Disease and Defense Unit 3 Flashcards

1
Q

Define and describe infection, infectious disease, pathogenicity, and virulence

A

Infection: process where a microbe enters into a relationship with a host. It may or may not cause disease (can be good or bad). usually stable, can be transient

Infectious disease: disease caused by infection with microbe. may be COMMUNICABLE, or not (transmitted from patient to patient, is not the case for all microbial bugs, e.g.: rabies). relationship that results in disease state.

Pathology: the ability (of a microbial species) to cause disease. Microbes that can cause disease are FRANK PATHOGENS. microbes that cause disease in compromised hosts but less in normal host are OPPORTUNISTIC PATHOGENS

Virulence: degree of pathogenicity (of a specific strain or species). a highly virulent microbe is likely to cause disease when introduced into host in small numbers.

  • note: both microbial and host factors contribute to the outcome of an infectious disease. “damage response” framework.
  • etiologic agents: organism/something that is directly or indirectly responsible for the disease state
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Explain how a microbe is shown to be the cause of a specific disease.

A

-Koch’s Postulates: criteria for a bacteria/microorganism/etc. that causes disease

  1. microbes are present regularly in characteristic disease lesions
  2. specific microbes can be isolated and grown in vitro
  3. injection of cultured microbes into animals reproduces disease seen in humans
  4. specific microbes can be re-isolated from lesions of the disease in animals

limitations:

  • some do not have a characteristic/pathogenic lesion
  • some microbes cause specific infectious diseases but can’t be grown in vitro
  • traditional concepts of pathogenicity focus on properties of microbe vs. hosts
  • characteristics of infectious diseases usually reflect complex interactions btw microbes and their hosts
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Describe typical stages in pathogenesis of an infectious disease and explain their importance.
(EESMDO)

A
  1. Encounter: how agent and host meet. when contracted endogenously (from flora) or exogenously (from environment). need to ask what is route and dose of infection.
    - source, route, dose
    - modes of spread are among people and from animals to people
  2. Entry: how the agent enters the host. what does it need to cross? is it actively (invasion) or passively (injection, bites, transfusion, transplant, etc.)
    a) colonization of body surfaces is an important 1st step
    b) adherence, by specific binding of microbial surface components (pili, non-pilus adhesions) to receptors on host tissues
    - colonization of body surfaces, adherence
  3. Spread: how agent spreads from entry site. microbial products can promote or inhibit spread
    a) hyaluronidase, elastase, collagenase, facilitate spread of microbes through tissues (spreading factors)
    b) coagulase inhibits spread of microbes by promoting deposition of fibrin to wall off and localize infections
    - crossing mucosal surfaces, spreads in tissues
  4. Multiplication: how agent multiplies in host. normal flora and pathogens must replicate the host at rates that exceed clearance by defense mechanisms. growth rate in vivo may be slower than in lab (in vitro)
  5. Damage: how tissue damage is caused by agent/host response, and are there virulence factors
    a) some microbial products cause direct damage to host
    b) some products cause damage to host via blocking or interfering with defense mechanisms
    c) most of products causing direct damage to host interfere with host defense mechanisms. they are surface components of microbes, products that can be secreted, products that can be injected into a target cell. can be used as protective antigens for vaccine development
  6. Outcome: does the microbial agent or host win the battle or do they coexist? what would be consequences?
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Compare mechanisms of innate and acquired host defense against infections.

A
  • **add in table from notes
  • Rhinovirus: same virus can range 200 fold based on route of infection (nasal cavity vs. pharynx)
  • route of infection and effect and determine the effective dose
  • Cholerae: depending on route of infection, but what also is taken with it, has a 10,000 fold difference in infectivity. EG: take with bicarb, neutralizes stomach pH, and makes the oral route more effective since not as much destroyed in stomach.
  • genetic differences in host: mouse pox example. route of infection was the same (injected footpad), mice were genetically different. Based on strain, the dose needed was huge.

Look at table to grasp idea but do not memorize it!!!

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Describe the composition and importance of the microbiome of the body.

A

-some prevalent bacteria of the normal flora of the skin, oropharynx, large intestine, vagina

  • some factors that differ among people and affect micro biome: diet, suppression of flora by antibiotics, anatomic abnormalities, genetic differences between individuals
  • examples of physiologic importance of micro biome: effects on tissue/organ differentiation, production of vitamins by gut flora, biochemical conversions, competition with pathogens for body surface colonizations
  • growth rate doubling times vary between viruses
  • slow doesn’t mean non-pathogenic
  • growth can be intra or extra cellular

**add in table from notes (don’t memorize it!!! just be familiar with trends)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Compare several disease paradigms that illustrate selected mechanisms of pathogenesis.

A
  • Cholera-toxin mediated disease
  • pneumococcal pneumonia-acute inflammation by invasive, extracellular bacteria
  • tuberculosis-infection by a facultative intracellular bacterium
  • rheumatic fever-pathology mediated by immune response
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Define the basic properties of viruses

A
  • submicroscopic, obligate, intracellular parasites
  • genetic material enters host cell and directs production of building blocks of new particles
  • genome is encased in a capsid, and it hijacks the cell to create what it wants
  • not alive, do not grow or divide
  • genome is replicated and directs synthesis of viral components and will be assembled to form progeny viruses
  • particles are produced from self assembly of newly synthesized components in a host cell
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

List the strategies viruses employ for survival.

A

-since not alive, one view is that they are chemicals.

  • employ 3 part survival strategy:
    1) house DNA/RN genomes in small, pertinacious particles (capsids)
    2) genome contains all info they need to initiate/complete infectious cycle
    3) establish a relationship in population of hosts that range from benign to lethal

-particle architecture, size/nature/topology of genomes, protein coding strategies, cell/tissue/host tropism, pathogenesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Describe two means of classifying viruses.

A
  1. classical system: viruses are grouped according to shared physical properties
    -nature of genetic material in iron (DNA/RNA)
    -symmetry of capsid (helical or icosahedral)
    -naked or enveloped
    -dimensions of viron and capsid
    -linnaen/biological categorization of verses governed by ICtV
    -starts at level of order and continues as:
    Order (-virales)
    Family (-viridae)
    Subfamily (-virinae)
    Genus (-virus)
    Species
    -Species names generally take the form of [Disease] virus,
    e.g. Hepatitis virus.
  2. Baltimore system: based on central dogma, DNA–>RNA–>protein
    - all viruses are parasites of the host mRNA translation machinery and therefore must produce mRNA to decode genome
    - categorizes virus based on how they produce mRNA

-originally were classified based on the fact that they were filterable (below a certain size)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q
Describe basic methods for studying viruses.
Electron microscopy
animal models
sequence analysis
cell culture
serology
other
A

Electron microscopy: type of microscope that uses a beam of electrons to create an image of the specimen. It is capable of much higher magnifications and has a greater resolving power than a light microscope, allowing it to see much smaller objects in finer detail.

