Sie sind auf Seite 1von 16

Herpesvirus (dsDNA, enveloped icosahedral capsid)

Virus & Disease Characteristics Epidemiology Pathogenesis Dx, Rx, Px


Alphaherpesviridae Reproductive cycle: HSV-1 and HSV-2 HSV-1: associated with infection above the Cell culture; look
HSV-1 Short share many waist for
HSV-2 Primary target cell: properties HSV-2: below the waist, and has greater cytopathogenic
Mucoepithelial cells Human are the only potential to cause viremia effect (CPE)
o Vesicular lesions of the skin; Site of latency: neuron natural hosts Formation of
fluid contain infectious virions, Cause lytic (Asymptomatic/ Local infection of the oral/mucoepithelial multi-nucleate
heals without scar. infection of most recurrent ds is cells syncytia

o Oral Herpes Simplex/ herpes cells and latent source of Contact with the cutaneous r/c of local
Serology for
labialis/cold sore infection of contagion) sensory nerves epidemiology
Most often occurs in neurons Risk group of 1ry Vesicular rash
infancy/childhood infection: newborn, Virus transported retrogradely along the culture (fluid,
Gingivostomatitis young children, microtubules of the axons scraping, swab
most common young adults CPE, PCR, IxFx
presentation in Virus attaches to and penetrates the CNS (CSF)
young children trigeminal ganglia
Transmission: PCR, virus

(painful) Contact Establishes latency isolation
Pharyngitis more Sexual *
common Prenatal Reactivation of virus Rx: Acyclovir/ other
presentation in nucleoside analogues
adolescent HSV-1: >90% positively Virus returns to initial site of infection
DDx: HFMD, seroconvert by year 2
chancre. Recurrent HSV infection
HSV-2: Seroconversion
o Genital Herpes (usually HSV- occur later in life
2) *UV, stress, pregnancy, trauma/surgery,
o HSV Infections of the Skin syst. ds, cancer
Herpes simplex
dermatitis
Herpetic Whitlows
(fingers; dentists)
Herpes Gladiatorum
(wrestlers and rugby
players)
Eczema Herpeticum
Erythema Multiforme
CPE Ballooning of cells
Other clinical findings:
Keratoconjunctivitis
Severe encephalitis (temporal
lobe)
Neonatal herpes (HSV-2, in-utero)
Alphaherpesviridae Varicella (Chicken pox) Unlike HSV, VZV is Day 0 Day 3-6 Rx: High dose acyclovir,
HSV-3 (VZV) Acute, mild, transmitted predominantly by Airborne route Mucosa of oral (prevent 2ry infection)
highly respiratory route. URT/conjunctiva replicate in
Varicella contagious Viremia Form skin lesions regional lymph node 1ry Dx:
o Incubation period: 14d disease, mostly all over the body viremia replicate in liver & Ab detection
o Fever, malaise, anorexia affecting spleen DNA detection
o Skin maculopapular rash, children Epidemiology Day 7 Day 14 Virus isolation
itch Generalized Worldwide Virus continues to replicate in the Histology smears
Trunk head vesicular Varicella; highly liver & spleen 2ry viremia (start
(scalp) limbs eruptions of the communicable, of sx), transported to skin
Macule papule skin and mostly children <10 (vesiculopustular rash), lungs
clear filled vesicle mucous Zoster; adult-elderly (major source of contagion), gut
cloudy vesicle membrane and sometimes brain
maculopapular Zoster (Shingles) Virus remain cell associated and
eruption scabbing Reactivation of transmitted by cell to cell
(heal) VZV present in interaction
o Cx; 2ry bact infection, latent form in Gain access to trigeminal and
pneumonia, CNS involvement neurons in dorsal root ganglia latency
Varicella in Pregancy sensory ganglia
o Virus cross placenta Localization of Reactivation
o 2 types of intrauterine sd one or few Virus replicates and released
Congenital Varicella dermatomes along the entire neuronal
Syndrome (1st gest) pathway to infect skin causing
Neonatal Varicella vesicular rash (Shingles)
(final phase);
unilateral, first 4wks
of life, pneumonitis,
encephalitis, damage
to msk system

