• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/129

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

129 Cards in this Set

  • Front
  • Back

to infect a cell, virus must

attach, enter, multiply, target appropriate organ

to be maintained in nature virus must

shed, taken up by vector (insect, needle), passed congenitally

local replication

confined to organ of entry

systemic replication

involved many organs

Respiratory tract entry

most important sit of entry, via aerosol of infected nasal secretions. large droplets - nose, small droplets = LRT alveoli

Respiratory tract barriers

mucus, cilia, alveolar macrophages, temperature gradient, IgA

Respiratory tract local infections

Rhinovirus - requires URT 35 degrees


Influenza - SA and tryptase Clara

Mumps virus

Systemic Respiratory tract virus.
Replicates in URT epith, binds SA. Infection of all organds including CNS, causes meningitis.


Salivary glands swell up

Measles virus

Replicates in URT epith cells, infects macro, lympho, DC and LN. Enter circulation and amplify in lymphoid tissue

Alimentary Tract entry

swallowed of infect oropharynx. Vviruses do not have an evelope. if no receptor, enter via breach in epith

Alimentary Tract barriers

constant movvement of contents (allow receptor contact), mucous, pH, proteolytic enzymes, lipolytic bile, IgA, macro

Local AT

M cells - deliver ag to lymphoid tissue by transcytosis. Viruses may trancytose of infect M cells

eg. Rotavirus double carpsid destroy villi and M cells = GE. Secrete NSP4 prot = increasse fluid secretion


e.g. Calici - ID w EM. cannot culture or animal model

Systemic AT

polio, rhino, hepato - viruses

Trascutaneous route

trauma - HPV warts


injection - HIV, Hep B/C


bite - Dengue, rabies



Genital tract

HPV, HSV2, HIV, HepB

Conjunctivia

adeno, HSV

mechanisms of spread

local spread on epith
subepith invasion and lymphatic spread


viremia


neural spread



Viremia types

free - primary and secondary phases, virus prod by liver, spleen. neutralised by ab and macro




cell ass - eg HIV CD4, measles monocytes. may persist



Poliovirus

Ingest, replicates in peters patches monoytes, travel to LN, go to blood as free virus


Can target MN = paralysis OR target gut = excreted.

Tissue invasion

Entry from blood to tissue - barriers are basement membrane and endothelial cell




Replicate and release progeny in endothelium
Trancytosis
Cross via cells - mono and lympho
pass through leaky junctions

Neural spread

via peripheral nerves. uncoated NC carried passively along axons and dendrites Replicate in nerve body can cross synapse.


Protexted from CTL - nerves = no MHC1



Rabies

neural spread. can infect muscle which is controlled by the iR. infects neurons and kills brain sites = aggression, thirst. infect salivary glands for transmission

Placenta transmission

to infect fetus.
cytocyidal = death, abortion. non cytocydal = developemental abnormalities.


baby at birth may be infected via the birth canal, fecal contamination or be immunoogically tolerant (carriage state)

Congenital Rubella

Slows rate of cell division = small babies, microencephaly, congenital heart defects, deaf, cataracts

ZIka virus

mosquitos, sexual transmission
subclinical or mild rash. severe = Guillain-Barre syndrome in adults, baby microencephaly.


NO VACC
infects human embryonic cortical neural progeniter cells, increases cell death and cell cycle dysregulation.

Congenital HCMV

Herpes, most common congenital inf.
primary inf of mother = 40% spread to fetus
recurrent inf of mother = lower risk to fetus, do not abort


no vaccine, no screening



Shedding

Respiratory tract = aerosols, nasopharyngeal secretions
Oropharynx and GIT - pharyngeal fluids, feces


Skin - warts, or vesicles/vesicular rash


Blood - Dengue and yellow fever viral levels are


Urine, milk, oral/genital secretions



Determinants of tropism

-receptor availability


-local factors - temp, ph, survival of enzymes


-accessibility - ability to replicate in cells or travel in blood, polarised release


-availability of cleavage activating proteases e.g. tryptase clara, furin


-transcription requirements e.g. HPV rep in differentiated skin cells

Pathogenicity vs virulence

P - inherent genetic capacity to cause disease


V - measure of degree of disease = qualitative.
relative virulence can be measured by mean time of death, mean time of symptoms appearing, degree of fever/weight loss, loss of immune cells