Animal models: allow for better understanding the disease process without the added risk of harming an actual human. The species chosen will usually meet a determined taxonomic equivalency to humans, so as to react to disease or its treatment in a way that resembles human physiology as needed.

sequence analysis: the process of subjecting a DNA, RNA or peptide sequence to any of a wide range of analytical methods to understand its features, function, structure, or evolution. Methodologies used include sequence alignment, searches against biological databases, and others.

cell culture: the removal of cells from an animal or plant and their subsequent growth in a favorable artificial environment.

serology: study of serum and other bodily fluids. In practice, the term usually refers to the diagnostic identification of antibodies in the serum.

other:
- we can grow large amounts of viruses in eggs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q
Identify the main structural characteristics of virus particles.
viral genomes
protein capsids (helical, icosahedral)
envelopes
virus attachment proteins
A

Viral genomes: made of DNA or RNA

Protein capsids: the genome delivery device, can be helical or icosahedral

  • Helical: simplest way to arrange identical subunits, use rotational symmetry and arrange around circumference to form a disk.
  • icosahedral: arrange proteins in a hollow, quasi-spherical structure enclosing the genome inside. number of ways to do this depending on shape of subunits. Nature uses triangle as repeating unit. solid shape of 20 triangles with 12 vertices and faces with 2, 3, 5 fold symmetry. smallest is 60 identical subunits

Envelopes: lipid bilayers acquired during assembly of viral particles, typically have viral glycoproteins embedded in membrane. many are acquired via budding though host membrane. don’t have to kill cell during production.
-glycoproteins are integral membrane proteins with 1 or 2 membrane spanning domains

Virus attachment proteins:

  • Virus attachment involves specific binding of a virus-attachment protein with a cellular receptor molecule.
  • Many examples of virus receptors are now known. Target receptor molecules may be:
    a) proteins (usually glycoproteins)
    b) carbohydrates (found on glycoproteins or glycolipids)
  • Carbohydrate receptors tend to be less specific than protein receptors because the same configuration of carbohydrate side-chains may occur on many different glycosylated membrane bound molecules.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Describe the seven basic virus genomes, protein expression strategies and replication strategies of each.

A
7 classes of viral genome configuration:
dsDNA
gapped circular dsDNA
ssDNA
dsRNA
ss(+)RNA
ss(-) RNA
ss(+)RNA with DNA intermediate
  • if dsDNA genomes are grouped with gapped circular dsDNA, then 6 types of genomes correspond to the classification in the Baltimore system.
  • Viruses are obligate intracellular parasites they rely on the host cell biosynthetic machinery to varying degrees for their propagation.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q
Describe the typical, generalized replication cycle of a virus.
Double stranded
Single stranded
Gapped circular
Viruses with RNA
A

-exact copies of viral genome must be produced and packaged

Double stranded strategies:

  • replication in nucleus, dependent on cellular factors. may involve viral polymerase and accessory factors
  • replication in cytoplasm (poxviruses) occurs when they have all the factors necessary for replication of genomes and are independent of cellular machinery

Single Stranded DNA:
-replication in nucleus with formation of double stranded intermediate as template for synthesis of single stranded genome

Gapped circular double stranded DNA:
-virally encoded reverse transcriptase to copy viral genome from mRNA transcribed from template genome

Viruses with RNA genome:
-transcription and genome replication is highly integrated for RNA viruses. genome is replicated using anti-genome as template. production of anti genome is done by RdRp.

Basic Steps:

1) Attachment of virus to the cell.
2) Entry of virus into the cell and uncoating of the viral genome.
3) Viral gene expression.
4) Viral genome replication.
5) Assembly of new viruses and egress of new virus particles from the cell.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Compare and contrast acute local disease versus acute systemic disease in incubation periods, virus shedding and transmission, host responses, and likelihood of re-infection.

A

** add in table from notes
Acute infection: high viral replication rate and the production of large number of progeny. persistent lasts for longer.

Acute Local: infections of epithelial cells at body surface (gut, respiratory tract, eyes). short incubation times, include many serotypes (rapidly mutating and result in short lived immunity, via IgA). re infections common. cold and diarrhea

Acute Systemic: primary infection also in epithelium. viremia and systemic infection result in secondary replication at various sites, lifelong immunity and includes IgA and IgG. Measles and smallpox

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Describe the expected outcome of viral infection and disease in a healthy host versus an immune compromised host, including symptoms, shedding and transmission, and length of primary infection.

A

-The expected outcome of viral infection and disease in an immune compromised host (relative to a healthy individual) is: worse and longer. The point is that when you are diagnosing someone or considering impact on community health, you must be aware that the textbooks symptoms and times have to be adjusted for certain cases.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Detail two differences between persistent and latent infection.

A
  • Persistent - usually refers to virus infections that continue to produce new virus over a long period of time, it never completely settles down.
  • Latent - refers to virus infection in which the virus genome is relatively silent, there is little gene transcription in most infected cells, and there is little to no disease in a healthy host for a long time, in many cases for life.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

List the potential outcomes and types of virus diseases, with the contributing factors.
acute viral and acute systemic
chronic viral (persistent, latent)
transforming

A
  • *** add in photo from slides
  • acute viral infection: local/systemic depends on initial events of infection/virus tropism
  • acute local: infections in epithelial cells at body surface. re-infection common.
  • acute systemic: primary infection also in epithelium, viremia and systemic infection result of secondary replication at various sites. lifelong immunity IgA,G.
  • chronic viral: ongoing virus infection/replication with mild inapparent disease in healthy host. coincident with immune suppression
  • persistent: continue to produce new virus
  • latent: vires is silent for a long time (even for life). may be reactivated/ recrudescence

-transforming viral: integrate into host genome and activate oncogenes. cause ongoing inflammation and contribute to tumerogenesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

List effects of viruses on infected cells, such as CPE, syncytia, growth, apoptosis.

A
Cytopathic effects (CPE) of virus infection: Direct cell damage and death from viral infection may result from diversion of the cell's energy, shut off of cell macromolecular synthesis,  competition of viral mRNA for cellular ribosomes,  competition of viral promoters, and enhancers for cellular factors inhibition of the interferon defense mechanisms.
-- any detectable morphologic changes in the host cell.

Indirect cell damage can result from integration of the viral genome, induction of mutations in the host genome, inflammation, and host immune response.

Syncytia are multinucleate masses of cytoplasm that have not separated into individual cells.

Growth and apoptosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Explain the interferon response and “anti-viral state”.

A
  • Infection of cells with viruses induces the production of proteins that are known as interferons because they were found to interfere with viral infection of neighboring cells.
  • The anti-viral state is a state that can be induced in neighboring cells to prevent the virus from spreading.

Mediators of the IFN-induced anti-viral state:

1) PKR- protein kinase that phosphorylates and thereby inactivates a cellular translation initiation factor, resulting in decreased protein synthesis.
2) OAS- 2’-5’Oligoadenylate synthetase activates a cellular ribonuclease (RNAseL) that degrades mRNA.
3) others, many unidentified

IFNs bind to specific receptors on cells & induce transcription of genes = antiviral state.
-PKR and OAS are made as inactive precursors and are activated by dsRNA.
-Once activated, these gene products shut down translation of both cellular and viral mRNAs.
IFNs can also induce the synthesis of gene products that arrest the cell cycle (blocking virus replication), or induce a pro-apoptotic state.
-IFNs can induce the synthesis of proteins that are involved in the presentation of virus proteins to cytotoxic T lymphocytes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Distinguish between innate and adaptive anti-viral responses.

A

Innate defenses : natural barriers, cells, soluble factors.
-Natural barriers: skin, mucus, ciliated epithelium, gastric acid, tears, and bile.
-Cells: macrophages, neutrophils, dendritic cells, and NK cells.
-Soluble factors: interferons, cytokines, complement, chemokines.
-Innate immunity is non-specific (responds to infection) and immediate (within hours of infection).
-Innate immunity and the inflammatory response are critical
precursors to the adaptive immune response to viruses. Innate responses are triggered in many ways.
-Interferons (IFN)

What does the adaptive immune response recognize?