Zoster
o Rash limited to skin
innervated by a single sensory
ganglion
o Intense inflammation and pain
precedes skin lesion
o Cx; Ophthalmic zoster,
Postherpetic neuralgia
o Trunk, head, neck (T3-L3),
unilateral
Ophthalmic Zoster
o Trigeminal nerve, ocular cx
o Followed by Postherpetic N
Betaherpesviridae Reproductive cycle: Endemic in all parts of the Normal Host Dx:
HSV-5 (CMV) long world. Incubation period: 4-8wks PCR (detect
Primary target cells: Systemic dissemination replicating virus)
Clinical Features Monocytes and Transmission May readily establish lifelong Isolation of virus;
Normal Host granulocytes Sexually: semen, latent infections throat washing
o Spontaneous Infectious Latency: T cells cervical secretions, Virus shed intermittently from and urine.
Mononucleosis syndrome saliva, even urine pharynx and in urine for months Serology (IgM,
(less severe pharyngitis and Inapparent Vertically: in-utero, to years after primary infection IgG)
lymphadenopathy) infection is breast milk Reactivation of latent infection
o Cx: hepatitis (jaundice) common during Organ transplant occurs by immunosuppression
childhood and and blood borne (eg: corticosteroids)
Immunocompromised adolescent (WBC)
o Pneumonia is a freq cx The most
o Disease of transplantation common Risk group
Bone marrow congenital Individuals who
transplant recipient; infection lead to attend/work at a
interstitial severe daycare centre CPE (Owls eye) on
pneumonitis congenital Pts who undergo fibroblast cell culture
Solid organ anomalies blood transfusion
transplant recipient; Frequently Person with multiple Rx: Ganciclovir (DOC),
leukopenia found in adults sex partners Acyclovir, Valacyclovir
Heart transplant; who are Tissue
graft atherosclerosis immunocompro transplantation Px:
Renal allograft; CMV mised Isolation of
related rejection newborns
o Untreated AIDS pt; Screening of
disseminated disease transplant donors
and recipients
Congenital & Perinatal
No vaccine
o Small size, microcephaly,
jaundice,
hepatosplenomegaly, rash.
o Virus detected in
Infants urine (1st wk)
Maternal breastmilk
Ab in breast milk
does not prevent
transmission
Betaherpesviridae Replication >90% of children
HHV-6 mainly in CD4 T over age 1 and
HHV-7 lymphocytes adults are virus
Virus present in positive
saliva Typically occur in
Clinical findings Latency site not early childhood
o 1ry infection Exanthem known Mode of
Subitum (Roseola transmission
infantum/Sixth Disease) presumed to be via
o Infection persists for life oral secretions
Gammaherpesviridae Major target cell: B cells - Worldwide (>90% Reactivation from latency Dx:
HSV-4 (EBV) Latent site: B cells seropositive) Clinically silent Isolation &
Replicates in: Epithelial - >50% symptomatic Evidenced by increased levels of identification of
Clinical findings cells of oropharynx, in adolescent virus in salica and of DNA in virus (NA
o Lymphoid & epithelial tumours parotid gland, uterine - No EBV vaccine blood cells hybridization,
o Infectious mononucleosis cervix Immunosuppression is known to viral Ag marker,
o Cancers reactivate infection, sometimes viral isolation)
Burkitt lymphoma; with serious consequences Serology
fever, night sweats, (ELISA, IxFx)
swollen lymph
nodes, weight loss Rx: Acyclovir reduces
NPC EBV shedding but doesnt
affect immortalized B cells
Gammaherpesviridae A new herpesvirus Transmission: orally, Dx:
HHV-8 (KSHV) discovered in 1994. sexually, vertically, blood No readily test
Replicate in: borne, organ transplant available
Clinical findings: Lymphocytes Blood test
o A blood cancer called Outstanding feature: - Frequently occur in
Kaposis sarcoma molecular AIDS patients Rx: Ganciclovir (effective
o Lymphoma piracy/molecular mimicry - Africa >50% in preventing KS, but
o Castlemans disease (some (Rather than dev its own - Italy, Greece, Israel, unsure on tumors that
forms of severe lymph node oncoprotein, it steals Saudi Arabia >10% already exist)
enlargement) many genes from host - North America
cell) <10%
Hepatitis Viruses

