: skin
blistering
▪ Coagulase: activates prothrombin →
coagulation
▪ Bacterial spread: protease degrades
proteins; lipase degrades lipids
▪ Protein A: inactivates immunoglobulins →
phagocytosis evasion
▪ Can form biofilms
▫ Biofilm: adherence of cells to polymer
241
Chapter 98 Staphylococcus
surfaces (e.g. catheters)
▫ Properties allow immune evasion
CAUSES
▪ Common cause of infections and toxinmediated diseases acquired from
community/healthcare environment
▫ Endocarditis, ocular infections,
pneumonia, meningitis, osteomyelitis,
septic arthritis, prosthetic device
infections, catheter-associated
infections
Skin/soft tissue infections
▪ E.g. surgical site infections, abscesses,
impetigo, cellulitis, erysipela, fasciitis,
mastitis
Toxic shock syndrome (TSS)
▪ Mediated by toxic shock syndrome toxin-1;
commonly caused by growth of S. aureus
in vagina/surgical sites → multiple organ
dysfunction
Staphylococcal scalded-skin syndrome
(Ritter disease)
▪ Mediated by epidermolytic toxins A and B
Foodborne illness
▪ Caused by ingestion of S. aureus
endotoxins
SIGNS & SYMPTOMS
Skin/soft tissue infections
▪ Erythema, swelling, warmth, localized
warmth/fever
▫ Staphylococcal scalded-skin syndrome
(Ritter disease): fever, erythema, fluidfilled bullae on the skin → skin loss
Systemic infections
▪ Joint pain, abdominal pain, headache, CVA
tenderness, new onset heart murmur
TSS
▪ Fever, hypotension, rash, coagulopathy,
tissue necrosis (site of infection)
Foodborne illness
▪ Nausea, vomiting, diarrhea, abdominal pain
DIAGNOSIS
LAB RESULTS
▪ Culture-based observation
▫ Clustered golden Gram-positive cocci
▫ Catalase-positive; coagulase-positive
▪ Polymerase chain reaction (PCR)
amplification
▪ May be guided by further studies
depending on site of infection
RISK FACTORS
▪ Immunosuppression, IV drug use, recent
invasive procedure, foreign material in body
(e.g. prosthetics, catheters, sutures), dialysis
COMPLICATIONS
▪ Sepsis, bacteremia, invasive infection
▪ Antibiotic resistance: all strains resistant
to penicillin G; some strains resistant to
methicillin (MRSA) or vancomycin (VRSA);
some strains have intermediate resistance
to vancomycin (VISA)
▫ Resistant strains are common
pathogens in nosocomial infections
Figure 98.1 Staphylococcus takes on a
golden colour when cultured with mannitol
containing media.
242
TREATMENT
MEDICATIONS
Topical antibiotics
▪ Superficial skin infections
Oral/IV antibiotics
▪ Treatment based on the pathogen’s
antibiotic susceptibility
▫ Oxacillin/nafcillin/cefazolin
▫ MRSA: vancomycin/trimethoprimsulfamethoxazole
▫ VRSA: linezolid
SURGERY
▪ Abscess drainage (if applicable)
▪ Foreign material removal in body (if
applicable)
STAPHYLOCOCCUS EPIDERMIDIS
osms.it/staphylococcus-epidermidis
PATHOLOGY & CAUSES
▪ Staphylococcus epidermidis: bacteria
commonly associated with infections of
surgical sites, indwelling catheters, and
prosthetic devices
▪ Part of skin and mucous membrane natural
flora
▪ Does not produce exotoxins
CAUSES
▪ Skin/mucous colonization → barrier rupture
→ infection
▪ Can form biofilms on foreign materials in
body; biofilm properties allow immune
evasion
▪ Polymer surface adhesion → extracellular
matrix production → polysaccharide
intercellular adhesin (PIA) secretion →
multi-layered bacteria
▪ Common S. epidermidis infections:
catheter-associated infection, intravascular
catheter infection, prosthetic joint infection,
endocarditis (frequently associated with
prosthetic valves), surgical site infection
RISK FACTORS
COMPLICATIONS
▪ Sepsis, bacteremia
▪ Neonates: pneumonia, urinary tract
infections, meningitis, enterocolitis,
omphalitis
SIGNS & SYMPTOMS
Local
▪ Pain, tenderness, swelling, warmth,
erythema, drainage at incisional site
Systemic
▪ Fever, hypotension, leukocytosis
DIAGNOSIS
LAB RESULTS
Culture-based observation
▪ Blood, urine, synovial fluid, surgical site
▫ Clustered Gram-positive cocci
▫ Catalase-positive; coagulase-negative;
novobiocin-sensitive
PCR amplification
▪ Immunosuppression, neonates, recent
invasive procedure, foreign material in the
body (e.g. prosthetics, catheters), dialysis
243
Chapter 98 Staphylococcus
TREATMENT
MEDICATIONS
SURGERY
▪ Remove foreign material from body (if
applicable)
Oral antibiotics
▪ Empiric treatment: vancomycin
▪ If proven methicillin sensitivity: oxacillin/
nafcillin; may be combined with rifampicin,
gentamicin
▪ Fusidic acid for skin infections (if available)
STAPHYLOCOCCUS
SAPROPHYTICUS
osms.it/staphylococcus-saprophyticus
PATHOLOGY & CAUSES
SIGNS & SYMPTOMS
▪ Staphylococcus saprophyticus: bacteria
that commonly produces urinary tract
infections (UTI) in young, biologicallyfemale individuals
▪ Frequently part of vagina’s natural flora
▪ Tropism for urinary tract: surface fibrillar
protein (Ssp) and hemagglutinin/adhesin
allow pathogen’s adherence to uroepithelial
cells
▪ Does not produce exotoxins
▪ Etiologic agent: community-acquired UTIs
(occasionally)
▪ Usually presents as symptomatic cystitis
(bladder inflammation)
▪ Dysuria (painful urination), urinary urgency,
increased urinary frequency, suprapubic
pain, occasional hematuria
RISK FACTORS
DIAGNOSIS
LAB RESULTS
Urinalysis
▪ Presence of leukocytes, erythrocytes,
bacteria, negative urine nitrate
Culture-based observation
▪ Clustered Gram-positive cocci
▪ Catalase-positive; coagulase-negative;
novobiocin-resistant
▪ Biologically-female (shorter urethra), recent
sexual activity (“honeymoon cystitis”),
diabetes, immunosuppression, indwelling
urinary catheter
Quantitative PCR
COMPLICATIONS
MEDICATIONS
▪ Pyelonephritis
TREATMENT
▪ Oral antibiotics
▪ Symptomatic therapy (e.g.
phenazopyridine)
244
AfraTafreeh.com exclusive
NOTES
NOTES
STREPTOCOCCUS
MICROBE OVERVIEW
Morphology
▪ Spherical, Gram-positive bacteria; appear in
chains/pairs; catalase, coagulase negative
▪ Produce extracellular substances
(e.g. cytolysins, enzymes) → enhance
pathogenicity
STREPTOCOCCUS AGALACTIAE
(GROUP B STREP)
osms.it/streptococcus-agalactiae
PATHOLOGY & CAUSES
▪ AKA Group B Streptococcus (GBS)
▪ Encapsulated, facultative anaerobe
▪ Colonizes human genital, gastrointestinal
(GI) tracts; upper respiratory tracts of young
infants
▪ Beta-hemolytic
▫ Blood agar plates, hemolysins degrade
lipid membranes → colonies surrounded
by narrow zone of hemolyzed cells →
complete (beta-)hemolysis
Virulence factors
▪ Complex capsular polysaccharides
▫ Inhibit complement deposition on
microbe surface components
▪ Hypervirulent GBS adhesin (HvgA)
▫ ↑ ability to invade blood-brain barrier
▪ Cluster of virulence responder/sensor
(CovR/S) mutation
▫ Accelerate failure of amniotic barrier
→ ↑ ability to penetrate chorioamniotic
membranes
▪ Pilins
▫ Act as adhesins → ↑ ability to invade
central nervous system, form biofilm
▪ Direct cytotoxicity to host phagocytes
Common infectious agent
▪ Adults (nonpregnant)
▫ Broad spectrum of infections
▪ Pregnant individuals
▫ Chorioamnionitis
▪ Neonates
▫ GBS infection, sepsis
RISK FACTORS
▪ Adults (nonpregnant)
▫ Chronic disease (e.g. diabetes, liver
disease, malignancy; > age 65, esp.
residents of nursing homes)
▪ Pregnancy
▪ Neonates
▫ Ascending infection from mother (e.g.
rupture of membranes, chorioamnionitis)
▪ Hospitalization
▫ Nosocomial/hospital-acquired infections
245
Chapter 99 Streptococcus
COMPLICATIONS
▪ Cystitis, pyelonephritis, urethritis,
prostatitis; osteomyelitis, septic arthritis;
endocarditis; meningitis; pneumonia; sepsis;
toxic shock-like syndrome
▪ Neonates
▫ Preterm birth, bacteremia, sepsis,
pneumonia, meningitis, neonatal
mortality
SIGNS & SYMPTOMS
▪ Fever, chills; malaise; cough
▪ Local tissue infection
▫ Red, warm, swollen, presence of
drainage
▪ Hippurate hydrolysis test
▫ Detections hippurate hydrolysis by GBS
OTHER DIAGNOSTICS
▪ Clinical history, physical examination
TREATMENT
MEDICATIONS
▪ Antibiotics (e.g. penicillin G, ampicillin)
OTHER INTERVENTIONS
▪ Prenatal screening
DIAGNOSIS
LAB RESULTS
Identification of microbe
▪ E.g. blood, cerebrospinal fluid
▪ Gram stain, characteristic morphology
▪ Culture
▫ Beta-hemolysis on blood agar
▪ CAMP test
▫ Identifies presence of CAMP factor
▪ Latex agglutination tests
▫ Detects antibodies produced in
response to GBS
Figure 99.1 The three classes of
streptococcus cultured on a blood agar plate.