  • All viruses encode proteins- these are foreign antigens that can elicit specific/adaptive immune responses.
  • Viral antigens may be found on virions, surfaces of infected cells, debris from infected cells, or as peptide fragments bound to host MHC molecules on infected cells or phagocytic cells.
  • recognizes cell mediated or humoral immunity
  • recognizes antigens in a more specific way than TLRs
  • Viral pathogens are recognized by the cells in which they replicate, leading to the production of interferons that serve to inhibit viral replication and to activate NK cells. The induced innate responses either succeed in clearing the infection or contain it while an adaptive response develops.
  • Adaptive immunity harnesses many of the same effector mechanisms used in the innate system, but is able to target them with greater precision. Thus antigen-specific T cells activate the microbicidal and cytokine-secreting properties of macrophages harboring pathogens, while antibodies activate complement, act as direct opsonins for phagocytes, and stimulate NK cells to kill infected cells. In addition, the adaptive immune response uses cytokines and chemokines, in a manner similar to that of innate immunity, to induce inflammatory responses that promote the influx of antibodies and effector lymphocytes to sites of infection.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Compare antibodies produced in primary and secondary responses.

A

-Antibodies produced during primary virus infections are usually of lower affinity than those produced later, and are often of the IgM isotype (unswitched).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

List and describe major cell types involved in anti-viral responses.

A

Innate: phagocytes, dendritic cells, NK cells, granulocytes

Adaptive: T and B cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

Compare the efficacy of antibody versus cell-mediated immunity in the anti-viral response.

A

All viruses multiply in the cytoplasm of infected cells. Once inside cells, viruses are not accessible to antibodies and can be eliminated only by the destruction of the infected cells on which they depend. This role in host defense is fulfilled by cytotoxic CD8 T cells. The elimination of infected cells without the destruction of healthy tissue requires the cytotoxic mechanisms of CD8 T cells to be both powerful and accurately targeted.

However, when outside the cells, antibodies can act as effector molecules

  • IgA (secretory Ab) inhibits virion/host attachment, neutralizes toxins and enzymes
  • IgG inhibits fusion of enveloped viruses with host membranes
  • IgG and IgM opsonize virions to enhance phagocytosis
  • IgM can coat and agglutinate some virions
  • IgG and IgM can facilitate complement lysis of enveloped viruses
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Explain means of virus evasion/manipulation of host defenses by various viruses.

A

A. Antigenic variation- mechanisms include point mutations in antigenic drift (e.g. HIV, influenza A) and genome shuffling in antigenic shift (e.g. influenza A)

B. Inhibition of the IFN pathway (e.g. influenza NS1).

C. Inhibition of apoptosis and cell cycle control (e.g. SV40 large T Ag and Adenovirus
E1A)

D. Immune tolerance (molecular mimicry or infection prior to competent immune system)

E. Infection of immunoprivileged sites such as the brain (e.g. HSV)

F. Direct infection of the immune system (e.g. HIV and EBV)

G. Restricted expression of viral genes- going invisible to the host defenses as in latent infections (e.g. HIV)

H. Production of viral molecules that act as inhibitors or decoys of host defense molecules such as TLRs, cytokines, receptors, and Ab’s (e.g. Pox and Herpesviruses)

I. Down-regulation of host proteins such as MHC class I or adhesion molecules (e.g. Pox and Herpesviruses)

Viruses can evade capture and death by:
having antigenic variation, mimicking host proteins, restricting the expresion of their genes, producing molecules that act as decoys or inhibitors of host defense molecules, down-regulating host proteins like MHC’s, infecting immunoprivelaged sites (brain), infecting the immune system, and inhibiting apoptosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

For the following virus
Herpes simplex type 1 (HSV1)
be able to discuss:
a.Type of virus (DNA vs. RNA, double strand versus single strand);
b.Cells targeted for primary infection and latency;
c.Transmission and incubation period;
d.Disease entities and their clinical presentations;
e.Diagnosis and key diagnostic tests;
f.Treatments;
g.Prophylaxis-including vaccines.

A

a. DS DNA, protected by isocadedral capsid, alpha

b.Cells targeted for primary infection: mucosal epithelium
and latency: neuron (ganglia)

c.Transmission: close contact, penetrates through mucosal surfaces or cracks in the skin
incubation period: 2-12 days, average is 4

d. Disease entities and their clinical presentations:
- orofacial lesions and (sometimes) genital lesions, encephalitis, herpes whitlow, herpes keratitis, neonatal herpes
- most common presentation is painful mucocutaneous ulcers (orofacial, ocular)
- “above the belt”

e. Diagnosis and key diagnostic tests;
- viral culture, direct fluorescent antibody stain of lesions, PCR of lesions

f. Treatments;
- nucleoside drugs used to treat, take advantage of activation of drug by thymidine kinase (coded by HSV)
- acycloguanosine (acyclovir) and other similar agents
- IV acyclovir: treat severe patients, neonatal, immunocompromised, encephalitis, meningitis patients
- oral antiviral for oral or genital HSV outbreaks

g. Prophylaxis-including vaccines.
- in compliant patients suppressive therapy considered for frequent painful oral/genital herpes
- must take 2x’s a day and not miss days

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

For the following virus
Herpes simplex type 2 (HSV2)
be able to discuss:
a.Type of virus (DNA vs. RNA, double strand versus single strand);
b.Cells targeted for primary infection and latency;
c.Transmission and incubation period;
d.Disease entities and their clinical presentations;
e.Diagnosis and key diagnostic tests;
f.Treatments;
g.Prophylaxis-including vaccines.

A

a. DS DNA, protected by isocadedral capsid, alpha

b. Cells targeted for primary infection: mucosal epithelium
latency: neuron (ganglia)

c.Transmission: close contact, usually sexually. penetrates through mucosal membranes or cracks in the skin
incubation period; 2-12 days, usually 4

d. Disease entities and their clinical presentations;
- genital lesions and (sometimes) orofacial lesions, encephalitis, herpes whitlow, herpes keratitis, neonatal herpes
- “below the belt”

e. Diagnosis and key diagnostic tests;
- most people with it have not been diagnosed, 50% or more of new cases are asymptomatic/unrecongized
- can often be diagnosed clinically, but always must make definitive diagnosis
- viral culture, direct fluorescent antibody stain of lesions, PCR of lesions

f. Treatments;
- nucleoside drugs used to treat, take advantage of activation of drug by thymidine kinase (coded by HSV)
- acycloguanosine (acyclovir) and other similar agents
- IV acyclovir: treat severe patients, neonatal, immunocompromised, encephalitis, meningitis patients
- oral antiviral for oral or genital HSV outbreaks

g. Prophylaxis-including vaccines.
- in compliant patients suppressive therapy considered for frequent painful oral/genital herpes
- must take 2x’s a day and not miss days
- used in patients without stable partner or who are sexually active

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

For the following virus
Varicella zoster virus (VZV)
be able to discuss:
a.Type of virus (DNA vs. RNA, double strand versus single strand);
b.Cells targeted for primary infection and latency;
c.Transmission and incubation period;
d.Disease entities and their clinical presentations;
e.Diagnosis and key diagnostic tests;
f.Treatments;
g.Prophylaxis-including vaccines.