Virus induced changes in cells

-transformation


-lysis


-chronic infection - release of virus w/o cell death


-latent infection - no damage to cell, can reemerge later for lysis

Cytocidal virally induced damage

-shutdown protein synth - e.g. polio 2A cleaves elF4G reuired for translation


-shutdown NA synth - due to shutdown of protein or DNAse


-viral proteins - accumulation = cytotoxic
-apoptosis

Noncytocidal virally induced damage

-Loss of function e.g. rhinovirus kills cilia


-Infected cells = target of immune system


-Transformaation - viral oncogenes = tumour



Immunopathology

-cytokine inflammation


-ag-ab complexes deposited on kidney/blood vessel


- FcR mediated uptake o commplexes into macro/mono enhances infection


- CD4 - induct cytokines, responsible to measles rash due to inflamm


- CD8 - kill hepatocytes





Autoimmunity

molecular mimicry - produce proteins that resemble host cells




polyclonal B cell activation

Immunosuppression

-HIV replicates in CD4 (and kills them) and mono (inhib function)


- measles temporary immunosuppresion from replication in Tc and macro

Host resistance

Genetic - inherited defects (no Ig class), polymorphisms (MHC genes), IFN inducible genes, receptor genes (CCR5)




Non genetic - age, malnutrition, homones and pregnancy, dual infections



Outcomes of viral infection

fatal


full recovery


recovery w permanent damage


persistent infection

Innate immunity

First line = immediate, passive
skin, mucous membranes, lysozyme, gastric juice, body flora




Second line = require activation
NK, macro, eosino, cyotkines, fever

Ligand signalling pathway

Ligand binds, TLR dimerise, adaptor molecules assemble.




Adaptors MyD88 and TRIF activate TF and MAP kinases.




NFkB - IL-6 and TNFa
IRF3 - Type 1 IFN (a/b)

RIG-1 and MDA5

RNA helicases. Bind dsRNA and and induce dimerisation and interaction with adaptor MAVS on mitochondria via the CARD domains.


Induces activation of IRF3 and NFkB

cGAS-STING

cGAS recognises cytoplasmic DNA, generates cGAMP to bind STING and activate RF3 and NFkB

Cyotkines

Low molecular weigh proteinns with regulatory functions to alter gene expression in target cells. Work locally for antiviral defense and in larger quantities product global effects eg. fever



autocrine (self), paracrine (nearby), endocrine (distant)


JAK STAT signalling

IFNa binds Jak1, Stat heterodimer dimerises and gets phos, enters nucleus to bind ISRE




IFNgamme binds Jack heterodimer, Stat homodimer dimerises and gets phos, enters nucleus to bind GAS

ISG

proteins only produced upon IFN stimulation



MxA

-MxA found in cyto, recognises capsids. used as a marker for infection

OAS

.-OAS forms a tetramer to dimerise and activate RNAseL to degrade viral RNA RNAse L is always present in small amounts

PKR

PKR binds dsRNA, gets phos and dimerises = active. PKR phos host translation factor ELF2a stop host translation, therefore stopping any viral translation.

Viral countermeasures

RNA recognition - interfere w recognition, helicase activation, MAVS signalling or TF activation


Inhibit IFN signalling by preventing phos or nuclear umport


Inhibit PKR - cleavage, preventing activation. most viruses do this

Latency characteristics

- infect non replicating cells or viral genome is replicated with the host genome


-genome is intact


-low or no gene expression


-low or no detection of virus



Viral persistence

-Latency


-Chronic infection


-Noncytopathic inf of inaccessible site to avoid IR induction


-Acute with late complications e.g. measles to SSPE

Antigenic variation

antigenic drift = change in antigenic structure of the virus to escape neautralisation by ab




drift in Tc epitopes - change anchor residues, flanking AA or TcR contact residues



Inhibit T cell priming

-block cytokine induced maturation


-block signal transduction


-block Tc stim





Evade CD8 Tc

-antigenic variation in CTL epitopes


-endocytosis of class 1


-binding TAP = no peptide translocation to ER


-bind MHC peptide complex to keep it in the ER


-bind tapasin = loads peptide onto MHC in ER


-decrease class 1 gene transcription


- evade proteosome degradation





Evade NK cells

Inhib receptor binds MHC1, activation receptor binds to target cell




-murine CMV keeps the activation receptor ligand in the ER


-human CMV encodes MHC1 like molecule to bind the inhib receptor, upregulates HLA-E

Interfere with cytokine function

-soluble cytokine receptor homologues


-redirect Tc response


-intracellular blocking of cytokine synthesis


-intracellular interference with cytokine function

Apoptosis

Cell death mediated by caspases when cell cycle is disrupted or when there is cell stress.