Virus Characteristics Epidemiology Clinical Features Dx, Px, Rx


Hepatitis A Nucleic acid: ssRNA, +ve Routes of transmission No chronic infection. Dx test:
Classification: Picornaviridae - Fecal oral/ Acute Infection
Contamination of Sporadic IgM anti-HAV
Non enveloped, acid food or water Transmission Chronic Infection
and heat stable. - Close personal o Oral (common) not applicable
One serotype, multiple contact o Percutaneous (rare)
genotypes. - Blood borne (rare) o No sexual/perinatal Immunity: IgG anti-HAV
In vivo replication: Average incubation period: 30d - Pre-exposure
cytoplasm of Jaundice: Adult (30%), Children (travellers to high
hepatocytes (humans (<5%) risk regions)
and higher primates Fulminant <1% - Post-exposure
Onset of symptoms (within 14 days)
o Fatigue
o Abdominal pain Case fatality rate: 0.1-2.7%
o Loss of appetite
o Nausea & vomiting
o Dark urine
o Jaundice
Hepatitis E Nucleic acid: ssRNA, +ve - Fecal oral Similar to HAV; more severe in preg. Dx test:
Classification: Unclassified, transmission Can cause severe acute hepatitis. Acute Infection
hepevirus (Contaminated No chronic infection. IgM anti-HEV
water supplies) Chronic Infection
Non enveloped, acid - Mainly young adults Transmission not applicable
stable - Can infect primates, o Oral (common)
One serotype with swine, sheep, rats o No sexual Immunity: not applicable
genetic heterogeneity - Maternal-infant o Yes perinatal Case fatality rate:
In vivo replication: transmission occurs Incubation period: 15-16d 0.5-4% in non-pregnant
cytoplasm of (last trimester) and Clinical illness at presentation: 70- 1.5-21% in pregnant
hepatocytes (humans & is often fatal 80% in adults
higher primates) Jaundice (common)
Fulminant <1%, in pregnancy 30%
Hepatitis C Nucleic acid: ssRNA, +ve Prevalence Transmission Dx test:
Classification: Flaviviridae - Worldwide (3%) o No oral Acute Infection
Enveloped - Africa (5.3%) o Percutaneous (common; HCV RNA, Anti-
- Eastern IVDU) HCV
Mediterranean o Yes sexual (rare) Chronic Infection
(4.6%) o Yes perinatal (low freq) HCV RNA, Anti-
- SEA (2.15%) Incubation period: 14-160d HCV >6mnths
- Americas (1.7%) Clinical illness at presentation: ALT (>x10)
- Europe (1%) jaundice (5-10%) Chronic pt may
Fulminant: rare have normal ALT
With/ without
jaundice

Immunity: no vaccine
available
Rx: Ribavirin & IFN therapy
Case fatality rate: 1-2%
Hepatitis B Nucleic acid: dsDNA Risk factors Transmission Dx test:
Classification: Hepadnaviridae - Perinatal o Oral (not likely) Acute Infection
- Sexual o Percutaneous (common) HbsAg, IgM anti-
Enveloped - IVDU o Sexual (common) HBC
Multiple serotypes and - Organ transplant o Perinatal (common) Chronic infection
genotypes A-F - Hemophiliac Incubation period: 60-180d HbsAg, IgG anti-
- Pt with chronic renal Jaundice: 5-20% HBC
failure/ Fulminant <1% HBeAg (high
hemodialysis Chronic infection infectivity)
- Healthcare workers o >90% infants Anti-HBc IgM
- Tatooing/ body o <5% adults (Acute Infection)
piercing Anti-HBe (Low
HBV Gene Products infectivity)
Risk of chronic infection: - Core gene; HBcAg, HBeAg Anti-HBs
Neonates - Surface; HBsAg (Immunity)
- Polymerase HBV DNA
Terminal protein (priming) (Viremia/Active
Reverse transcriptase replication)
Rnase H
Immunity: HBIg, HBV
Vaccine (IgG anti-HBc, anti-
HBs)
Rx: IFN-, Pegylated IFN-,
Lamivudine, Adefovir,
Entecavir, Telbivudine,
Tenofovir
Hepatitis D Nucleic acid: ssRNA - HDV requires HBV to replicate and Dx test:
Classification: Unclassified, delta frequently assoc with severe Serological test for
virus acute/chronic hepatitis delta Ag or HDV
- Co-infection or superinfection IgM and IgG
HBV Envelope
One serotype, 3 Immunity: no vaccine
genotypes
Respiratory Viruses