A

a.DS DNA, protected by isocadedral capsid, alpha

b. Cells targeted for primary infection: mucosal epithelium. enters via respiratory tract, spreads to lymph nodes, then replicates in liver, spleen, spreads to particles of skin
latency: neuron (ganglia)

c.Transmission: contact or respiratory route
incubation period: 10-21 days

d. Disease entities and their clinical presentations;
- chickenpox, shingles (zoster)
- starts with fever, malaise, headache, cough
- rash in “crops”/waves
- lesions start as flat, become “dew drop on rose petal” (vesicle with erythematousus base)
- rash on face/trunk, spreads to extremities
- vesicles become pustules then rupture and scab. (usually you find various stages in a patient)
- takes about 7 days for the all lesions to scab, then they are not infectious
- adolescents/adults are higher risk
- pregnancy= risk of varicella pneumonia and death

e. Diagnosis and key diagnostic tests;
- clinically is done typically
- if unclear, can do: direct fluorescent antibody (swab lesion to look for antigens), VZV PCR, viral culture (HSV grows quickly, VZV grows slowly)

f. Treatments;
- self limited in children, doesn’t mandate therapy
- if antivirals given in first 48-72 hours, course is shortened
- acyclovir ot outpatients or IV if very sick
- immunocompromised, pregnant, or severe cases should be treated
- teenagers and adults= high rrisk
g. Prophylaxis-including vaccines.
- live attenuated vaccine (varivax) is recommended for prevention
- given to children as a 2 dose series via subQ injection
- initial dose 12-15 months, booster 4-6years
- can cause disease in immunocompromised
- for shingles, treat with acyclovir in 48-72 hours of onset to lesson lesions and pain
- if post-herpetic neuralgia occurs, you need to do pain control
- live attenuated vaccine for shingles but it has 14x’s more than the chickenpox dose (Zostavax)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

For the following virus Cytomegalovirus (CMV)]
be able to discuss:
a.Type of virus (DNA vs. RNA, double strand versus single strand);
b.Cells targeted for primary infection and latency;
c.Transmission and incubation period;
d.Disease entities and their clinical presentations;
e.Diagnosis and key diagnostic tests;
f.Treatments;
g.Prophylaxis-including vaccines.

A

a.DS DNA, protected by isocadedral capsid, beta

b. Cells targeted for primary infection: epithelia, monocytes, lymphocytes, others
latency: monocytes, lymphocytes, others

c.Transmission: contact, blood transfusions, transplant, congenital
incubation period: 2 weeks-2 months

d. Disease entities and their clinical presentations;
- infectious mononucleosis like syndrome
- in immunocompromised: renitis, pneumonia, colitis
- in newborns: congenital CMV
- common pathogen in 80% of the population
- if pregnant woman develops: 3-5% chance baby born with congenital CMV infection (low birth weight, microcephaly, hearing loss, mental impairment, hepatosplenomegaly, rash, jaundice, chorioretinitis)

e. Diagnosis and key diagnostic tests;
- viral culture, PCR, fluorescent antibody staining, serology, histology

f. Treatments;
- treatment in people with normal immune systems isn’t indicated
- immunocompromised people are treated with antiviral Ganciclovir
- CMV-IG with ganciclovier are used for CMV with pneumonia

g. Prophylaxis-including vaccines.
- no vaccine
- CMV-IG given IV once monthly to immunocompromised patients at high risk

29
Q
Describe the virion structure and replication cycle of herpesviruses.
Structure
Entry
Replication
Assembly and Egress
A

-herpisviridae family are all DS DNA in an isocahedral capsid.
-nucleocapsid = genome + capsid
-surrounded by glycoprotein rich envelope. entire virus particle is a virion
-3 subfamilies based on latency
alpha-sensory ganglia- HSV1,2 VZV
beta-monocytes and lymphocytes-CMV, HHV6, 7
gamma-B cells-EBV, KSHV

Entry:

  • attach and bind to proteins (receptors) found on host cells, and the membranes fuse
  • nucleocapsid is released into cytoplasm, transported to nucleus
  • genome enters through nuclear pores

Replication:

  • Immediate Early (IE) genes are brought in with the virus and are expressed prior to protein synthesis, and are required for the expression of early and late genes
  • E and L genes are de novo synthesized
  • IE genes contain: transcriptional activators
  • E genes encode: proteins involved in DNA replication (viral DNA polymerase, TK, helicase, etc)
  • drug targets often hit these virally encoded proteins, e.g.: ganciclovir acts on TK, foscarnet on DNA polymerase
  • L genes encode: structural proteins like capsids/glycoproteins

Assembly and Egress:

  • assembly in the nucleus, capsids assemble and packaged with DNA
  • bud through the membrane to get envelope as they pass through the Golgi
  • leave the cell through exocytosis or lysis
30
Q

Recognize primary infections, and the concepts of latency and reactivation.

A

Primary Infections:

  • first infections ever
  • no serum antibody is present when symptoms appear
  • most are not symptomatic, but if symptoms occur the disease is likely to be more severe in recurrences
  • common primary infections: HSV 1 and 2 (HSV1 usually occurs in childhood), gingivostomatitis, herpetic whitlow, genital herpes, herpes keratitis, encephalitis, neonatal herpes

Latency:

  • period of time where virus is dormant or silent
  • after primary infection, the virus spreads, enters the nucleus and persists indefinitely in a latent state

Reactivation:

  • when the virus emerges form latency and begins to replicate again.
  • reactivation and shedding of virus without symptoms is a major way things can be transmitted without knowledge (e.g. genital herpes)
  • common reactivations: herpes labials, keratitis, recurrent genital herpes
31
Q
Explain the clinical manifestations of primary infection and reactivation for each of herpesviruses.
HSV1
HSV2
Gingivostomatitis
Herpetic whitlow
Genital herpes
Herpes Keratitis
Encephalitis
Neonatal Herpes
Keratitis
A

HSV1
primary infection: childhood, asymptomatic
reactivation: virus emerges from latency and replicates. travels from infected ganglion down axons to cause recurrence-lesions. Herpes Labilis (common and self limited)

HSV2
primary infection:
reactivation:

Gingivostomatitis
primary infection: occurs with HSV-1. mouth vesicles and ulcerations of gums, lips, tongue in anterior part of mouth, fever, drooling
reactivation:

Herpetic Whitlow
primary infection: inoculation of HSV from oral secretions onto fingers. erythema, swelling, grouped vesicles, pustules at site of entry. oral secretions onto a finger
reactivation:

Genital herpes
primary infection: HSV 2 mostly. commonly asymptomatic. can have painful vesicles, ulcers genital/perianal region
reactivation: lesions =less in number and heal quicker.

Herpes Keratitis (can be primary or reactivation)
primary infection: HSV infects cornea of eye, most common with HSV1. dendritic lesions of the cornea. Pain
reactivation:

Encephalitis (can be primary or reactivation)
primary infection: fulminant and hemorrhagic, necrotizing encephalitis. temporal lobes. 30% mortality. 1=childhood and adult. 2=neonatal
reactivation: neuronal spread of virus occurs in retrograde fashion to brain through trigeminal tract. presents similar to primary infection.

Neonatal Herpes
primary infection: typically diagnosed in mothers with no known history of herpes infection. 3 forms: 1) SEM (skin, eye, mucosa), 2) CNS, 3) disseminated (most severe)
reactivation:

Keratitis
primary infection: infection of any of the 3 branches of cranial nerve 5. latent infection of nerve cells in trigeminal nerve
reactivation: migrates down the nerve axon to cause corneal disease. internal spread of HSV can cause this too. lesions are dendritic appearance-one of the leading causes of blindness in US

32
Q

Identify which herpesviruses have asymptomatic shedding associated with reactivation.

(there are 4)

A
  • asymptomatic reactivation with shedding is a common event, there may be no lesions and only viral shedding may occur
  • viral shedding may occur 1-3 days before the development of a symptomatic outbreak

HSV 1 and 2 (virus shed from mucous membranes and is transmissible, major way genital herpes its transmitted unknowingly)

Recurrent Genital Herpes (may reactivate and shed HSV virus from mucous membranes, anus, perineum. Most patients shed virus asymptomatically even if outbreaks disappear. how 70% of new infections happen)

CMV (virons appear in urine and saliva, can lead to vertical transmission but more to mom and baby. normal immune system=asymptomatic)

**note: VZV is the ONLY herpesvirus that doesn’t exhibit asymptomatic viral shedding in normal hosts that experience reactivation
other members of the herpesvirus family may manifest reactivation ONLY by asymptomatic viral shedding

33
Q

List the 6 complications of chickenpox.