Alpha (left) shows partial hemolysis, beta
(centre) shows complete hemolysis and
gamma (right) shows no hemolysis.
STREPTOCOCCUS PNEUMONIAE
osms.it/streptococcus-pneumoniae
PATHOLOGY & CAUSES
▪ Causative agent for numerous clinical
syndromes in children, older adults
▪ Alpha-hemolytic, lancet-shaped diplococci
▪ Lysis by bile (deoxycholate), optochin
sensitive
▪ Fastidious; prefers 5% carbon dioxide
▪ Pyogenic
▪ Virulence factors
▫ Resistance to phagocytosis (conferred
by 92 polysaccharide serotypes)
▫ Adherence proteins
▫ Biofilm formation
▫ Pneumolysin toxin
▪ Asymptomatic colonization → direct spread
from site of colonization,hematogenous
spread → clinical syndromes
246
▪ Typical infections caused by S. pneumoniae
range from mucosal to invasive diseases
▫ Meningitis
▫ Otitis media
▫ Pneumococcal community-acquired
pneumonia
▫ Sinusitis
RISK FACTORS
▪ Age (< 2, ≥ 65 years)
▪ Underlying disease (e.g. liver, kidney, heart,
lung, diabetes, malignancies)
▪ Crowded conditions (e.g. daycare centers,
military camps, prisons)
▪ Immunodeficiency (e.g. HIV, genetic
immune defects, solid organ/bone
transplant)
▪ Smoking, alcohol abuse
COMPLICATIONS
▪ Pneumococcal endocarditis, empyema,
bacteremia, sepsis
SIGNS & SYMPTOMS
▪ Common clinical presentation
▫ Fever, altered mental status, malaise
▪ Typical findings related to clinical syndrome
▫ Meningitis: headache, neck stiffness
▫ Otitis media: ↓ tympanic membrane
mobility/bulging membrane, otorrhea,
pain
▫ Pneumonia: cough, bronchial breath
sounds, rales
▫ Sinusitis: purulent rhinitis, mucous
membrane edema, headache
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Infiltration, consolidation (pneumonia)
LAB RESULTS
Identification of organism
▪ Gram-positive diplococci, positive culture,
polymerase chain reaction (PCR)
▪ Urine antigen analysis (bacteremia)
OTHER DIAGNOSTICS
▪ Clinical history, physical examination
TREATMENT
MEDICATIONS
▪ Antibiotics
▫ Pneumonia: beta-lactam antibiotic
▫ Otitis media: amoxicillin-clavulanate
(children)
▫ Sinusitis: amoxicillin (amoxicillin–
clavulanic acid may be preferable)
OTHER INTERVENTIONS
Prevention
▪ Pneumococcal vaccine
247
Chapter 99 Streptococcus
STREPTOCOCCUS PYOGENES
(GROUP A STREP)
osms.it/streptococcus-pyogenes
PATHOLOGY & CAUSES
▪ AKA Group A Streptococcus (GAS)
▪ Colonizes human skin, mucous membranes
▪ Cell-wall structure
▫ Peptidoglycan backbone + lipoteichoic
acid components → structural stability
▪ Beta-hemolytic
▫ Blood agar plates, hemolysins degrade
lipid membranes → colonies surrounded
by clear zone of hemolyzed cells →
complete (beta-) hemolysis
▪ Primarily infects skin, soft tissue
Virulence factors
▪ Vary with specific strain
▪ M proteins
▫ Protect microbe from humoral
immune surveillance, phagocytosis by
polymorphonuclear leukocytes
▪ Binding proteins
▫ Bind to IgG, IgM, IgA → may interfere
with complement activation
▫ Protein F: binds to fibronectin → ↑
adherence to epithelial surfaces
▪ Cytolysins
▫ Streptolysins: bind to cholesterol on
eukaryotic cell membranes → cell lysis
▫ Hyaluronidase: hydrolyzes hyaluronic
acid → facilitates infection spread
▫ Streptokinase: proteolytically converts
bound plasminogen to active plasmin
→ cleavage of fibrin; medically useful as
clot-busting drug
▫ Nicotinamide adenine dinucleotidase
(NADase): exact function unclear; likely
↑ invasiveness
▫ Deoxyribonuclease: promotes
production of anti-deoxyribonuclease
(DNase) antibody following pharyngeal/
skin infections
▪ Pyrogenic exotoxins (type A, B, C)
▫ Induce fever, act as superantigens
→ T-cell proliferation → ↑ cytokine
production → promotes shock
▪ Streptococcal inhibitor of complement (SIC)
▫ Inactivates complement membrane
attack complex
▪ Opacity factor (OF)
▫ Lipoprotein lipase
Causative agent in several disorders
▪ Pyogenic diseases
▫ Pharyngitis, cellulitis (abscess formation
in dermis, subcutaneous fat layers),
necrotizing fasciitis (progressive
destruction of deep soft tissue),
impetigo
▪ Toxigenic disease
▫ Scarlet fever, toxic shock syndrome,
GAS endometritis (puerperal sepsis)
▪ Immunologic disease
▫ Rheumatic fever (antibodies against
streptococcal cell cross-react with
cardiac tissue); poststreptococcal
glomerulonephritis (immune complexes
deposited in glomeruli)
RISK FACTORS
▪ Susceptible host + encounter with
streptococcus expressing specific virulence
factors
COMPLICATIONS
▪ Local spread (e.g. otitis media, sinusitis,
mastoiditis); tissue destruction; valvular,
renal disease; sepsis, shock, multiorgan
failure; disseminated intravascular
coagulation; pediatric autoimmune
neuropsychiatric disorder associated with
group A streptococci (PANDAS)
248
DIAGNOSIS
SIGNS & SYMPTOMS
▪ Pharyngitis
▫ Acute onset of sore throat, fever,
pharyngeal edema, patchy tonsillar
exudates
▪ Cellulitis
▫ Erythema, edema, abscess formation
▪ Impetigo
▫ Papules, vesicles, pustules surrounded
by erythema pustules → breaks down,
forms crusts
▪ Scarlet fever
▫ Erythematous rash
▪ Toxic shock syndrome
▫ Shock, multiorgan failure
▪ GAS endometritis
▫ Postpartum fever, uterine tenderness
LAB RESULTS
Identification of microbe
▪ Gram positive cocci
▪ Positive culture
▪ Blood studies
▫ Rapid antigen detection test (RADT) for
GAS
OTHER DIAGNOSTICS
▪ Clinical history, physical examination
TREATMENT
MEDICATIONS
▪ Antibiotics (e.g. penicillin G, clindamycin)
SURGERY
▪ Surgical debridement
STREPTOCOCCUS VIRIDANS
osms.it/streptococcus-viridans
infections (e.g. abdominal, central
nervous system, lung, skin, soft tissue,
sepsis)
▫ Abscess formation
▫ Viridans streptococcal shock syndrome
PATHOLOGY & CAUSES
▪ Heterogeneous collection of alpha/
nonhemolytic streptococci, cause variety of
diseases
▪ Some species produce greenish color on
blood agar plates
▪ Not bile soluble, optochin resistant
▪ Approx. 30 species classified into six
groups
▪ Part of microbiome of oropharynx, GI,
genitourinary tract
▪ May be invasive, produce variety of
diseases
▫ Dental caries, periodontal disease,
maxillofacial infections, exudative
pharyngitis, infective endocarditis
▫ Invades circulation → systemic
RISK FACTORS
Immunocompromised state
Periodontal disease
More common in children than adults
Comorbidities (e.g. mucositis, cystic fibrosis,
malignancies)
▪ Altered microbiome
▪
▪
▪
▪
SIGNS & SYMPTOMS
▪ Clinical presentation varies widely
depending on infection
249
Chapter 99 Streptococcus
DIAGNOSIS
LAB RESULTS
Identification of organism
▪ Gram-positive cocci, positive culture
OTHER DIAGNOSTICS
▪ Clinical history, physical examination
TREATMENT
MEDICATIONS
▪ Antibiotics (depending on sensitivity,
resistance)
▫ Penicillin + aminoglycoside; broadspectrum cephalosporin, vancomycin
SURGERY
▪ Abscess debridement/drainage
250
NOTES
NOTES
SYSTEMIC MYCOSES
GENERALLY, WHAT ARE THEY?