Virus Characteristics Epidemiology Pathogenesis & Clinical Features Dx, Px, Rx


Orthomyxoviruses Virion: spherical, Epidemiologic pattern differ: Surface Ag of influenza undergo antigenic Dx:
Influenza A pleiomorphic, helical Influenza A can variation Usually based on
Influenza B nucleocapsid sweep across - Antigenic drift: Minor changes (point characteristics of
Influenza C Genome: ssRNA, -ve, continents mutation) amino acid changes symptoms
segmented (pandemic) escape from immune system local Specimen (nasal
Envelope: Contain HA Influenza B cause outbreaks washing, gargles,
(hemagglutinin) and NA epidemics - Antigenic shift: Major changes in HA throat swab)
(neuraminidase) (exclusive human & NA (Only Influenza A) new RT-PCR
Replication: Nuclear virus) subtypes pandemic Isolation and
transcription, particle Influenza C cause identification of virus
mature by budding sporadic respiratory Gene reassortment: (CPE)
from plasma disease Infection of two/more members of the Serology
membrane influenza virus of the same genus, mixtures of
Outstanding 3-5 million cases, parental gene segments may be assembled Rx:
characteristics: 2.5-5mil deaths into progeny virions Acetaminophen
Influenza A undergo worldwide Antihistamines
Airborne droplet/contracted with contaminated
genetic reassortment; Epidemic outbreak: Zanamivir and
hands/surface
cause worldwide seasonal Oseltamivir (target
epidemics Escape Ab neutralization NA)
Subtypes: based on People at risk Amantadine and
HA and NA (Only Seronegative Infect and kill epithelium Rimantadine
Influenza A has people (uncoating step)
subtypes) Adults: classic flu Progeny virus kill adjacent cells
Vaccine
syndrome
Viral NA lower the viscosity of the mucus
Children:

asymptomatic to Spread virus-containing fluid to lower portion of
severe RTI the respiratory tract
High risk groups:
elderly and Many cells are killed
immunocompromis
ed, people in Pathology
nursing homes or Incubation period: 1-4d
with underlying IFN and cytokines detected in
cardiac/ respiratory respiratory secretion
problems Cellular destruction is repaired may
be exposed to 2ry bacterial infection
(staph, strep, H.influenzae)

Clinical findings
Acute influenza (Adults): Rapid onset
fever, chills, headache, dry cough,
myalgia, malaise, anorexia
Acute influenza (Children): Higher
fever, GI manifestation, febrile
convulsion, otitis media, croup
Cx: pneumonia (1ry viral, 2ry
bacterial), neurologic sd (Reyes sd,
GBS, encephalitis)
Paramyxoviruses Morphology: spherical Measles (Rubeola) Replication: Lab Dx RSV:
Respiratory Genome: ssRNA, -ve, linear, Acute, highly - Attach to host cells - Antigen detection
Syncytial Virus non-segmented infectious disease - Virion envelope fuses with the cell (IxFx, ELISA)
(RSV) Envelope: contain viral HN charac by fever, membrane by F protein - Isolation and
Measles Virus (hemagglutinin that sometime resp sx, and a - Transcription, translation, RNA identification of virus
Mumps Virus carries neuraminidase activity) & maculopapular rash replication takes place in the - Nucleic acid
fusion (F) glycoprotein fusion, Cx are common cytoplasm (-ve sense use +ve detection
hemolysis and viral entry (pneumonia, sense as template) - Serology
Replication: cytoplasm; particle diarrhea, - Maturation
buds out from plasma encephalitis) budding of cell surface
membrane Leading cause of - Assembly & release;
Outstanding characteristics: death in young M protein important
antigenically stable, particle are children NA activity prevent self-
labile yet highly infectious In pregnancy aggregation of virus
abortion, stillbirth, Exit without killing cell
Classification based on specific premature birth
features Airborne/droplet RSV CPE (Syncytial
Pneumovirus cause Encephalitis caused by
Escape Ab production
formation)
pneumonia (eg:RSV) measles:

Henipavirus cause 0.5% occurrence, Infect respiratory epithelia Dx Mumps:
zoonotic infection (eg: 1.5% mortality - Usually clinical
Nipah virus) 2 ways: Common cold, croup [Type 1,2,3 RSV] - Sx of mumps are
Classification based on HN & F: postinfectious non-specific
Respirovirus & encephalitis Viremia [Measles, Mumps]/Spread to lower - Classic finding of
Rubulavirus possess (immune mediated), trachea & bronchi Pneumonia/bronchitis parotid gland
direct infection of [RSV, type 3]
HA and NA activities tenderness and
Measles has HA but neurons swelling (day 3)
Subacute RSV:
lacks NA activity, has - Lab Ix: Isolated viral
F. sclerosing Inapparent infection or the common culture from saliva,
cold pneumonia in infants to
F of RSV possess panencephalitis urine and CSF
(SSPE) very rare bronchiolitis in very young babies - Serology
fusion property but not
hemolysin. RSV lacks and late sequelae Wheezing
HA and no NA of measles Reinfection is common in both adults Rx Mumps:
and children
Mumps cx: In elderly may cause symptoms To provide comfort;
Measles (Rubeola) - Aseptic meningitis similar to influenza virus disease. Analgesics
Structure differs from (50%) Pneumonia may develop (acetaminophen,
other paramyxoviruses - Meningoence- RSV is an important cause of otitis ibuprofen), warm or
Spikes carry HA but not phalitis after media cold packs.
NA function inflammation of MMR Vaccine (80%
F protein is also a salivary glands Measles: effective)
haemolysin - Meningitis > Regional lymph nodes
Only one serotype encephalitis
- Usually resolve Viremia/monocytes & lymphocytes
Mumps without sequelae
Lytic infection of cells of conjunctiva, RT, UT,
Highly contagious viral - Others; arthralgia in
lymph, blood vessels and persistant infection of
infection young male, CNS
Self-limited disease but myocarditis,
presence of abortion in pregnant Rash caused by T cell response
complications might be women
severe Prodromal illness; high fever, cough,
Envelope: HN coryza, conjunctivitis, photophobia
(agglutinate RBC and Koplik spots (red spots with white
attachment), F (fusion, centre on buccal mucosa) appear
hemolysin, antigenic) after 2d
No cross immunity with Within 24hr exanthema start below
parainfluenza virus ears and spread over
Replication: Nasal or
URT epithelial cells Mumps:
Viremia to salivary Prodromal period: malaise & anorexia
glands & other major Classic parotitis; tender, earache
organ systems Propensity to replicate in various
visceral organs; kidney, CNS, testes
and ovaries