DNA is fragmented


Dying cells release apoptotic bodies that APC take up and present on MHC2 for activation of CTL.



Apoptosis pathways

Extrinsic pathway


TNF/Fas bind recept, caspases, apoptosis




Intrinsic pathway


mitochondria permeabilised by BCL2, cytochrome C released, caspases, apoptosis

Autophagy

The cytoplasm gets engulfed by double membrane autophagosome, lysosome degrades.


Antiviral - targets viral components for degradation (xenophagy) and has a role in initiating innate/adaptive IS.




Viruses may use autophhagy components for replication

Complement evasion

Virus encodes homologues of control proteins that bind to complement components to stop the cascade




e.g. Vaccinnia protein binds C3b

Passive evasion of host defence

-cell tropism


-spreading via cell fusion


-free passage through barriers (needle, transplants)



Active evasion of host defence

-limit expression of viral genes


-non cytopathc infection


-block specific immune defences (cyto, antibody, MC, coreceptors_


-block apoptosis


-genetic evolution


-immunosuppressive or anergic epitopes



Herpesvirses and persistance sites

all herpesviruses form lifelong persistent inf w periodic reactivation (cytopathic)




alpha - HSV1/2, VZV = neuronal persistance


beta - CMV = monocyyte/secretory gland


gamme - EBV = lymphocytes (Bc)

Herpesvirus latent genomes and ori

dsDNA, regulated gene expression in latency




EBV = replicating latency
OriP for rep during latency in B cells
OriLyt for replication during lytic infection


HSV= non replication latency
Has 2 ori that are both used for lyic infection

HSV-1 diseases

primary inf = lips and tongue gingivistomatitis
CNS enncephalitis rare, most common in newborns, very fatal. diagnose w PCR of CSF. Virus retrograde axonal flow to trigeminal ganglion. reemerges down the SAME axon




Recurrent = cold sores

HSV1 and 2 latency

virus enters skin/mucous membrane, local multiplication and enters nerves, goes to trgem (HSV1) or sacral (HSV2) ganglion via retrograde axonal flow, latent, reactivate an travels down SAME neuron.




virus is maintained in the cell bodies, multiple copies. express LATs

LATs

Latency Associated Transcripts




non translated transcripts, no protein produce


associated with reactivation

Selection of replication and latency

IE genes are blocked by interferons. LAT is expressed to inhibit apoptosis and therefore promote latency, resulting in immune suppression

Varicella Zoster Virus infection

Primary inf = chicken pox viremia (via DC and Tc) produces vesicular rash.


Neural spread virus goes to DRG, latent, reactivate and moves back down ALL the neurons of the DRG to produce shingles dermatome rash




reactivation in elderly - immune deficit/decline

VZV vaccine

chickenpox vaccine = live attenuated, recommended for seronegative children and at risk e.g. teacher

EBV infection

glandular fever, acq by contact of infected saliva w oroparynx.


Primary inf = lytic, orophayngeeal cavity epith cells.


Then it spreads to Bc to generate wide IR, LN swelling. Bc = latency reservoir, no replication





EBV Latency

associated with expression of genes


-EBNA1-6 - EBN1 involved in replication in dividing Bc


-LMP1 and 2 = latent membrane proteins




Has 2 ori - OriP and OriLyt

EBV tumorogenesis

reactivation of EBV from Bc leads to


Hodgkins disease


Burkitts Bc lymphoma


Nasopharyngeal carcinoma

CMV

infection in older = mild, leads to persistence


reactivated by immunosuppression




congenital inf = deaf, microencephaly.


mother primary inf - 32% chance fetus gets inf,


15% smptomatic, 70% long term sequelae

HIV Tat

Tat - produced by alternative splicing,activate transcription through binding TAR RNA element to recruit RNAP



HIV Rev

regulator of structural gene expression through binding RRE to transport unspliced or partially spliced RNA to the cytoplasm.


accumulation of Rev leads to late transcription

HIV non essential proteins

Vif inncreases infectivity and blocks the IR for ssDNA


Vpr for nuclear import of cDNA, activate transcription, stop cell growth


Vpu regulates release and env processing


Nef multifunction protein



HIV entry and receptors

binds C type lectin receptors = selects for R5 strains because DC bind virus w C type lectin receptors and R5 receptor.




bind CD4, CCR5 promotes fusion. Later CXCR4 used to inf Tc in the draining LN, then viremia.