PATHOLOGY & CAUSES
DIAGNOSIS
▪ Fungal infections in internal organs (esp.
lungs)
DIAGNOSTIC IMAGING
CAUSES
LAB RESULTS
▪ Dimorphic species of fungi
▪ Transmitted by spore inhalation;
lymphohematogenous dissemination
SIGNS & SYMPTOMS
▪ X-ray, CT scan, MRI
▪ Culture-based observation, direct
microscopy, serologic tests, lab tests (e.g.
abnormal blood exams)
TREATMENT
MEDICATIONS
▪ Cough, chest pain, fever
▪ Antifungal agents
BLASTOMYCES SPP.
osms.it/blastomyces
PATHOLOGY & CAUSES
▪ Blastomycosis
▫ Systemic fungal infection caused by
Blastomyces dermatitidis, B. gilchristii;
usually manifests as chronic pneumonia
▪ Incubation period: 3–6 weeks
▪ Spore inhalation → conversion to yeast in
lungs → phagocytosis by macrophages →
acute suppurative inflammation
▪ Immune response: mainly cellular, mediated
by T-lymphocytes, macrophages
▪ Common sites of infection: primarily lungs
(90%); skin, bones, genitourinary tract,
central nervous system (CNS)
Blastomyces spp.
▪ Size: 8–15μm
▪ Broad-based budding (wide connection
between two cells before splitting apart
during reproduction)
▪ Thermal dimorphism
▫ Mold form (< 37°C/98.6°F): produces
spores
▫ Yeast form (37°C/98.6°F): multinucleate;
antiphagocytic (e.g. thick cell wall)
▪ Virulence
▫ Thick cell wall: resistance to
phagocytosis
▫ BAD-1: cell surface glycoprotein;
adhesin
▫ Binds yeast to extracellular matrix,
macrophages
251
Chapter 100 Systemic Mycoses
▫ Blocks production of tumor necrosis
factor (TNF) alpha (proinflammatory
cytokine)
Clinical syndromes
▪ Pulmonary blastomycosis
▫ Pneumonia, mostly chronic
▫ Frequently affects upper lobes
▪ Primary cutaneous blastomycosis
▫ Ulcerative/verrucous skin lesions
▪ Disseminated blastomycosis
▫ Osteomyelitis; prostatitis, epididymoorchitis (inflammation of epididymis/
testicles); meningitis; intracranial
abscesses
RISK FACTORS
Outdoor occupations (e.g. farming)
Recreational exposure to soil
Immunosuppression
High prevalence in North America
Recent travel to endemic areas (e.g. Ohio,
Mississippi river valleys)
▪ Comorbid conditions
▪
▪
▪
▪
▪
COMPLICATIONS
LAB RESULTS
Culture-based observation
▪ tissue, sputum, body fluids
▫ Sabouraud dextrose agar without
cycloheximide
▫ Confirmation requires conversion (mold
→ yeast) at 37°C/98.6°F
Direct microscopic examination
▪ Periodic acid–Schiff stain
▪ Differentiation: size, yeast morphology
Lab tests
▪ Anemia, leukocytosis, ↑ erythrocyte
sedimentation rate
Tissue biopsy
▪ Pyogranulomatous response
▪ Skin
▫ Epithelial hyperplasia, intraepidermal
abscesses, multinucleated cells
▪ Fungus observation
Serologic tests
▪ Polymerase chain reaction (PCR)
▫ Antigen detection assays
▪ Acute respiratory distress, multiorgan
disease, chronicity
SIGNS & SYMPTOMS
▪ Cough, fever, weight loss, sputum
production, dyspnea, night sweats, chills,
hemoptysis, arthralgia, soft tissue swelling
▪ Verrucous skin lesions with irregular
borders, ulcerative skin lesions
DIAGNOSIS
TREATMENT
MEDICATIONS
▪ Antifungal treatment
▪ Amphotericin B; followed by azole
▫ Liposomal amphotericin B: CNS
infections
SURGERY
▪ Resection of abscesses, devitalized bone,
empyemas, pericardial effusion
DIAGNOSTIC IMAGING
X-ray
▪ Pneumonia
▫ Lobar consolidation, alveolar infiltrates,
fibronodular infiltrates, cavitation,
nodules, pleural effusion
▪ Osteomyelitis
▫ Well-defined, osteolytic bone lesions
252
COCCIDIOIDES SPP.
osms.it/coccidioides
▫ Metalloproteinase: inhibits phagocytosis
▫ Alteration of pulmonary surfactant
proteins
PATHOLOGY & CAUSES
▪ Coccidioidomycosis
▫ Systemic fungal infection caused by
Coccidioides immitis, C. posadasii;
usually manifests as acute pneumonia
▪ AKA San Joaquin Valley Fever/“desert
rheumatism” (associated with arthralgia)
▪ Arthroconidium inhalation → conversion to
spherules → activation of T-lymphocytes →
production of cytokines → inflammation →
acute pneumonia
▫ In infected tissue, spherules grow,
septate → release endospores →
infection spreads
▪ Immune response
▫ Mainly mediated by Th1 cells
▫ Interleukin 17, TNF alpha, interferongamma
▪ Incubation period: 1–4 weeks
▪ Common sites of infection: lungs, skin,
bones, CNS
▪ High prevalence areas: arid, dry regions in
U.S. (e.g. California, Southwest), Mexico,
Central America, South America
Coccidioides spp.
▪ Size: 20–70μm
▪ Dimorphism
▫ Mold form: found in soil
▫ Yeast form: parasitic
▪ Produces arthroconidia (barrel-shaped,
multinucleated spores)
▫ Production stimulated by human sex
hormones
▫ Arthroconidia convert to spherules
(2–5μm; in infected tissues)
▪ Infectious particles: arthroconidia
▪ Virulence
▫ Enzyme with elastase activity: ↑
infection, inflammation
▫ Cell surface glycoprotein with adhesin
activity
Clinical syndromes
▪ Acute pneumonia
▪ Dermatologic lesions
▫ Wart-like lesions on face
▫ Erythema nodosum/multiforme
▪ Osteomyelitis
▪ Meningitis
RISK FACTORS
▪ Outdoor occupations; recreational
exposure to soil (e.g. gardening, camping);
immunosuppression; recent travel to
endemic areas; comorbid conditions
COMPLICATIONS
▪ Adult respiratory distress syndrome; fatal
multilobar pneumonia; pyopneumothorax;
meningitis; chronicity
SIGNS & SYMPTOMS
▪
▪
▪
▪
▪
Mostly mild/asymptomatic
Non-specific: fever, malaise
Cough, pleuritic pain, hemoptysis, arthralgia
Erythema nodosum/multiforme
Wart-like lesions on face (e.g. nasolabial
folds)
DIAGNOSIS
DIAGNOSTIC IMAGING
X-ray
▪ Pneumonia
▫ Parenchymal infiltrates, thin-walled
cavities
▪ Osteomyelitis
▫ Osteolytic bone lesions
253
Chapter 100 Systemic Mycoses
LAB RESULTS
TREATMENT
▪ Culture-based observation
Serologic tests
▪ IgM, IgG antibody detection (e.g. enzyme
immunoassays)
▪ Antigen detection
Direct microscopic observation
▪ Spherules in sputum, blood, body fluid
samples
Lab tests
▪ ↑ erythrocyte sedimentation rate
▪ Eosinophilia (mostly with dissemination)
MEDICATIONS
High risk of dissemination
▪ E.g. immunosuppression, pregnancy
▫ azoles
Severe
▪ Amphotericin B
SURGERY
▪ Debridement of abscesses, devitalized
bone, pyopneumothorax
Extrathoracic tissue biopsy
▪ Essential for diagnosis of Coccidioides
dissemination
▪ Pyogranulomatous inflammation
▪ Presence of spherules
Spherulin skin test
▪ Positive after resolution; not available in
U.S.
Figure 100.1 Numerous spores in the lungs
of an individual with coccidioidomycosis.
254
HISTOPLASMA CAPSULATUM
osms.it/histoplasma-capsulatum
PATHOLOGY & CAUSES
▪ Histoplasmosis
▫ Systemic fungal infection caused
by Histoplasma capsulatum; usually
manifests as acute pneumonia
▪ Most frequent systemic mycoses in U.S.