A
  • many complications are potentially fatal
    1. secondary infection or cellulits (group A strep is the most common secondary skin infection causer)
    2. Pneumonia (bacterial is second most common complication due to Strep group A) ( Viral VZV in immunocompromised or pregnant)
    3. Necrotizing fasciitis (infection goes into the fascia, progresses along these planes, shears off nerve endings and blood supply, usually due to Strep A)
    4. Encephalitis (infection with spread to CNS)
    5. Hepatitis (mild elevations of liver function tests, severe is only in immunocompromised)
    6. Congenital Varicella syndrome (contracted by pregnant in 8-20 week range, fetus has multiple tissue/organ abnormalities)
34
Q

Discuss the importance of T-cell mediated immunity to VZV infection.

A
  • Virus reactivation occurs because the virus remains in the ganglia for the latent period, which is often lifelong for people
  • reactivation means shingles
  • once it reactivates it travels down the sensory nerve to an area of skin the nerve innervates
  • lowered cell mediated immunity to variable is the critical factor for risk of shingles
  • more common in elderly due to declining cell mediated immunity (T cells) in old age
  • declining cell mediated immunity may be due to less exposure since most kiddos are vaccinated-exposures used to boost the immunity in elderly regularly
35
Q

Explain the consequences of maternal herpesvirus infection during pregnancy including the risk of the infant developing neonatal HSV, or congenital VZV and CMV syndromes.

A

Neonatal HSV:

  • can be acquired in utero, peripartum, postpartum
  • majority occur in peripartum period, like during birth when exposed to maternal secretions with active HSV 1 or 2.
  • maternal infections can be asymptomatic, so it can pass
  • 3 forms: 1) skin, eye, mucous membrane (SEM), 2) CNS, 3) disseminated (most severe)
  • if a vesicle is found at
36
Q

Describe and be able to recognize how CMV can be diagnosed histologically in infected tissues.

A
  • can do a histology diagnosis for those with organ involvement, cells have “owl’s eye” appearance (showing intranuclear inclusions)
  • tissue biopsy done to look for these characteristic CMV infection cells
  • intranuclear inclusions are accumulations of proteins and virons
  • there may be smaller intracytoplasmic inclusions

** add in photo from notes

37
Q

Describe how to interpret serology (IgM and IgG tests) in the diagnosis of CMV disease.

A

-for primary CMV disease serology establishes the illness, can distinguish btw. primary and recurrent infection,
-primary has CVM IgM and negative CVM IgG
positive IgM
-negative IgG=acute CMV
negative IgM, negative IgG=never had CMV
negative IgM, positive IgG=previously infected at some point
both are positive=recent reaction

38
Q

For Influenza, list the basic structure, important proteins, and describe the roles they play in pathogenesis.

A

Basic Structure:

  • peak in october through may
  • RNA virus with segmented genome
  • 8 different pieces of single strand RNA with several different viral proteins
  • core surrounded by lipid envelope with lining of matrix proteins on inner side
  • 3 types of influenza virus, A and B strains
  • named by type, geographic origin, strain number, year isolation, virus subtype: A/Moscow/21/99 (H3N2)

Important Proteins:

  • hemagglutinin (H) and neuraminidase (N) glycoproteins
  • both are surface proteins
  • different types are designated by numbers: H1, H2, N1, N2, etc.
  • viral subtypes are identified by combination of H and N proteins on viral coat (H1N1, etc)

Roles they play in pathogenesis:

  • A types are cause of epidemics and pandemics
  • A types can infect animals (horses, seals, swine, birds, etc)
  • repeated epidemics happen because A and B types undergo constant and rapid change due to antigenic drift
39
Q

Describe how Influenza causes disease - including outlining routes of viral entry and cells/organs infected, mechanisms of infection, and the consequences of infection on clinical disease presentation.

A
  • flu is transmitted primarily by respiratory route: small droplets/particles are aerosolized (cough/sneeze) and spread
  • also read by contamination on hands or inanimate objects (fomites)
  • viruses live on human hands for 5 minutes, plastic/steel 24-48 hours, cloth/paper 8-12
  • incubation is about 1-3 days
  • acute onset: fever, chills, myalgia, headache, cough
  • presentation varies with age (neonates=bacterial sepsis, apnea) (infants/toddlers=gastrointestinal symptoms, fever, anorexia, respiratory symptoms)(older children/adults=onset rapid with high fever, cough,chills, malaise, other)
40
Q

Describe transmission and effective prevention strategies for Influenza.

A

-transmitted by respiratory route

Treat with antivirals:

1) matrix protein inhibitors: only work on susceptible type A virus (amantadine, rimantadine)
2) neuraminidase inhibitors: effective for susceptible viruses of A and B (oseltamivir, zanamivir, peramivir)
- note: resistance to antivirals is increasing. 2006 CDC announced almost all A strains were resistante to amantidine/rimantidien
- H1N1 was resistant to oseltamivir
- problem is that zanamivir is not approved for people

41
Q

Outline basic principles of vaccine strategies, including listing the different stages of vaccine development for Influenza, the differences between the types of influenza vaccines, and the differences in populations in terms of who can and cannot receive certain types of vaccine.

A
  • 2 types of seasonal flu vaccines:
    1. inactivated influenza-all people 6months or older
    2. live attenuated-people 2-49 years old
  • influenza vaccines contain the same 3 strains of influenza and quadrivalent have the extra B strain
  • reformulated annually to provide protection fro what is circulating in the following year
  • every year after flu season, it is decided what 3-4 will be likely to circulate next year (based on what’s going on in the Southern Hemisphere)
  • 2 A and 1 B are selected for trivalent and one extra B for quadrivalent
  • guess work
  • well matched years: predominant circulating strain is in the vaccine
  • new strains are not in the vaccine-mismatched years

-efficacy declines in elderly, but if vaccinated have a 50% reduction in hospitalization risk or death

42
Q

Describe antigenic drift (point mutation) and antigenic shift (reassortment of genome segments) of influenza and recognize that antigenic drift does NOT change the subtype of the virus (i.e. H2N2).

A

antigenic drift:

  • gradual change in virus that occurs through a slow series of mutations, substitutions, deletions in AA constituting hemagglutinin or neuraminidase surface antigens
  • occurs only after viral strain has become established in humans
  • shows adaptation to host antibody development
  • prevail for 2-5 years then are replaced by next strain
  • gradual evolution

antigenic shift:

  • occurs when type A (and ONLY type A) with a completely new H or N gene segment is introduced into the human population
  • usually acquired from other species (bird, swine, etc.)
  • gene reassortment occurs when 2 or more flu viruses infect 1 human or animal
  • has segmented genome so “swapping” occurs between them
  • if pig infected it can then infect pigs, birds and humans (unique), so its a site where rearrangements between human and avian strains-“mixing vessel”
  • occurs less frequently but is more dramatic.
  • H1N1 is a result
  • influenza type B are NOT subject to antigenic shift as they infect only humans
43
Q

Describe the origin, as well as the basic epidemiology and morbidity/mortality of Pandemic H1N1 influenza virus, recognizing that it has caused a human pandemic.