Virus entrance into URT



Local lymph nodes/1ry viremia

Salivary glands/Testes, ovaries, CNS, pancreas

2ry viremia

Kidneys

Viruria
Enterovirus & Poliovirus

Virus Epidemiology Pathogenesis Dx, Rx, Px


Picornaviridae Enterovirus Enterovirus pathogenesis Dx polio:
Enteroviruses - Resist pH3 to pH9, detergent, mild 1. Entry into the body Virus usually
o Polio sewage treatment, heat a. Cell surface receptors mediate tissue present in the
o Coxsackie A - Transmitted fecal-oral route or tropism throat and in the
o Coxsackie B respiratory route (aerosol droplet) b. Poliovirus receptor (restricted) stools before onset
o Echovirus - Asymptomatic shedding can occur up c. Coxsackie and Echovirus receptors are Virus may be found
Rhinoviruses to months more widespread on tissues (broader in the blood of pt
Hepatoviruses - Schools and day care settings disease spectrum) with aseptic
Parechoviruses 2. Viral replication and disease progression meningitis
Kobuviruses Poliovirus a. Primary infection Ab to virus appear
- Vaccine era i. Mucosa and lymphoid tissues of early in the disease
Virion: Icosahedral Wild type poliovirus has tonsils and pharynx Specimen: throat
Genome: ssRNA, +ve, linear been eliminated from ii. Lymphoid cells of Payers swabs, rectal
Replication: Cytoplasm developed countries patches and underlying intestinal swabs, stool
Non enveloped Paralytic polio is still mucosa Cell culture
prevalent in Nigeria, b. Primary viremia Genome and
Genera of diseases Afghanistan, Pakistan c. Secondary viremia and sx serology: RT-PCR,
Enteroviruses Mutation in live vaccine virus i. Disseminated to target organ IF, ELISA, IgM
disease of GIT and re-establishes ii. Potential target organ: CNS and (four fold)
able to cause CNS neurovirulence skin
disease Salk (killed virus) IPV 3. Immunity: protection via Ab Symptomatic Rx:
Rhinovirus Sabin (live attenuated virus) 4. Release of virus back to environment by fecal - Acetaminophen
Nasopharyngeal ds trivalent OPV shedding (fever)
- Children are more susceptible than
Hepatovirus HAV - Mouthwashes and
adults, causing infantial paralysis, but Poliomyelitis; acute infectious ds that affects the CNS in its
Parechovirus GIT sprays (oral
infection in adults would produce serious form. The destruction of motor neurons in the spinal discomfort)
and RT
more severe sx cord results in flaccid paralysis.
Kobuvirus Viral GE - Fluids
- Levels of hygiene Only a fraction develop paralytic disease (dehydration)
Asymptomatic infection (90%) - Cold milk
Enterovirus serotypes
Polio (1-3) Abortive/minor illness (5%) (neutralize acid
Coxsackie A (1-22, Non-paralytic progression to CNS or aseptic juices that irritate
24) meningitis (1-2%) mouth ulcers)
Coxsackie B (1-6) Paralytic polio (0.1-2%) - Topical
Echovirus (1-9, 11- o Spinal polio destruction of motor neuron diphenhydramine
27, 29-34) o Bulbar (cranial) polio destruction of (hand and foot
Other enteroviruses cranial nerve discomfort)
(68-71) - IVIg treatment
Paralytic poliomyelitis
- Spinal polio
Viremia virus infect skeletal ms and
travel up innervating nerves spread
along axons of peripheral nerves to CNS
destruction of motor neuron of anterior
horn of spinal cord and motor cortex of
brainstem
Nerve damage extend of paralysis
Asymmetric flaccid paralysis with no
sensory loss
- Bulbar polio
Involve the brainstem where the cranial
nerves are located
Involve muscles of the pharynx, vocal
cords and respiration
75% death inability to swallow, breath

Postpoliomyelitis; years after infection

Coxsackie viruses
Group A
o Herpangina (vesicular pharyngitis),
HFMD, acute hemorrhagic conjunctivitis,
diarrhea, pneumonitis
Group B
o Pleurodynia (pleuritic chest pain),
myocarditis, pericarditis, severe infantile
generalized disease
A number of group A and B serotypes can give rise
to meningoencephalitis and paralysis

Enterovirus 68-71
Nonspecific fever, aseptic meningitis, paralysis,
myocarditis, sepsis
Enterovirus 70 hemorrhagic conjunctivitis
Enterovirus 71 HFMD, polio-like syndrome
Arboviruses

Virus Epidemiology Pathogenesis Dx, Rx, Px


Family: Togaviridae Transmission pattern of Arboviruses: General clinical manifestations:
Genera: Alphavirus Human-arthropod cycle (Eg: Urban Four types of reactions to Arboviruses
- Chikungunya yellow fever, dengue, chikungunya) A mild fever: sometimes with a rash
Lower vertebrate-arthropod A long term fever: persistent arthritic
Family: Flaviviridae Human accidental host (Eg: Jungle symptoms, often with a rash
Genera: Flavivirus yellow fever, St. Louis encephalitis) Hemorrhagic fever: serious
- Dengue Arthropod-arthropod cycle, Encephalitis: serious, often fatal
- Japanese Encephalitis occasionally humans and lower
- West Nile Fever vertebrate (Eg: Colorado tick fever)
- Yellow Fever
- St. Louis Encephalitis