▪ Microconidia inhalation → conversion to
yeast form → macrophage phagocytosis →
inflammation → pneumonia
▪ Immune response: mainly cellular, mediated
by T-lymphocytes, macrophages, TNF
alpha, interferon-gamma
▪ Infection sites: primarily lungs; may
disseminate to other organs
Histoplasma capsulatum
▪ Size: 2–3μm x 3–4μm
▪ Thermal dimorphism
▫ Mold form (< 35°C/95°F); produces
microconidia (spores, 2–5μm)
▫ Yeast form (37°C/98.6°F)
▪ Infectious particles: microconidia
▪ Bird, bat fecal material promotes growth
Pulmonary histoplasmosis clinical
syndromes
▪ Pneumonia: acute (diffuse/localized);
chronic
▪ Broncholithiasis
Disseminated histoplasmosis clinical
syndromes
▪ Progressive disseminated histoplasmosis:
excessive reticuloendothelial infection
▪ Adrenal perivasculitis (common)
▪ Endocarditis
▪ Mediastinal granuloma
▪ Mediastinitis
▪ Meningitis
▪ Ocular histoplasmosis (e.g. retinal lesions)
▪ Lesions: intestinal (e.g. ulcers, polyps), skin
(e.g. dermatitis, papules)
RISK FACTORS
▪ Outdoor occupations (e.g. construction,
excavation)
▪ Outdoor activities (e.g. camping)
▪ Immunosuppression
▪ High prevalence regions: U.S. (Ohio,
Mississippi river valleys), Mexico, Central
America, South America
▪ Recent travel to endemic areas
▪ Comorbid conditions
▪ Extremes of age
COMPLICATIONS
▪ Fatal acute diffuse pneumonia, mediastinal
granuloma, mediastinitis, chylothorax,
pleural effusion
SIGNS & SYMPTOMS
▪ Mostly asymptomatic
Acute pulmonary histoplasmosis
▪ Systemic
▫ Fever, headaches, fatigue
▪ Chest pain (pleuritic/substernal), dry cough,
myalgia, arthralgia, erythema nodosum/
multiforme
Chronic pulmonary histoplasmosis
▪ Systemic
▫ Fever, fatigue, night sweats, weight loss
▪ Productive cough, hemoptysis, dyspnea
▪ Consolidation: dullness to percussion,
crackles
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Hilar/mediastinal lymphadenopathy, patchy/
nodular pulmonary infiltrates, occasional
cavitation
255
Chapter 100 Systemic Mycoses
LAB RESULTS
▪ Culture-based observation
▪ Direct microscopic observation
Serologic tests
▪ Antibody detection (e.g. immunodiffusion,
complement fixation assays)
▪ Antigen detection in urine, sputum, body
fluids (e.g. enzyme immunoassays)
Lab tests
▪ Anemia, ↑ erythrocyte sedimentation rate,
progressive disseminated histoplasmosis
(e.g. pancytopenia)
Tissue biopsy
▪ Granulomas, lymphohistiocytic aggregates,
mononuclear cell infiltrates, fungi
visualization
TREATMENT
Figure 100.2 Grocott methenamine silver
stain highlights spores in the lung tissue
of an immunocompromised individual with
histoplasmosis.
MEDICATIONS
▪ Progressive disseminated/prolonged/severe
pulmonary histoplasmosis
▫ Corticosteroids: ↓ inflammation
▫ Antifungal treatment: amphotericin B,
azoles
PARACOCCIDIOIDES BRASILIENSIS
osms.it/paracoccidioides-brasilienses
PATHOLOGY & CAUSES
▪ Paracoccidioidomycosis
▫ Systemic fungal infection caused by
Paracoccidioides brasiliensis, P. lutzii;
usually manifests as chronic lung
disease
▪ AKA South American blastomycosis
▪ Spore inhalation → conversion to
yeast form in lungs → phagocytosis
by macrophages → inflammation →
pneumonia
▪ Immune response: mostly cell-mediated
▪ Common sites of infection: mainly lungs;
oral mucosa, skin, adrenal glands, CNS
Paracoccidioides spp.
▪ Size: 4–40μm
▪ Thermal dimorphism
▫ Mold form (22–26°C/71.6–78.8°F):
present in soil
▫ Yeast form (37°C/98.6°F): “pilot’s wheel”
appearance
▪ Stimulated by sex hormones
Clinical syndromes
▪ Acute/subacute paracoccidioidomycosis
( juvenile)
▫ Hepatosplenomegaly,
256
lymphadenopathy, skin lesions,
pulmonary manifestations (rare)
▪ Chronic paracoccidioidomycosis (adult)
▫ Progressive pulmonary fibrosis (esp.
lower lobes), ulcers (mouth, larynx);
adrenal involvement
RISK FACTORS
▪ High prevalence regions: Central, South
America (80% in Brazil)
▪ Outdoor occupations
▪ Outdoor activities: contact with soil
▪ More common in individuals who are
biologically male
▪ Immunosuppression
COMPLICATIONS
▪ Bone marrow, adrenal dysfunction; chronic
respiratory failure
SIGNS & SYMPTOMS
▪ Generally asymptomatic (95%)
▪ Non-specific symptoms: fever, weight loss
▪ Cough, dyspnea, hepatosplenomegaly,
lymphadenopathies, odynophagia,
sialorrhea, skin lesions (ulcers, nodules)
▪ Compressive manifestations: jaundice (bile
duct obstruction)
Figure 100.3 A child with numerous lesions
on the face caused by Coccidioidomycosis
brasiliensis.
Serologic tests
▪ Detection of antibodies through
immunodiffusion
TREATMENT
MEDICATIONS
▪ Antifungal treatment: azoles
▪ Trimethoprim-sulfamethoxazole
DIAGNOSIS
DIAGNOSTIC IMAGING
X-ray/CT scan
▪ Acute/subacute paracoccidioidomycosis
▫ Lymph node enlargement
▪ Chronic paracoccidioidomycosis
▫ Pulmonary disease: alveolar/interstitial
infiltrates, consolidation, masses/
nodules, cavitation
▫ CNS disease: ring enhancing lesions
▫ Articular disease: effusion, erosions,
space narrowing
LAB RESULTS
▪ Direct microscopic observation
▪ Culture-based observation
257
NOTES
NOTES
TOGAVIRUSES
MICROBE OVERVIEW
▪ Pathogenic viruses in Togaviridae family
▪ Capsid symmetry: icosahedral
▪ RNA structure: linear, positive polarity
EASTERN EQUINE ENCEPHALITIS
VIRUS (EEEV)
osms.it/eastern-equine-encephalitis
PATHOLOGY & CAUSES
▪ Highly pathogenic; causes central nervous
system illness in humans, horses (equines)
▪ Genus: Alphavirus
▪ Spherical, approx. 69nm diameter
(including glycoprotein spikes)
▪ Enveloped, single-stranded, positive-sense
RNA genome
▪ Glycoproteins associated with
neurovirulence, cellular apoptosis
▪ Potential bioterrorism agent use (aerosol
route)
▪ Range
▫ Atlantic, Gulf-coast states in eastern
USA
▪ Four lineages
▫ Group I: endemic in North America,
Caribbean (causes most human disease)
▫ Groups IIA, IIB, III: primarily cause
equine illness in Central, South America
▪ Viral life-cycle: wild birds, Culiseta
melanura mosquito (enzootic vector)
▫ C. melanura rarely bites humans
▫ Human transmission requires other
mosquito species (e.g. Aedes,
Coquillettidia, Culex) to bridge between
infected birds, humans
▫ Infected mosquito bite → 4–10 day
incubation period → prodromal period
→ neurological symptom development
occurs rarely
RISK FACTORS
▪ Rural residence; living in/visiting woodland
habitats, swampy areas
▪ Outdoor occupation/recreational activity
COMPLICATIONS
▪ Encephalitis; cerebral edema; coma;
residual brain damage (mild–severe, esp.