CCR5Δ32 homo deletion mt = HIV resistant

HIV phases

1 - high viral load, CD4 loss, hard to spot. ELISA and WB

2 - asymptomatic, load decreases and plateaus. Has CTL and non neutralizing ab for decoy ag. HIV evades IR


3 - symptomatic and AIDS




HIV IR evasion

sequence variation


high replication level


viral proteins to downreg MHC1


infect IS cells - Tc clonal exhaustion


latency in resting cells = priveleged site

HIV rate of progression

dependent on age, strain of virus, presence of CCR5Δ32 mt, HLA type, IR

HIV reservoirs

-LN


-Blood


-Resting memory Tc


-macro in tissue and LN


-accumuates in LN germinal centres as immune complexes on FD


-in microglia and astrocytes


-testes and secretions

Drug development issus

must have proof of principle


must quickly and completely block replication and spread, avoid persistance


resistance must be managable


safe


cheap


easy to make/deliver

Interferons

IFNab bind cell receptor to activate TF for IFN regulated genes that inhib penetration, uncoating, mRNA synth, translation, assembly, release




purified recombinant proteins for therpeutic use - side effects fever, nausea

Haemagluttanin inhibitors

HA binds to SA for infection




Amantadine


M2 ion channel blocker, binds M2 and prevents acidification of HA during fusion.



Neuraminidae inhibitors

NA cleaves SA resdues for release from infected cells




Relenza (inhale) and Tamiflu (oral)


bind to the active site of NA with higher affinity than SA does

HIV drugs

Acyclovir


nucleoside analogue, no 3'OH to inhibit DNAP. requires activation by viral TK phos to triphosphate form, therefore it is specific to infected cells


Addition of side chain changes bioavailability - can pass digestive tract

Nucleoside vs nucleotide

nucleoside has no phos groups




nucleotide has more than one phos group. used for polymerisation od DNA

Ribavarin

guanasine analogue, inhibits DNAPand RNAP


monophosphate form reduces NA synth


triphosphate form alters viral mRNA formation




Hep C, Influenza

AZT

HIV RT inhibitor


Thymine analogie, phosphorylated by cellular kinase to triphosphate form. Used in preferennce of thymine.


does not eliminate provirus


drug resistant mutant viruses occur



NRTI

nucleoside reverse transcriptase inhibitor for HIV1




T/G/C analogues, kinase phos to triphos form


used in preference to cellular nucleosides to prevent chain elongation




used individually = mutant.s therefore HAART

NNRTI

Non nucleoside reverse transcriptase inhibitors for HIV1




do not bind the nucleotide binding sit of RT, are not DNA analgoues.


inhibit RT enzyme directly by other mech

HIV protease inhibitors

peptidomimetics that have close structural similarity of the AA recognition sequence


bind to the active sit of protease

Raltegravir

blocks integrase strand transfer of HIV-1 DNA


causes the LTRs to anneal and form 1 and 2 LTR circles that degrade or recomb

Drug Resistace

-high replication and mutation rate


-selective preessure favoring combo therapy


-resistant mutants preexisting


-resistant virus often has reduced replicative capaciy and pathogenicity

HAART

potent


durabble antiviral response


minimise resistance




Rapid viral rebound when HAART stopped due to integrated provirus = undetectable viral load

Viral hepatitis

non cytolytic, replicates in hepatocytes of the liver.




acute disease (A&E) = jaundice


chronic (BCD) = cirrhosis, liver cancer


disease = immune mediated, age relatedearly exposure = less severe acute disease but higher rates of chronic inf



Diagnosis of hepatitis

sero tests (ELISA) - IgM = acute, IgG rising titre confirms acute infection




NA test - PCR blood/faeces

Hep A

picornaviridae, non env +ssRNA


resistant to stomach acid


single serotype


replicates in cell culture - inactivated vacc





Hep A and E pathogenesis

ingest orally


replicate in intestine epith


viremia


replicates in liver hepatocyte


secretes in bile/faeces



Hep E

non env BUT FRAGILE


associated with contaminated water, low person to person spread



Hep B

have double walledvirus particles and incomplete particles with only env protein


gapped circle genome, many starts one stop





Hep BCD pathogenesis

sex or injection


penetrate mucosal epith


viremia


replicate in liver


shed in blood, semen, secretions. perinatal transmission also high

Hep B sequelae

Chronic carrier (HBsAg+)