Family: Bunyaviridae
Genera: Bunyavirus, Plebovirus,
Nairovirus, Hantavirus
Dengue Fever (DF) Tropic areas Viral pathogenesis Dx:
4 main serotypes DENV o Poorly planned Bite: Skin inoculation by vector Clinical finding
1,2,3,4 urbanization Travel history
Virus infects immature dendritic cells/macrophage
Vector; female Aedes o Global population growth
FBC
o Aedes aegyptii o Increased air travel Migration of virus to lymph nodes Hematocrit (Correlate
(day) 50-100mil infections per year well with plasma
o Aedes albopticus worldwide Activation of cellular and humoral immune response volume loss and
(day and night) >2.5 billion people at risk of disease severity)
infection Viral replicates in macrophage for 3-6 days (incubation Serology;
period to viremic state)
Fatality rate <1% with appropriate o Hemagglutinin
therapy Inhibition (HAI)
Clinical criteria of dengue infection
Warm and wet conditions are test detect
Asymptomatic (50-90%)
favoured by the mosquitoes early but cross
Classic DF rapid onset high fever, HA,
Transmission react with
retro-orbital pain, myalgia, vomiting, sore
o Epidemic introduced as other
throat, maculopapular rash flaviviruses in
an isolated event, involves
single viral strain Dengue hemorrhagic fever/Dengue shock late infection
o Hyperendemic syndrome bleeding and endothelial leak Direct detection
continuous circulation of o RT-PCR
multiple viral serotypes Clinical Manifestations and Pathophysiology o NS-1 Ag
o Persons with DENV in their 1. Incubation period detection by
blood can transmit the 2. Febrile phase ELISA (IgM Ab
viruses to the mosquito 1 a. High grade fever (2-7d) capture
day before onset of febrile b. Usually: facial flushing, skin ELISA)
erythema, generalised body ache,
period and remain myalgia, arthralgia, HA
infectious for the next 6-7d c. Sometimes: sore throat, injected IgG ELISA
o A. aegypti bites pharynx, conjunctival infection - Paired samples
interruptedly during blood d. Common: anorexia, nausea and - Differentiate between
meals vomiting 1ry and 2ry infections
o Mosquito remains e. DHF: petechiae, bruising - Confirmatory test
infectious for its 1-month 3. Critical phase (Day 3-7)
life a. Progression of leukopenia Lab confirmation: Plague
(neutropenia) reduction and neutralization test
b. Mod-severe thrombocytopenia (PRNT)
c. Plasma leakage; hemoconc. And
hypoproteinemia Prognosis:
d. Sweat, cool extremity, restless, DHF; treated 2.5% mortality,
skin and mucosal bleeding untreated 50% mortality
e. Shock
4. Recovery phase Management
a. Resorption of leaked plasma - Acetaminophen for
b. Stable hemodynamic status pain/fever
c. Profound fatigue for several - Monitor development of
weeks plasma leakage
(haematocrit)
DHF/DSS - Monitor hydration status
Ab-dependent enchancement - Blood transfusion if
Pre-existing non neutralizing Ab (IgG) bing significant bleeding
to DENV [AgAbCx] - Careful volume
Viral entry through monocytes and other repletion
FcR-presenting target cells
Suppression of innate immune response; Control; No vaccine, surveillance
release of inf cytokines and chemokines, control, vector control
enhanced disease
Japanese Encephalitis (JE) 50,000 cases annually, with - Target cell: T lymphocytes and periph. - Antigen relatively
Vector; Culex mosquitoes permanent neurological or Mononuclear cells neurons homogenous &
4 distinct genotypes psychological handicap, 25% died - More severe in young and elderly recovery results in solid
Birds pigs/cattle/dogs individual protection
Humans are dead-end host - Early: Fever, GI symptoms, HA - Cut trans of disease,
Cannot be spread from human to - After 3-14d: rapid LOC, stupor coma education and
hman and eating pork - Survivor: brain damage vaccination
Rural areas - Seizures in children - No effective drug avail,
All genotypes: Malaysia, Indonesia ribavirin can be given
Zika Virus Transmission: Mosquito bites, mother to Sx: fever, rash, joint pain, conjunctivitis
Vector; Aedes child (transplacental), sexual contact, blood In pregnancy: microcephaly, other fetal brain
transfusion defects
Retroviridae

Virus Epidemiology Pathogenesis Dx, Rx, Px


Lentiviruses In 2010, WHO estimated that 34 mil HIV primarily infects CD4 T cells and cells Lab Dx:
Human Immunodeficiency people were living with HIV, of whom of macrophage lineage Screening tests