young children); death (some)
SIGNS & SYMPTOMS
▪ May be asymptomatic
▪ Prodromal period: high fever, headache,
nausea, vomiting
▪ Neurologic presentation: cranial nerve
palsy, seizure, stupor → coma
▪ Infants: fever, bulging fontanel, generalized
flaccid/spastic paralysis
258
DIAGNOSIS
TREATMENT
DIAGNOSTIC IMAGING
▪ No specific treatment
MRI
▪ Focal lesions in basal ganglia, thalamus,
brainstem
MEDICATIONS
LAB RESULTS
▪ Leukocytosis; left shift
▪ Hyponatremia
▪ Serology
▫ IgM antibody presence
▪ Cerebrospinal fluid (CSF) examination
▪ Lymphocytic pleocytosis, ↑ neutrophils;
↑ protein; IgM antibodies (assay); virus
isolation
▪ Supportive: anticonvulsants,
corticosteroids (↓ inflammation)
OTHER INTERVENTIONS
▪ Supportive: IV fluid, respiratory support,
monitor intracranial pressure
▪ Prevention
▫ Insect repellent (DEET, picaridin,
IR3535, oil of lemon eucalyptus)
▫ Protective clothing
▫ Vector control
OTHER DIAGNOSTICS
Electroencephalography (EEG)
▪ Generalized slowing; disorganized pattern
RUBELLA VIRUS
osms.it/rubella-virus
PATHOLOGY & CAUSES
▪ Highly communicable virus → German
measles
▪ Enveloped, positive-sense, single-stranded
RNA virus
▪ Family: Togaviridae
▪ Genus: Rubivirus
▪ Three structural proteins
▫ C: capsid protein surrounding virion
RNA
▫ E1, E2: glycosylated proteins forming
transmembrane antigenic sites
▪ Humans are the only natural hosts
▪ Transmission: droplet inhalation/direct
contact with infectious nasopharyngeal
secretion
▪ Viral contact → 12–23 day incubation
→ nasopharyngeal cell, regional lymph
node viral replication → viremia →
maculopapular rash eruption → rash
resolution (approx. two days)
▫ Contagious via virus shedding before,
after rash appears
▫ ↑ contagiousness during rash eruption
▪ Spreads transplacentally
RISK FACTORS
▪ Unvaccinated
▪ Travel (especially abroad)
▪ Contact with febrile rash individuals
COMPLICATIONS
▪ Thrombocytopenic purpura
▪ Encephalitis (rare)
▪ If infected during pregnancy: congenital
rubella syndrome (CRS)
259
Chapter 101 Togaviruses
▫ ↑ risk of miscarriage, fetal death,
stillbirth
▫ CRS: A ToRCHeS (see mnemonic)
infection; ↑ first trimester risk;
extramedullary hematopoiesis
(“blueberry muffin” rash), cataract, heart
defect, hearing impairment, intellectual
disability
MNEMONIC: ToRCHeS
Perinatal infections passed
from mother to child
Toxoplasmosis, toxoplasma
gondii
Other infections
Rubella
Cytomegalovirus
Herpes Simplex virus-2/
neonatal herpes simplex
SIGNS & SYMPTOMS
▫ Reverse transcription-PCR (rubella virus
RNA performed on amniotic fluid)
OTHER DIAGNOSTICS
▪ Clinical diagnosis
▪ High suspicion index
▫ Febrile rash, unvaccinated status
TREATMENT
▪ No specific antiviral therapy
OTHER INTERVENTIONS
▪ Infection control measures
▫ Prompt isolation for seven days after
rash development
▪ Vaccine: live-attenuated measles-mumpsrubella (MMR)/measles-mumps-rubellavaricella (MMRV)
▫ First dose: 12–15 months old
▫ Second dose: 4–6 years old
▪ Maculopapular rash
▫ Pink/light red macules: coalesce to form
evenly-colored desquamating rash
▫ Initially: face → generalized rash within
24 hours
▫ Duration: three days
▪ Lymphadenopathy; primarily posterior
auricular/suboccipital lymph
▪ Low-grade fever
▪ Mild nonexudative conjunctivitis
▪ Forchheimer spots on soft palate
▪ Arthralgias
▪ Orchitis
▪ Asymptomatic (half of cases)
DIAGNOSIS
LAB RESULTS
▪ Polymerase chain reaction (PCR) testing/
molecular typing
▫ Throat, nasal, urine specimens
▪ Serologic testing
▫ Enzyme immunoassay (EIA) detects
rubella-specific IgM antibodies
▪ Pregnancy
Figure 101.1 A child with rubella showing a
characteristic maculopapular, erythematous
rash.
260
WESTERN EQUINE ENCEPHALITIS
VIRUS (WEE)
osms.it/western-equine-encephalitis
PATHOLOGY & CAUSES
▪ Causes central nervous system illness in
humans, horses (equines)
▪ Genus Alphavirus
▪ Spherical, approx. 69nm diameter
(including glycoprotein spikes)
▪ Enveloped, single-stranded, positive-sense
RNA genome
▪ Contain glycoproteins associated with
neurovirulence, cellular apoptosis
▪ Range: most commonly US states,
Canadian provinces west of Mississippi
River
▪ Virus life-cycle: wild birds, other
vertebrates, Culex tarsalis mosquito
(enzootic vector)
▫ Culex tarsalis (another human vector)
▪ Potential bioterrorism agent use (aerosol
route)
▪ Infected mosquito bite → 2–10 day
incubation period → sudden onset of
severe headache, fever/chills, dizziness,
chills, myalgias, malaise, tremor, irritability,
photophobia, neck stiffness → rapid
neurological manifestation development →
recovery
▫ Most adults: no residual neurological
effects
▫ Infants, children: ↑ long-term neurologic
sequelae risk
RISK FACTORS
▪ Most cases June–September
▪ Bimodal age pattern: < one year; ↑ risk in
elderly
▪ Biologically-female
▪ Rural residence
▪ Outdoor occupation/recreational activity
COMPLICATIONS
▪ Encephalitis, coma, respiratory failure,
death
▪ Infants: intellectual disability, cerebellar
damage, spastic paralysis, developmental
delay
SIGNS & SYMPTOMS
▪ Neurological manifestations
▫ Generalized weakness; somnolence;
hand, tongue, lip tremor; cranial nerve
palsy; motor weakness; ↓ deep tendon
reflexes
▪ Infants: poor feeding, fussiness, fever,
vomiting, tense/bulging fontanelle
DIAGNOSIS
LAB RESULTS
▪ Serology
▫ Enzyme-linked immunosorbent assay
(ELISA): IgM antibodies
▫ Hemagglutination-inhibition,
neutralizing antibody presence
▪ CSF
▫ ELISA: IgM antibodies
▫ Lymphocytic pleocytosis
▫ ↑ protein
TREATMENT
▪ No specific treatment
MEDICATIONS
▪ Supportive: anticonvulsants, corticosteroids
261
Chapter 101 Togaviruses
OTHER INTERVENTIONS
Prevention
▪ Insect repellent (DEET, picaridin, IR3535, oil
of lemon eucalyptus)
▪ Protective clothing
▪ Vector control
262
NOTES
NOTES
TREMATODES (FLATWORMS)
GENERALLY, WHAT ARE THEY?
PATHOLOGY & CAUSES
▪ AKA flukes; parasitic flatworm; infects
internal organs
▪ Phylum: Platyhelminthes
Characteristics
▪ Adult morphology: flattened oval/elongated
▪ Structures
▫ Tegmentum (outer body covering)
▫ Ventral, oral suckers
▫ Pharynx → esophagus → caeca
▫ Testes, uterus/ovary (hermaphrodites)
▪ Eggs generally operculated (lidded); except
schistosomes
▪ Obligate parasites of mollusks, vertebrates
Development
1. Egg
2. Miracidium (ciliated, lacks mouth; infects
first intermediate host)
3. Sporocyst (elongated sac, produces rediae)
4. Redia (larval stage with oral sucker)
5. Cercaria (larval stage; may be infectant)
6. Metacercaria (encysted cercaria)
7. Adult
DIAGNOSIS
DIAGNOSTIC IMAGING
▪ Medical imaging (e.g. ultrasound, CT scan,
MRI)
LAB RESULTS
▪ E.g. blood tests
▪ Direct microscopy
OTHER DIAGNOSTICS
▪ Serologic tests
TREATMENT
MEDICATIONS
▪ Anthelmintic
SIGNS & SYMPTOMS
▪ See individual trematodes
263
Chapter 102 Trematodes (Flatworms)
CLONORCHIS SINENSIS
osms.it/clonorchis-sinensis
PATHOLOGY & CAUSES
▪ Parasitic fluke; invasion of biliary tree →
liver infection
▪ AKA Chinese liver fluke
▪ Morphology
▫ Adult: flat, elongated body; 25 x 5mm
▫ Egg: oval-shaped; 30 x 15μm
▪ Intermediate hosts
▫ First: freshwater snail (e.g.
Parafossarulus manchouricus)
▫ Second: freshwater fish/shrimp
▪ Reservoirs: cats, dogs
▪ Transmission
▫ Ingestion of raw/undercooked fish/
shrimp
▪ Infectious form: metacercariae
▫ Ingestion of metacercariae → excyst in
duodenum → migration to biliary tract
→ inflammation, epithelial hyperplasia
▪ Endemic to Eastern Asia (e.g. China, Japan,
Philippines, Vietnam)
RISK FACTORS
▪ Recent travel to endemic areas
▪ Consumption of raw/undercooked fish/
shrimp
COMPLICATIONS
▪ Pancreatitis, cholangitis, liver abscesses,
cholangiocarcinoma
DIAGNOSIS
DIAGNOSTIC IMAGING
Ultrasound, CT scan, MRI
▪ Enlarged gallbladder, hepatomegaly,
bile duct inflammation, dilated/thickened
intrahepatic bile ducts
Endoscopy
▪ Visualization of adult organisms
LAB RESULTS
▪ Acute infection
▫ Eosinophilia, ↑ IgE
▪ Chronic infection
▫ ↑ alkaline phosphatase, ↑ bilirubin
▪ Direct microscopy
▫ Detection of Clonorchis eggs in stool
samples
▫ Formalin ethyl-acetate concentration
technique (FECT) → parasite separation
from faeces
▪ Serologic tests
▫ E.g. ELISA
▪ Polymerase chain reaction (PCR)
TREATMENT
MEDICATIONS
▪ Anthelmintic (e.g. praziquantel)
SIGNS & SYMPTOMS
▪ Mostly asymptomatic
▪ Acute infection: fatigue, right upper
quadrant abdominal pain, indigestion,
diarrhea, flatulence
▪ Chronic infection: fatigue, weight loss,
abdominal discomfort, diarrhea, dyspepsia,
jaundice (severe cases)
Figure 102.1 An adult Clonorchis sinensis
worm.