A
  • could have arisen from incubation in pig
    1) pig infected with 2 different flu strains of different species (human, pig, or bird) at the same time
    2) strains infect the same cell
    3) genes reassort between the strains
    4) creates a new predominantly human strain that has a novel H or N from the bird or pig strain
    5) newly formed strain gets passed onto a human host
  • 1957, 1968 influenza pandemics were caused by reassortment of avian flu A virus
  • swine influenza was the result of multiple reassortments between swine, birds, human strains. had genes from swine flu in pigs, and bird, and human genes
  • lab testing showed most of gene segments were similar to flu viruses in pigs
  • first noted March/April 2009 in Mexico.
  • first typed in CA: so A/California/7/2009 (H1N1)
  • became the dominant circulating flu strain worldwide, called pandemic in June
  • rate was highest among those
44
Q

Discuss newly-emerging influenza viruses including the type (H5N1), including an outline of its geographic occurrence and its importance to public health.

A
  • avian influenza is a disease from birds with influenza type A
  • circulate in birds but rarely kill birds, shed a lot in stool
  • 16 types of H and 9 types of N in wild birds
  • reservoir for influenza

H5N1:

  • in some bird populations that causes severe disease in wild, domesticated birds and can be transmitted to humans
  • outbreak in China=40% mortality in geese
  • highly pathogenic, mainly in birds, can be deadly
  • spreading through eastern europe, more than 20 countries
  • close contact with birds is a risk factor, most people live with/close to their animals allowing for spillover to occur
  • very low in human so far
  • avian flu and human flu have species specificity (due to H binding the receptor, avian binds alpha 2-3 link and humans bind alpha 2-6 link)
  • sometimes they haver receptor specificity for humans-eg: a mutation in H gene that prefers a 2-6 binding.
  • this results in avian strain
  • we fear that it will be like the 1918 strain which killed 50 million people (was avian, we had no immunity)

other way avian flu pandemic could emerge:

  • use pigs as mixing vessel for avian/human strains
  • novels strain could result from mixing that H gene assorts with avian segments so it binds human receptors
45
Q

Describe the necessary qualities in an influenza virus strain to potentially cause a pandemic (new gene segment from another species or ability of a non-human strain to bind to human receptors), stages of a pandemic, and stages leading up to a pandemic.

A

3 conditions must be met to be a pandemic:

  1. emergence of new flu type
  2. virus must infect humans and cause serious illness
  3. virus must have sustained human to human transmission and spread easily among humans
  • Pandemic H1N1 met this
  • H5N1 meets 2 out of 3, only causes disease on limited basis, so there has not been sustained human to human spread (not condition 3)
  • if a pandemic occurs, it would be dealt with by limiting the spread
  • limiting public gatherings, wearing masks, quarantine sick, quarantine zones
46
Q

Explain the effects of tissue tropism, virulence and host responses on the nature of viral disease.

A

Tropism- a given virus is likely to infect certain tissues and not others. Enterotropic viruses replicate in the gut and neurotropic viruses in nervous system tissue. Tropism is determined by a combination of factors: access to the tissue in which it can replicate, receptors required for virus binding and entry, expression of host genes required for virus infection and production of new progeny virus, and a relative failure of host defenses.

Virulence: affect ability of virus to replicate, modify host defense mechanisms, facilitate virus spread among hosts, are directly toxic to host cells

Host factors in susceptibility to viral disease- these factors include expression of appropriate receptors for virus entry, accessibility and permissivity (ability of cells to support virus replication) of infected cells, age of host, genetic background, exposure history, immune status

47
Q

What are pathogenticity and virulence, and how can they be measured?

A

Pathogenicity: refers to the ability of an organism to cause disease (ie, harm the host). This ability represents a genetic component of the pathogen and the overt damage done to the host is a property of the host-pathogen interactions. Commensals and opportunistic pathogens lack this inherent ability to cause disease.

  • frank pathogens: a microorganism capable of producing disease in either normal healthy persons or immunocompromised persons
  • opportunistic pathogens: an organism capable of causing disease, but rarely does so in normal, healthy individual. HIV is an example, also genetic. wouldn’t make most people sick, but immune compromised, folks with genetic defects, folks on immunosuppressants, etc are susceptible to these. Pneumocystis is an example of what happens in HIV patient with AIDS, normal immunity fights it off.

Virulence: if you look at all organisms in a particular taxonomic group, there are differences in relative abilities of these organisms to cause disease (readily vs not). ability of organisms to cause disease (usually used for same family of microbes)

How can these be measured?

  • # of bugs it takes to induce a disease.
  • ID-50: is infective dose that would take to infect 50 subjects
48
Q

Describe how viral genomes are copied, and the first and second principles of template and function of viruses.

A
  • Viral genomes are RNA or DNA and have very diverse structure and composition of genomes
  • mRNA has a translatable open reading from is always (+) strand, ribosome ready, and able to be translated into a protein
  • complementary sequence is (-) strand
  • DNA copied from mRNA is (-) strand
  • DNA equivalent polarity to the mRNA is also the (+) strand
  • First principle: genomes are the templates for synthesis of progeny genomes, small finite number of nucleic acid copying strategies
  • Second principle: function of genomes in the cell is to make RNA. all viruses are parasites of the host cells mRNA translation system. all viral genomes must provide mechanisms for mRNA synthesis
49
Q

Describe the typical, generalized replication cycle of a virus.
Assembly
Egress

A

Assembly:

  • for icosahedral capsids packaging into capsid can be done by: assembling around genome, genome is fed into capsid
  • for helical nucleocapsids: genome coated with nucleocapsid protein during synthesis
  • assembly results in CPE (cytopathic effect) because the capsids are produced at high levels. can result in appearance of cytoplasmic/nuclear inclusions detectable by light microscopy.
  • size and location of inclusions is highly characteristic of particular virus infections

Egress:

  • exit is different for viruses that have envelope vs. those that don’t
  • viruses with naked capsids often release via lysis
  • viruses that have envelopes bud off: plasma membrane to EC environment directly, bud into Golgi or ER, secreted
50
Q

One step growth curve and the latent period-describe these for me.

A

One step growth curve: experiment done in cultured cells were all cells are infected

  • don’t grow exponentially
  • virus is released as burst due to fact that they are formed from preformed components
  • 0-12 hours after adsorption is where no infection detectable inside/outside of cell (eclipse period). time where viruses are broken down and release genome
  • no longer infectious and can’t be detected as plaque forming unit (PFU)
  • latent period: time it takes fro initiation of infection to release of new infectious particles from the cell, 16 hours (adenovirus)

what happens in latent period?

  • attachment of virus to cell
  • entry of virus into cell and uncoating
  • viral gene expression
  • viral genome replication
  • assembly of new virus, and egress of new virus particle
51
Q

Describe attachment, entry, and uncoating of viruses for me.

A

Attachment: involves specific binding of virus-attachedment protein with cellular receptor molecule.

  • target receptor molecules may be proteins, carbs
  • carb receptors are less specific than protein because same configuration of protein may occur on diff. glycosylated membrane bound molecules
  • usually to epithelial cells and mucosal surfaces
  • 2 step process

Entry: penetration occurs short time after attachment of virus to receptor in cell membrane.