Viruses >30 mil were living in low-middle income o Requires blood
Virus causes lytic infection of CD4 T cells
o HIV-1 countries and persistent low level productive sample
o HIV-2 2014 36.9 mil people living with AIDS infection of macrophage lineage cells o Detect the bodys Ab
worldwide, more than half do not know response to HIV
All retroviruses have 3 essential they are infected, 1.2 mil died Virus causes syncytia formation, with response
genes which encode polyprotein cells expressing large amounts of CD4 o Test does not detect
precursors for viral replication Transmission antigen (T cells); subsequent lysis occurs HIV
Gag (group specific - Blood o 4th generation Ab/Ag
Virus alters T cell and macrophage cell
antigen) gene encodes Open cuts tests p24 (detected
function
o Viral matrix (MA) Breaks in the skin during window period)
p17 Mucous membranes Virus reduces CD4 T cell number and o Assay for viral Ag and
o Capsid (CA) Direct injection helper cell maintainence of CD8 T cell and viral genome can also
p24 Occupational exposure macrophage function detect very early/very
o Nucleo-protein - Sexual contact late infections
(NC) MSM, FSF, MSF CD8 T cell number and macrophage o Exposed should be
function decrease
Pol gene encodes Semen retested at least 6
o RT including Vaginal secretion months after last
Three Phases of HIV exposure
RNase H - Perinatal
o Intergrase (IN) Breast milk o EIA/ELISA
provirus - Not transmitted by casual contact Confirmatory tests
integration o IFA
o Protease (PR) HIV and STDs o Western Blot; show
encodes prod STDs increase infectivity to HIV atleast 2 of p24, gp41
that cleaves gag o Increase in HIV viral shedding and gp120/160
Env(elope) gene o More WBC carrying HIV in Viral load tests
o Surface mucosa of genital area
glycoprotein (SU) STDs increase susceptibility to HIV EIA/ELISA
o Transmembrane o Ulcerative and inf. STDs +ve
Phase I: Primary infection/Acute Repeat EIA/ELISA
(TM) polyprotein compromise the
infection +ve
mucosal/cutaneous surfaces of IFA/Western blot
- Lasts 3-8wks
Morphology: Type C Lack of genital tract that normally acts +ve
morphologically recognizable as barrier against HIV - Virus replicates to high titer
Positive for HIV
(usually of RS type which is M
intracytoplasmic form. o Ulcerative STDs: syphilis,
tropic MO tropic)
chancroid, genital herpes HIV Testing
- When CTL develop,
Genome: ssRNA, +ve, 2 copies of o Inf. STDs: chlamydia, - Requires informed consent
RNA genome gonorrhoea, trichomoniasis seroconversion occurs and viral
- Premarital testing
load diminished in blood
Replication: Depends on RT Effect of HIV on immune system recommended
Enveloped increases risk of STDs - Prenatal testing not required
o Supressed IR due to HIV can: Phase II: Asymptomatic/latent/ but recommended
Classification of HIV-1 Genital ulcer chronic phase
Major group M reactivation - Lasts for months to >15y, HIV Risk Reduction
Outlier group O Rate of abnormal average 10 y - Avoid drug and alcohol use
New group N cell growth - Continuation of virus replication - Dont share needles used by
- More than 90% of HIV-1 Difficulty in curing mainly in lymph node others for; drugs, tattoos, body
infections belong to HIV-1 reactivated/newly - Active IR and antigenic escape piercings
group M acquired genital ulcer - Concluded by development of - Avoid exposure to blood
Risk of becoming opportunistic infection (CD4 T products
infected with cell <200/mm3) - Practice safe sex; use
additional STDs condoms
Phase III: AIDS
HIV Post Exposure Prophylaxis - Host immune response HIV vaccines
HIV Occupational Exposure collapses - Preventive vaccines
Medical follow up Constitutional sx - Therapeutic vaccines
Baseline and follow-up HIV testing Opportunistic
4wk course of medication initiated one infections
to two hours after exposure Neoplasm
LFT to monitor medication tolerance - Clinical progression to AIDS
Exposure precautions practiced - Syncytium inducing T tropic T
lymphocyte tropic (x4 viruses)
are detected in most if not all
cases

Das könnte Ihnen auch gefallen