264
AfraTafreeh.com exclusive
PARAGONIMUS WESTERMANI
osms.it/paragonimus-westermani
PATHOLOGY & CAUSES
SIGNS & SYMPTOMS
▪ Parasitic flatworm; causes pulmonary
paragonimiasis
▪ AKA Japanese lung fluke
▪ Morphology
▫ Adult: oval-shaped body with spines;
15 x 8mm
▫ Egg: oval-shaped, thick shell; 100 x
55μm
▪ Intermediate hosts
▫ First: freshwater snails (e.g.
Semisulcospira spp.)
▫ Second: crustaceans (e.g. crabs,
crayfish)
▪ Transmission
▫ Ingestion of raw/undercooked
crustaceans (e.g. crab, crayfish)
▪ Infectious form: metacercariae
▫ Ingestion of metacercariae → excyst
in duodenum → penetration of
peritoneal wall → migration to lungs →
encapsulate, mature → inflammation,
fibrosis
▪ Endemic to Eastern Asia (e.g. China, Japan,
Philippines, Vietnam)
Pulmonary
▪ Early infection
▫ Systemic: fever, malaise
▫ Pulmonary: dyspnea, cough, pleuritic
pain
▫ Gastrointestinal: diarrhea, epigastric
pain
▪ Late infection
▫ Malaise
▫ Recurrent, chocolate-colored
hemoptysis
RISK FACTORS
X-ray, CT scan, MRI
▪ Brain
▫ Skull X-ray: soap-bubble calcifications;
calcified cysts
▫ CT/MRI: grape clusters; conglomerated,
cystic lesions
▪ Lungs
▫ Pleural effusion, parenchymal cysts/
nodules, cavitary lesions, parasite
migration tracts
▪ Poor sanitary conditions
▪ Seafood consumption in endemic areas
COMPLICATIONS
▪ Meningitis, encephalitis, seizures
Extrapulmonary
▪ Cerebral: headache, fever, vomiting,
seizures, papilledema, paresis/paresthesias,
visual disturbances (e.g. diplopia)
▪ Abdominal: nausea/vomiting, hematoquezia
(bloody stool), pain, hematuria
▪ Subcutaneous: tender, firm, painless,
mobile nodules
DIAGNOSIS
DIAGNOSTIC IMAGING
265
Chapter 102 Trematodes (Flatworms)
LAB RESULTS
▪ Eosinophilia, ↑ IgE
▪ Direct microscopy
▫ Detection of eggs in stool, sputum,
bronchoalveolar lavage
▪ Serologic tests
▫ Enzyme-linked immunosorbent assay
(ELISA), immunoblot
TREATMENT
MEDICATIONS
▪ Anthelmintic (e.g. praziquantel,
triclabendazole)
SCHISTOSOMES
osms.it/schistosomes
PATHOLOGY & CAUSES
▪ Blood flukes; parasitize mesenteric veins/
vesical venous plexus → gastrointestinal/
genitourinary tract infections
▪ AKA bilharziasis/snail fever
▪ Morphology
▫ Adult: elongated body, 1–2cm/0.39–
0.79in
▫ Eggs: not operculated
▪ Intermediate host
▫ Snails
▪ Transmission
▫ Contaminated freshwater contact
▪ Infectious form: cercariae
▫ Contact with cercariae in fresh water →
skin penetration → schistosomulae →
migration to liver through circulation →
adult form → migration to mesenteric
venules/vesical venous plexus → egg
deposits → inflammation → fibrosis
▪ High-prevalence area is sub-Saharan Africa
RISK FACTORS
▪ More common in individuals who are
biologically male, rural areas
▪ Recent contact with fresh water bodies in
endemic areas
COMPLICATIONS
▪ Bacteremia, infertility, intestinal obstruction,
nephrotic syndrome, renal failure,
cardiomegaly, acute myelopathy
Figure 102.2 A scanning electron
micrograph of a S. japonicum flatworm.
SIGNS & SYMPTOMS
Acute infection
▪ Swimmer’s itch
▫ Pruritic papular/urticarial rash, esp. legs/
feet
▪ Acute schistosomiasis syndrome/Katayama
fever
▫ Non-specific symptoms (fever, urticaria,
chills, arthralgia, myalgia, headaches)
▫ Angioedema, dry cough, abdominal
pain, diarrhea
Chronic infection
▪ Intestinal: abdominal pain, poor appetite,
diarrhea
▪ Hepatosplenic: hepatosplenomegaly, portal
266
hypertension (e.g. collateral circulation,
gastrointestinal bleeding, ascites)
▪ Pulmonary: dyspnea; pulmonary
hypertension → cor pulmonale (enlarged
right cardiac chambers)
▪ Urogenital: hematuria, pyuria, dysuria,
increased urinary frequency
▪ Neuroschistosomiasis (acute myelopathy):
seizures, sensory/motor impairment,
cerebellar syndrome (incoordination)
prednisone)
▪ Manageme pruritus (e.g. antihistamines)
OTHER INTERVENTIONS
▪ Prevention
▫ Water sanitation programs
▫ Mass therapy
▫ Control of snails (e.g. molluscicides)
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray, CT scan, MRI, abdominal
ultrasound
▪ Brain: contrast-enhancing infiltrates
▪ Bladder: wall irregularities/fibrosis
▪ Liver: widened periportal space, periportal
fibrosis, collateral pathways
▪ Lungs: patchy infiltrates, miliary nodules
▪ Spinal cord: radicular thickening,
intramyelinic lesions
LAB RESULTS
Figure 102.3 Calcified eggs of the flatworm
Schistosoma japonicum in the submucosa of
the colon of an individual previously treated
for schistosomiasis.
▪ Bladder/rectum biopsy
▫ Egg-filled granulomas in mucosa
▪ Direct microscopy
▫ Detection of eggs in stool/urine samples
▫ Kato–Katz method (thick smear)
▫ FLOTAC stool concentration method
▪ Lab tests
▫ Acute infection: eosinophilia
▫ Chronic infections: anemia
▫ Portal hypertension: thrombocytopenia
(splenic sequestration)
▫ Urogenital infection: hematuria/
leukocyturia in urinalysis
▪ Serologic testings
▫ E.g. ELISA, indirect hemagglutination
TREATMENT
MEDICATIONS
▪ Anthelmintic (e.g. praziquantel,
oxamniquine)
▪ Corticosteroids (e.g. prednisolone,
Figure 102.4 A CT scan of the abdomen
and pelvis demonstrating calcification of the
bladder secondary to schistosomiasis.
267
Chapter 102 Trematodes (Flatworms)
268
NOTES
NOTES
TRICHOMONA
MICROBE OVERVIEW
▪ Trichomonas vaginalis: pear-shaped
(pyriform), flagellated protozoa; infects
genitourinary tract
▪ Causative agent of trichomoniasis (trich),
common sexually transmitted disease
(STD)
Morphology
▪ Size
▫ 9 x 7 micrometers
▪ Motile via four flagella, undulating
membrane
▪ Rigid axostyle runs through cell from
anterior to posterior end
▪ Contains hydrogenosomes (unique energyproducing organelles)
Replication/Multiplication
▪ Humans only host
▫ Does not survive well in external
environments
▫ Multiplies when vaginal pH basic
▫ Incubation period: 5–28 days
▫ No cyst stage
TRICHOMONAS VAGINALIS
osms.it/trichomonas-vaginalis
PATHOLOGY & CAUSES
▪ Resides in lower genital tract of individuals
who are biologically female; urethra,
prostate of individuals who are biologically
male → trophozoite stage (infective
stage) → transmitted sexually → infects
squamous epithelium of lower genital tract
→ replicates by longitudinal binary fission
→ inflammatory response
RISK FACTORS
▪ Sexual activity with infected partner
▪ Multiple sexual partners
▪ More common in individuals who are
biologically female
COMPLICATIONS
▪ Pregnancy
▫ ↑ risk of premature rupture of
membranes, preterm delivery, low birth
weight
▪ Urethritis, cystitis
SIGNS & SYMPTOMS
▪ May be asymptomatic
▪ Individuals who are biologically female
▫ Watery, foul-smelling vaginal discharge;
burning; pruritus; dysuria, urinary
frequency; lower abdominal pain;
dyspareunia; vulvar, vaginal erythema
▪ Individuals who are biologically male
▫ Urethral discharge; pruritus; burning
after urination/ ejaculation
▪ ↑ risk of contracting HIV due to genital
inflammation
269
Chapter 103 Trichomona
DIAGNOSIS
LAB RESULTS
Microbe identification
▪ Saline microscopy (wet mount of genital
secretions)
▫ Characteristic organism
▫ ↑ polymorphonuclear leukocytes
▪ ↑ vaginal pH (> 4.5)
▪ T. vaginalis assay
▫ Detects species-specific ribonucleic acid
(RNA); vaginal swab/urine specimen
▪ Nucleic acid amplification testing (NAAT)
▪ Trichomonas rapid test
TREATMENT
MEDICATIONS
▪ Systemic 5-nitroimidazole drugs (e.g.
metronidazole, tinidazole)
▫ Treat both partners
OTHER INTERVENTIONS
▪ Rate of transmission decreased with
consistent use of condoms, spermicidal
agents (e.g. nonoxynol-9)
OTHER DIAGNOSTICS
▪ Speculum exam
▫ Punctate hemorrhages cervix
(strawberry cervix)
270
NOTES
NOTES
TRYPANOSOMA
GENERALLY, WHAT ARE THEY?