  • energy dependent process
  • may involve: endocytosis, fusion of virus envelope with membrane

Uncoating: term for events where after viral particle has entered cell where virus capsid is completely/partially removed and genome is exposed

52
Q

Tell me abut susceptible, resistant, permissive, susceptible and permissive cells

A
  1. A susceptible cell has a functional receptor for a
    given virus–the cell may or may not be able to
    complete the replication cycle
  2. A resistant cell has no receptor–it may or may not
    be competent otherwise to replicate the virus.
  3. A permissive cell has the capacity to replicate
    virus–it may or may not be susceptible
  4. A susceptible AND permissive cell is the only cell
    that can take up virus and replicate it
53
Q
Tell me about gene expression strategies:
DNA viruses
RNA  viruses
\+ stranded RNA viruses
- stranded RNA viruses
Retroviruses
A

DNA viruses: Double stranded, gapped genomes or single stranded genomes must transcribe mRNA using the (-) strand of the DNA genome as a template. gaps must be filled before genes can be transcribed. RNA pol 2 dies this, in nucleus

RNA viruses: make use of a unique viral enzyme known as RNA-dependent RNA polymerase (RdRp), which is not found in host cells. This enzyme is used for both the production of mRNA and the replication of RNA genomes

+ Strand RNA viruses: can be translated directly by cellular ribosomes. Amplification of mRNA copy number or production of sub-genomic mRNAs is mediated by RdRp.

  • Strand RNA viruses and DS RNA viruses: (+)sense mRNA must be transcribed from the genome in order to have gene expression. Because animal cells do not contain this enzymatic function and the genome is not “ribosome-ready”, these viruses must bring this enzyme with them into the infected cell. Thus, (-) stranded RNA viruses package a RdRp in the virus particle.

Retroviruses: accomplished by an enzyme packaged into the virus particle called reverse transcriptase. Animal cells do not contain an enzyme that can perform this function. The double-stranded DNA copy of the virus genome then integrates into the host cells DNA. Once integrated, mRNA is transcribed from the virus genome using host cell-encoded RNA polymerase II

54
Q
Briefly describe the following infections:
lytic
persistent 
latent
transformation
A

Lytic infections result in the destruction of the host cell.

Persistent infections include enveloped viruses that are released by budding (without lysis of the cell) and may occur over relatively long periods of time.

Latent infections show a delay between the infection and appearance of symptoms.

Transformation can be caused by some viruses, conferring cell growth without restraint. These are called oncogenic viruses.

55
Q

Tell me about Interferons, the types, and what they are produced by.

A

~ Infection of cells with viruses induces the production of proteins known as interferons, so called because they were found to interfere with viral infection of neighboring cells.

1. Type I IFNs (include IFNa and IFNb) - antiviral cytokines transiently produced and secreted by most infected cells within hours of infection.
Type I (a/b) IFNs regulate control genes through ISREs (interferon-stimulated response elements).
2. Type II IFN (IFNg) is produced mainly by T cells and NK cells, more restricted than Type I IFN production.
Type II (g) IFN regulate control genes through GAS (gamma activated site) elements.

*Type I (a/b) and Type II (g) IFNs are distinct! They are produced by different cells and at different stages of infection!

Cells respond to IFN through receptor activation of Jak/Stat signaling.

56
Q

Tell me about the steps of pathogenesis of virus diseases.

A

Route of infection: primary initial are respiratory, alimentary, genitourinary

Initial site of replication: often epithelial cells near body surface or barriers. must have sufficient virus

Spread from initial site: some viruses remain local, others spread to tissues causing disseminated to systemic infection. must breach local barriers.

57
Q

What is tropism?

A
  • give virus is likely to infect certain tissues and not others.
  • enterotropic viruses replicate in gut and neurotropic viruses in nervous system tissue
  • determined by combination of factors
  • can drive population variants in individuals
58
Q

Tell me about transmission and shedding

A
  • release of infectious particles from infected host.
  • local infectors shed at site of initial infection
  • for systemic/disseminated it occurs in multiple or distant sites. may be in single host or alternate infection of hosts with another
  • enveloped viruses are fragile and sensitive to environmental stresses
  • most transmission is horizontal, but can occur from parent to child in germ line
  • some viruses show geographic patterns
59
Q

Discuss the varicella and shingles vaccines (who we do and do not give the vaccine to).

A

Varicella Vaccine:

  • recommended as part of routine immunizations
  • given at 12-15 months, and 4-6 years old
  • you do not give this vaccine to immunocompromised (since it is a live vaccine)

Shingles Vaccine:

  • same live attenuated virus as chickenpox but with 14x’s more virus
  • boost response to VZV and decreases likelihood of getting shingles
  • 50 years old or older, given and 1 dose
60
Q

For RSV, list the basic structure, important proteins, and describe the roles they play in pathogenesis.

A

-causes significant lower respiratory tract infection in all ages, but mostly in young children

Basic Structure: single strand, non segmented RNA

  • 2 types, A and B
  • A is more severe, more prevalent

Important Proteins:

  • F protein: fuses viral envelope to host cell, fusion of membranes of infected cells to cause “syncytia”
  • G protein: initial binding to host cell

Roles they play in pathogenesis:

  • occurs worldwide
  • both subtypes, A and B, circulate in 1 season, can alternate
  • most common cause of bronchitis
61
Q

Describe how RSV cause disease - including outlining routes of viral entry and cells/organs infected, mechanisms of infection, and the consequences of infection on clinical disease presentation.

A

Routes of viral entry: invades conductive, nasopharynx

Cells/organs infected:
-lungs, bronchioles

mechanisms infection:

  • large droplet spread
  • lives on surfaces 1 hour

clinical presentation:

  • 3-5 day incubation
  • syncytia on cellular level
  • constricts smooth muscles in bronchioles to produce:
    a) edema/inflammation of airway;
    b) ventilation/perfusion mismatch-hypoxia
    c) hyper expansion by mucous plugging (seen on x ray)
  • seen as: respiratory distress, wheezing, cough, rhonchi on auscultation, hypoxia, copious secretions

Testing:
-cell culture, direct antigen detection, PCR

62
Q

Describe transmission and effective prevention strategies for RSV.

A

Transmission: droplet, surface

Prevention Strategies:

  • vaccine in 60’s: formalin inactivated RSV. did not produce immunity, and those immunized got worse disease
  • new vaccines on horizon
  • immunophrophyaxis with palivizumaub and motavizumab
  • palvizumaub: human pooled antibody with high RS titers
  • motavizumab: developed from pal, but higher affinity binding F protein
63
Q

Outline basic principles of vaccine strategies, including listing the different stages of vaccine development for RSV, and the differences in populations in terms of who can and cannot receive certain types of vaccine.

A
  • people get recurrent infections
  • antibody/immune protection is incomplete for infection, but may prevent a more severe disease
  • vaccine in the 1960’s, formalin inactivated. showed risk of ‘priming’ for an infection without developing immunity (occurs with dengue)
  • RSV immunophrophylaxis with palivizumab and motavizumab
  • palivizumab: mouse model anti-RSV antibody developed: PALAVISUMAB developed in 1988. once monthly IM injection in RSV season for high risk groups of young children
  • Motavizumab: developed from pal. appears non-inferior to pal. need more studies. expensive
64
Q

For Inhibition of viral neuraminidase (oseltamivir, zanamivir, peramivir), describe the:
mechanism of action,
pharmacokinetic properties,
adverse drug reactions,
and role of pharmacotherapy in treatment of viral infections.