liver damage


cirrhosis, liver failure


liver cancer in 2-10% of patients, usually partial integration of the HBV genome

Diagnose Hep B infection

-HBsAg = marker of inf


-anti-HBs Ig = recovery or immunity


-IgM=acute, IgG = chronic


-HBeAg = active replication


-HBV DNA = active replication

Hep D

deltravirus with delta ag capsid




coinfects with HBV = acute


superinfects with HBV = chronic

Hep C

most infections via drug use


no vaccine, animal model, high mutation


poor immunity




NS5B RNAP mediates replication, error prone - 6 genotyes



Hep C treatment

no vaccine, only drugs


use IFNa and Ribavirin.


quasispecies may not respond

Hep B treatment

vaccine, no drugs. cannot eradicate - latency


vaccine = yeast purified sub viral particles




antiviral therapy - IFN, RT inhibitors, nucleoside analogue (Tenofivir) - used to decrease viral load in mothers



Eradicate vs Cure

eradicate = elimination of virus from Earth



cure:


complete =eliminate cccDNA, prod ab


functional = lose surface ag, prod ab. may still lead to latency

Hep B drug targets

target cccDNA -eradication involves cleavage of the genome - can reform with homologous recomb




prot x = hijacks SMC56 that chromatinises DNA in the nucleus - required for chronic.




coat protein stabilises cccDNA

Transfection

transfer of NA into eukaryotic cells




use molecular conjugates - CaPO4 precipitate of DNA or catioic liposomes for DNA/RNA

Transduction

viral vector delivery systems

Viral vector design


-replication defective - cant prod and release new infectious particles. remove sequences for pathogenicity/rep


- seperate struct genes into transpackaging cell line


- flank transgene w cis acting seq for translation and packaginng signals





Retrovirus vector

place gag/pol/env in packaging cell line


inf dividing cells
remove nonessential genes


keep LTR and packaging signals




integration into an oncogene = cancinogenic




limites env tropism


Retrovirus vector replication reversion

modify to preserve RT and integration but stop LTR transcription by replacing LTR with CMV




LTR inactive as promotor

Lentivirus vector

Similar to retrovirus


infects NON dividing cells - terminally dierentiated

Retrovirus vector applications

Gene correction therapy due to no vector IR




Cancer therapy - transient high level expression. Cytotoxicity specific to tumour cells




vaccines

Adenovirus vector

E1 genes into cell line, keep the ITR




efficient for mammalian cells, high immunogenic, high titres, short term expression, wide tropism
response against vector = no reimmunisation



use for cancer

Adeno associated vectors

small, do not require replicating cells for gene transfer, no insertional mutagenesis




can integrate into host DNA = persistant expression





RNA replicons

self replicating RNAs derived from viral genome


high expression of heterologous genes


delivered as VLP and naked RNA/DNA






delete genes for strucctural proteins

West Nile Virus replicons

high expression, non cytopathic, no integration, no recomb




use for vacc and cancer




preexisting immmunity and small size

- RNA virus vectors

-RNA genome = segments, expression depends on location of the gene.




product via genetic reassortment


parents = virulent virus with target ag and a weakly pathogenic strain


select for attenuated strains exp target ag

Smallpox vaccine

Cowpox antigenically similar, vaccinia not




able to eradicate because:


no secondary host, one stable sero, virus is infectious after symptoms occur, no persistance

Polio vaccine

not 100% successful due to provirus


SALK = inactivated vacc


SABIN = live atten vacc, lots of passages, provides mucosal IR, MAY REVERT TO WT

Measles vaccine

measles - no more SSPE, reduced encephalitis


mumps - no more orchitis, reduced encephalitis


rubella - no more congenital rubella syndrome, reduced encephalitis

Live attenuated vaccine pros and cons

pro - fewer doses, long lasting IR that is similar to the pathogen response




con - consider immunocomp, needs boosters, can revert to virulence and be shed



Vaccinia vector

construct plasmid with viral TK disrupted (TK-) by early gene promotor with gene inserted downstream.




infect cells with plasmid + WT vaccinia. select for TK- cells with BUdr = toxic in TK+ cells




DNA vaccines

plasmid contains ori, euk promotor, vaccine antigen, poly A and marker (antibiotic resistance)




can target specific cells by expressing specific molecules

VLP

capsid or env proteins can self assemble, are taken up efficiently by APC




HepB vaccine from yeast


HPV Gardasil vaccine from yeast L1 capsid prot