DIAGNOSIS
PATHOLOGY & CAUSES
▪ Genus of flagellated parasitic protozoa
Morphology
▪ Elongated body
▪ Flagellum: forms undulated membrane
along body
▪ Kinetoplast: functions as mitochondrion
Transmission
▪ Through vectors
▪ Incubation period: 1–2 weeks
▪
▪
▪
▪
Direct microscopy
Serologic testings
Laboratory findings
Additional diagnostic tests may be
necessary (severity, infection sitedependent)
TREATMENT
▪ See individual pathogens
SIGNS & SYMPTOMS
▪ See individual pathogens
TRYPANOSOMA BRUCEI
osms.it/trypanosoma-brucei
PATHOLOGY & CAUSES
▪ Protozoan, extracellular parasite → African
trypanosomiasis
▪ AKA “sleeping sickness”
▫ Neurologic alterations during
meningoencephalitis stage (e.g.
somnolence)
Virulence factors
▪ Antigenic variation
▫ Changes variant surface glycoprotein
(VSG) → immune response evasion
▪ ↑ interferon gamma → ↑ host T. brucei
susceptibility (mechanism unknown)
Morphologic forms (life-cycle)
▪ Epimastigote → procyclic trypomastigote
(in tsetse fly midgut) → metacyclic
trypomastigote (infectious form)
Subspecies
▪ Trypanosoma brucei rhodesiense (acute,
more severe disease course)
▪ Trypanosoma brucei gambiense
(progressive, milder disease course)
Reservoirs
▪ Domestic animals, lions, hyenas, antelopes
271
Chapter 104 Trypanosoma brucei
Vector
▪ Male/female Glossina flies, AKA tsetse flies
▫ Ideal conditions: warm, humid
climate (e.g. near river/lake); altitude <
1800m/5905ft
Transmission
▪ Saliva inoculation via fly bite; vertical/
parenteral transmission very rare
▪ Endemic regions: sub-Saharan Africa;
Democratic Republic of Congo (most cases)
Pathogenesis
▪ Glossina bite → subcutaneous metacyclic
trypomastigote inoculation → lymph
vessels → bloodstream → ↑ tumor necrosis
factor (TNF) alpha, interleukin 6 (IL-6),
nitric oxide → ↑ capillary permeability →
vasculitis → organ invasion, e.g. central
nervous system (CNS)
Disease stages
▪ Hemolymphatic (early) stage
▪ Meningoencephalitis (late) stage
▪ Symptom severity related to number of
organisms in affected tissue (e.g. blood,
CNS)
RISK FACTORS
▪ Recent endemic area travel
▪ Dense vegetation near human settlement
COMPLICATIONS
▪ Meningitis
▪ Myocarditis, heart failure
▪ Aspiration → bacterial pneumonia;
associated with altered state of
consciousness (meningoencephalitis stage)
SIGNS & SYMPTOMS
Hemolymphatic stage
▪ Systemic symptoms
▫ Intermittent fever, headache, malaise,
weakness, pruritus, rash
▪ Trypanosomal chancre
▫ Rubbery, painful, erythematous, wellcircumscribed lesion at fly bite site
approx. one week post-inoculation
▪ Lymphadenopathy
▫ Winterbottom’s sign: enlarged mobile,
soft posterior cervical triangle lymph
nodes
▪ Hepatosplenomegaly
▪ Dyspnea
▪ Chest pain
▪ Altered thyroid function
▪ Impotence (biologically-male), amenorrhea
(biologically-female)
▪ Pain, Kerandel sign (deep hyperesthesia)
Meningoencephalitis stage
▪ AKA “Sleeping sickness”
▪ Cachexia
▪ Sleep disturbances (e.g. diurnal
somnolence, nocturnal insomnia)
▪ Headaches
▪ Altered state of consciousness
▪ ↓ cognitive function
▪ Personality, behavioral change
▪ Muscle spasms, ataxia, tremor, flaccid
paralysis, choreiform movements
▪ Psychiatric manifestations (e.g. psychosis)
DIAGNOSIS
DIAGNOSTIC IMAGING
MRI
▪ Cerebral
▫ May show multifocal white matter
hyperintensity (indicates late-stage
disease)
LAB RESULTS
Serologic testings
▪ Card agglutination test for trypanosomiasis
(CATT)
▫ Blood + drop of reagent with
trypanosomal antigen
▪ Immunofluorescence
▪ Enzyme immunoassays
Cerebrospinal fluid (CSF) examination
▪ Disease staging essential
▪ ↑ leukocytes
▪ ↑ proteins
▪ IgM/Trypanosoma presence
272
▪ Morula/Mott cells (pathognomonic)
▫ IgM-filled plasma cells
Direct microscopy
▪ Organism observation in lymph node
aspiration, bone marrow, CSF, blood (thin/
thick Giemsa-stained smears)
Laboratory findings
▪ Hemolytic anemia; leukocytosis;
thrombocytopenia; ↑
erythrocyte sedimentation rate;
hypergammaglobulinemia;
hypoalbuminemia, hypocomplementemia;
↑ C-reactive protein; coagulation
abnormalities
Histological observation
▪ Meningoencephalitis stage (CSF sample)
▫ Morula/Mott cells in white
matter (pathognomonic), edema,
microhemorrhages, perivascular
proliferation
OTHER DIAGNOSTICS
Electroencephalogram (EEG)
▪ Late stage: abnormal, slow delta waves
TREATMENT
MEDICATIONS
▪ Antiprotozoal medication
▫ Hemolymphatic stage: pentamidine,
suramin
▫ Meningoencephalitis stage: eflornithine,
eflornithine + nifurtimox, melarsoprol
OTHER INTERVENTIONS
▪ Prevention
▫ Vector control, surveillance
▫ Protective clothing
TRYPANOSOMA CRUZI
osms.it/trypanosoma-cruzi
PATHOLOGY & CAUSES
▪ Protozoan, intracellular parasite
▫ Causes American trypanosomiasis, AKA
Chagas disease
▪ Morphologic forms (life cycle)
▫ Amastigote (intracellular, no flagellum)
→ epimastigote (in triatomine midgut)
→ trypomastigote (infectious form)
▪ Reservoirs
▫ Opossums, armadillos, canines
▪ Vectors
▫ Triatomine bugs (“kissing bugs”)
▫ Common species: Rhodnius prolixus,
Triatoma dimidiata, Triatoma infestans
▫ Characteristics: size (2–3cm/0.79–
1.18in); obligated hematogenous; feeds
at night; lives in dark, warm sites (e.g.
closets, thatched roofs)
Transmission
▪ Triatomine bite → fecal wound
contamination
▪ Contaminated food/water ingestion
(infection through mucous membranes)
▪ Parenteral (e.g. blood transfusion, sharing
syringes)
▪ Vertical (mother → fetus)
Endemic regions
▪ Rural areas of southern U.S., Latin America
Pathogenesis
▪ T. cruzi trypomastigote inoculation →
bloodstream → organ invasion (heart,
enteric nervous system) → interstitial
inflammation → tissue destruction →
fibrosis
Disease stages
▪ Acute phase: 8–12 weeks
▪ Indeterminate phase: decades
273
Chapter 104 Trypanosoma brucei
▪ Chronic phase: cardiac/gastrointestinal
disease
RISK FACTORS
▪ Recent endemic area travel,
immunosuppression, blood transfusion,
organ transplant, intravenous drug use
COMPLICATIONS
▪ Heart failure, acute myocarditis,
meningoencephalitis, systemic/pulmonary
embolism, sudden death
SIGNS & SYMPTOMS
Acute phase
▪ Mostly asymptomatic
▪ Systemic: malaise, fever, anorexia,
headaches
▪ Chagoma: nodular skin lesion at infection
site; usually on face/extremities
▪ Romaña’s sign: unilateral eyelid edema,
conjunctivitis, preauricular lymphadenitis;
follows conjunctival inoculation
▪ Lymphadenopathy
▪ Hepatosplenomegaly
Indeterminate phase
▪ Asymptomatic
Chronic phase
▪ Cardiac manifestations: dyspnea, fatigue,
palpitation, chest pain, edema, mitral/
tricuspid regurgitation murmur, splitting of
S2
▪ Gastrointestinal manifestations: megacolon
(constipation, bloating, abdominal pain);
megaesophagus (dysphagia, regurgitation)
Congenital disease
▪ Systemic: low birthweight, anasarca, fever
▪ Petechiae
▪ Hepatosplenomegaly
▪ Neurologic abnormalities (e.g. hypotonia,
tremor)
DIAGNOSIS
DIAGNOSTIC IMAGING
▪ Further studies: stage, clinical syndrome
dependent
Figure 104.1 The kissing bug, Triatoma
infestans, is found in Central and South
America and is a vector for Chagas disease.