A

Mechanism of action:

  • inhibition of enzyme neuraminidase (NA)
  • NA: cleaves N-acetyl nueraminic/sialic acid from cell receptors for A and B flu.
  • egress of new irons results from tethering to plasma membrane via H and sialic acid
  • without NA, virus aggregates at cell surface, and can’t bud or translocate, so infectivity is reduced
  • resistance =low

Pharmacokinetics:

  • Oseltamivir: oral, prodrug, eliminated renally, half life=6-10 hours, given 2x’s /day for 5 days
  • Zanamivir: inhalation 2x’s per day for 5 days, renal elimination
  • Peramivir: single IV dose for ill or immunocompromised

Adverse Drug Reactions:

  • Oseltamivir: minor nausea/vomiting
  • Zanamivir: bronchospasm (uncommonly) with COPD or asthma
  • Peramivir: diarrhea, GI effects, neutropenia, rare skin reactions

Pharmacotherapy Role in treatment:

  • for flu, if started in 48 hours may decrease duration and severity by 1-2 days of A or B
  • effective as prophylactic measure
  • indicated for use in high risk ppl, institutional outbreaks
65
Q

For Inhibition of uncoating (amatadine, rimatadine), describe the:
mechanism of action,
pharmacokinetic properties,
adverse drug reactions,
and role of pharmacotherapy in treatment of viral infections.

A

Mechanism of action:

  • blocks virally encoded H+ ion channel (M2 protein) to prevent intracellular pH changes needed for uncaring
  • resistance occurs in response to mutations of transmembrane domains of M2 proton channel

Pharmacokinetics:

  • effective orally-accumulate in lungs
  • Amatadine: excreted unchanged in urine (90%) so dosage adjust if impaired renal function
  • Rimatadine: hepatic elimination (half life 12 hours, 1-2 daily dose)
  • excreted breast milk, so not recommended if breast feeding

Adverse Drug Reactions:

  • amatadine: insomnia, concentration difficulty, lightheadedness/dizzy, headache
  • rimatadine: tolerated due to poor CNS penetration
  • teratogenic in animals, category C in pregnancy

Pharmacotherapy Role in treatment:

  • for prophylaxis in flu A
  • given 2-3 weeks in conjunction with vaccine in high risk pop
  • if given 1-2 days before and 6-7 days during reduces severity/incidence
  • most H3N2 and H1N1 are resistant
66
Q

For Inhibition of viral genome replication, describe the:
mechanism of action,
pharmacokinetic properties,
adverse drug reactions,
and role of pharmacotherapy in treatment of viral infections.

A

Mechanism of action:

  • acyclovir (zorivax), valacyclovir (valtrex), peniciclovir (denavir), famciclovir (famvir)
  • thymidine kinase does initial phosphorylation-primary mechanism of viral vs. host selectivity
  • acyclovir TP binds with greater affinity to viral DNA than host
  • protein kinases convert acyclovir-MP to TP form. TP competes with cellular dGTP for viral DNA polymerase, then incorporates into replicating viral DNA. once incorporated, acyclovir TP terminates DNA replication/strand elongation. DNA containing acyclovir TP binds and inactivates DNa polymerase
  • resistance=due to reduced/loss of TK expression, altered viral TK substrate, altered affinity of viral DNA polymerase activity

Pharmacokinetics:

  • acyclovir: oral is poor, not affected by food. topical or IV
  • valacyclovir: valyl ester prodrug of acyclovir. given orally, plasma levels are 3-5 times higher than acyclovir (equivalent to IV)
  • peniciclovir: acyclic guanosine analoge. poor oral, topical only
  • famciclovir: peniciclovir prodrug, increases bioavalablity to 70%.
  • vedarabine-trifluridine: topical only (toxic IV)
  • elimination: acyclovir=kidneys, neonatla clearance is 1/3 of adults

Adverse Drug Reactions:

  • headache, nausea, vomiting, renal dysfunction (reversible)
  • IV has been associated with encephalopathy
  • category B pregnancy

Pharmacotherapy Role in treatment:

  • use with HSV
  • oral acyclovir shortens primary and recurrent symptom duration
  • IV acyclovir: treatment for encephalitis HSV, neonatal, serious HSV or VZV infections, esp. immunocompromised
  • topical are limited to use for HSV keratoconjuntivis and epithelial keratitis
  • use with VZV
  • oral acyclovir decreases number lesions and duration at higher dose
  • suppression with oral acyclovir for immunocomporomised
67
Q

For Ribavirin (drugs for respiratory syncytial virus infections), describe the:
mechanism of action,
pharmacokinetic properties,
adverse drug reactions,
and role of pharmacotherapy in treatment of viral infections.

A

Mechanism of action:

  • converted to ribavirin TP by cellular kinases
  • inhibits GTP dependent 5’ capping of viral mRNA
  • may increase mutagenesis
  • no resistance to date

Pharmacokinetics:

  • oral is good (45-64%), increases with fatty food
  • elimination hepatic and renal of unchanged drug
  • half live varies: inhalation, children=6-11 hrs; oral, adults=43 hours, increases to 150

Adverse Drug Reactions:

  • inhalation: well tolerated. can cause conjunctival/bronchial irritation
  • acute deterioration in those with bronchospastic lung disease
  • oral: high incidence hemolytic anemia. with interferon=more cough, pruritus, rash
  • teratogenic, should not be given in pregnancy, category X

Pharmacotherapy Role in treatment:

  • respiratory syncitial virus: arosolized ribavirin effective against RSV, pneumonia
  • palivizumab is monoclonal antibody to RSV F glycoprotein that is indicated for RSV immunoprophylaxis for infants/young children. given IM monthly
  • hepatitis C: oral combined with interferon alpha2a or alpha2b. replaced by directing antiviral agents
68
Q

For Gangiclovir (drug for cytomegalovirus infection), describe the:
mechanism of action,
pharmacokinetic properties,
adverse drug reactions,
and role of pharmacotherapy in treatment of viral infections.

A

Mechanism of action:

  • cellular uptake and initial phosphorylation mediate by PK UL97 in CMV
  • primary mechanism of host vs. viral selectivity
  • PKs convert granciclovir-MP to TP form, 10 fold higher in CMV cells
  • ganciclovir-TP competes with cellular dGTP for viral DNA polymerase, incorporates nucleotide into replicating strands
  • slows and ceases elongation
  • resistance: mutations in UL97, and polymerase alter activity

Pharmacokinetics:

  • poor oral bioavailability, good distribution body fluids, so IV
  • valganciclovir prodrug deesterified and converted to this drug by GI and hepatic esterase’s
  • excreted via urine half life is 4 hours

Adverse Drug Reactions:

  • less selective toxicity than acyclovir, since kinases can phosphorylate
  • myelosuppresion with nutropenia and thrombocytopenia is major side effect/concern. can be reversed
  • CNS toxicity (rarely) and abnormal liver function
  • Category C pregnancy

Pharmacotherapy Role in treatment:

  • treatment and suppression of CMV retinitis in immune-compromised
  • effective controlling CMV in transplant patients
  • gel is good for HSV keratitis
  • activity against HBV when oral
69
Q

For Foscarnet (drug for cytomegalovirus infection), describe the:
mechanism of action,
pharmacokinetic properties,
adverse drug reactions,
and role of pharmacotherapy in treatment of viral infections.

A

Mechanism of action:

  • no cellular activation
  • binds noncompetitively to pyrophosphate binding site of RNA and DNA polymerases. Inhibits cleavage of pyrophosphate from deoxy TP
  • blocks viral replication
  • resistance: alterations to DNA polymerase. some strains resistant to both foscarnet and ganciclovir

Pharmacokinetics:

  • oral is poor, given as IV
  • eliminated unchanged in urine
  • half life is bimodal and complex, initial=4-8hurs, terminal is 3-4 days

Adverse Drug Reactions:

  • nephrotoxicity, hypoclcemia, can be severe/fatal
  • CNS abnormalities: headache, tremor, seizures, hallucinations
  • rash, fever, nausea

Pharmacotherapy Role in treatment:

  • effective against CMV retinitis (immunocompromised)
  • effective against ganciclovir resistant CMV infections and acyclovir resistance HSV and VZV infections