Chest X-ray, MRI, echocardiogram
▪ Enlarged cardiac silhouette (cardiomegaly)
▪ Pericardial effusion
▪ Valvular regurgitation
▪ Left ventricular aneurysm
Barium studies
▪ Megacolon, megaesophagus
LAB RESULTS
▪ Polymerase chain reaction (PCR)
274
▪ Blood culture
Direct microscopy
▪ Organism observation in thin/thick blood
smears; for acute phase disease (high
parasitemia)
Serologic testing
▪ E.g. enzyme-linked immunosorbent assay
(ELISA), immunofluorescence
Xenodiagnosis
▪ Feed laboratory triatomes with person’s
blood → examine feces weeks later
Cardiac tissue microscopy
▪ Acute disease: intracellular pseudocysts
(amastigotes inside myocardiocytes),
interstitial inflammation
▪ Chronic disease: mural thrombi, interstitial
fibrosis, myocardiocyte necrosis
Figure 104.2 Trypanosoma species seen on
a peripheral blood smear from an individual
with Chagas disease.
OTHER DIAGNOSTICS
ECG
▪ Arrhythmia evidence: bundle branch/AV
block
TREATMENT
MEDICATIONS
▪ Antitrypanosomal treatment
▪ Advanced cardiac disease: cardiac arrest
prevention, ventricular fibrillation through
antiarrhythmic medication
OTHER INTERVENTIONS
Prevention
▪ Vector control, treat parasitemia before
conception
275
NOTES
NOTES
TUBERCULOSIS
MICROBE OVERVIEW
▪ Tuberculosis (AKA Mycobacterium
tuberculosis) mycobacterium that primarily
infects lungs but may infect any bodily
organ/tissue
▪ Important properties
▫ Curved rod shaped bacteria often
wrapped together in cord-like
formations
▫ Obligate aerobe
▫ Impervious to Gram staining due to
waxy cell wall composed of fatty acids
(e.g., mycolic acid)
▪
▪
▪
▪
▪
▫ Staining: acid-fast stains like Ziehl–
Neelsen, fluorescent stains like
auramine/rhodamine
Clumped colonies
Distinctly slow growing (up to 6 weeks for
visible growth)
Grown on Lowenstein–Jensen media
Resistant to weak disinfectants, can survive
on dry surfaces for months
Can avoid mucus traps, getting into deep
airways (alveoli)
MYCOBACTERIUM TUBERCULOSIS
osms.it/mycobacterium-tuberculosis
PATHOLOGY & CAUSES
TYPES
Primary tuberculosis
Reactivation tuberculosis
▪ In about 5–10% cases of primary TB
Extrapulmonary tuberculosis
▪ May involve any organ (most commonly
kidneys, meninges, lymph nodes, etc.)
▪ Systemic miliary tuberculosis
STAGING
▪ Transmitted by inhaling infectious aerosol
droplets from individual with active TB (e.g.
coughing, sneezing, speaking, etc.)
▪ TB enters lungs, gets phagocytized by
macrophages → TB produces enzymes that
inhibit lysosome and phagocytic vacuole
fusion → bacteria survives, proliferates,
creates localized infection → primary
tuberculosis development
▫ TB infiltrated macrophage fusion →
Langhans giant cells
▫ Cell-mediated immunity activation →
granuloma forms within infected area →
caseous necrosis inside granuloma →
Ghon focus
▫ Lymphatic dissemination of TB → lymph
node caseation
▫ Ghon focus + involved lymph node →
Ghon complex
▫ Ghon complex fibrosis, calcification →
Ranke complex
▪ Primary infection resolution
▫ Mycobacteria killed by immune system
▫ Bacteria walled off in granuloma
remains dormant but viable →
latent tuberculosis with no further
complications in immunocompetent
276
individuals
▪ Compromised immune system → more
caseous necrosis areas → cavity formation
→ reactivation tuberculosis
RISK FACTORS
▪ Immunocompromised states
▫ HIV
▫ Diabetes mellitus
▫ Hematologic malignancy
▫ Chronic lung disease (especially
silicosis)
▫ Malnutrition
▫ Aging
▪ Substance abuse
▫ Alcoholism
▫ Injection drug users
▪ Close contact with individuals with active
TB infection
▫ Healthcare providers
▫ Incarceration
▪ Lower-income, medically underprivileged
countries
▫ Recent immigrants from highprevalence countries
COMPLICATIONS
▪ Bronchopneumonia
▪ Pneumothorax
▪ Extrapulmonary tuberculosis
▫ Kidney → dysuria, pyelonephritis with
sterile pyuria
▫ Meninge → meningitis
▫ Lumbar vertebrae → Pott disease
▫ Liver and gallbladder → hepatitis,
obstructive jaundice
▫ Lymph nodes → cervical tuberculous
lymphadenitis (scrofula)
▫ Peritonitis
▫ Pericarditis
▪ Systemic infection
Figure 105.1 The gross pathological
appearance of a Ghon focus.
SIGNS & SYMPTOMS
▪ Primary tuberculosis
▫ Usually asymptomatic (90-95% of
cases)
▫ Mild flu-like illness
▫ Rarely pleural effusion
▪ Reactivation tuberculosis
▫ Constitutional symptoms (fever, chills,
night sweats, fatigue, appetite loss,
weight loss, pleuritic chest pain)
▫ Cough (dry cough, prolonged
cough producing purulent sputum,
hemoptysis—suggesting advanced TB)
▫ Crepitations during lung auscultation
▪ Extrapulmonary tuberculosis
▫ Depending on affected organ/tissue
▪ Miliary (disseminated) tuberculosis
▫ Can affect any organ (e.g. choroidal
tubercles in eye, granulomas within
organs)
▫ Weight loss
▫ Fever, chills
▫ Dyspnea
277
Chapter 105 Tuberculosis
DIAGNOSIS
DIAGNOSTIC IMAGING
Chest X-ray
▪ Used in PPD/IGRAs positive
▪ Ranke complex → sign of healed primary
TB
▪ Cavities → active TB sign
Antibiotic resistance
▪ Multiple-drug-resistant TB
▫ Resistant to isoniazid and rifampin
▪ Extensively drug-resistant TB
▫ Resistant to both isoniazid and rifampin,
any fluoroquinolone, at least one
second-line drug
LAB RESULTS
PPD intradermal skin test (tuberculin test)
▪ Screening test for people at high risk for TB
▫ Tuberculin injection between dermal
layers, induration area measurement
within 48–72 hours
▪ Induration area ≥ 5mm: positive in
immunocompromised individuals, persons
with primary TB radiographic evidence/
close contact with those with active TB
▪ Induration area ≥ 10mm: positive in
residents/immigrants from high-prevalence
countries, children > four years of age, high
risk populations (e.g., medical employees)
▪ Induration area ≥ 15mm: considered
positive in individuals with no known risk
factors
▪ Cannot be used for differentiation between
active and latent TB
▪ PPD result interpretation
▫ Positive → exposure evidence
▫ False-positive → previously immunized
with BCG vaccine
▫ Negative → no exposure evidence
▫ False-negative → sometimes seen
in individuals with sarcoidosis,
malnutrition, Hodgkin’s lymphoma
Figure 105.2 An X-ray image of the chest
demonstrating diffuse interstitial granular
densities in an individual with milliary
tuberculosis.
Sputum testing
▪ Used for definitive diagnosis
▪ Staining, culture, PCR
OTHER DIAGNOSTICS
Interferon gamma release assays (IGRAs)
▪ Alternative for PPD
▪ Unlike PPD, doesn’t show false-positive
results in BCG vaccinated
Figure 105.3 Multifocal patchy opacities
in the right upper lobe of an individual who
presented with night sweats, weight loss and
persistent cough. The presenting symptoms
and radiological appearance are consistent
with pulmonary tuberculosis.
278
TREATMENT
MEDICATIONS
▪ Prophylactics
▫ BCG vaccine (some countries)
▪ Latent TB
▫ Isoniazid for 9 months
▪ Active TB
▫ First line anti-TB drugs: isoniazid,
rifampin, pyrazinamide, ethambutol/
streptomycin
▪ Antibiotic resistance
▫ For multiple-drug-resistant TB,
treatment requires second-line drugs
(amikacin, kanamycin, capreomycin)
OTHER INTERVENTIONS
Figure 105.4 The histological appearance
of a tuberculosis granuloma. The granuloma
is formed of epithelioid macrophages and
giant cells with a focus of caseating necrosis
at the centre and a rim of lymphocytes at the
periphery.
▪ Active TB
▫ Compulsory isolation (until sputum
negative for TB)
279
Download