+ All Categories
Home > Documents > VIRAL LATENCY

VIRAL LATENCY

Date post: 30-Oct-2014
Category:
Upload: walid-fouad
View: 115 times
Download: 3 times
Share this document with a friend
Description:
A STRATEGY OF IMMUNE EVASION
Popular Tags:
49
WALID NABIL FOUAD Microbiology Department Medical Research Institute Alexandria University June 2010 VIRAL LATENCY A STRATEGY OF IMMUNE EVASION
Transcript
Page 1: VIRAL LATENCY

WALID NABIL FOUADMicrobiology Department

Medical Research InstituteAlexandria University

June 2010

VIRAL LATENCYA STRATEGY OF IMMUNE EVASION

Page 2: VIRAL LATENCY

1. Acute infections: Infections that are of relatively short duration with rapid recovery. Examples include viruses causing: routine respiratory infections (cold viruses and influenza viruses), and gastrointestinal infections (e.g. rotaviruses and noroviruses).

2. Persistent infections: In contrast to acute viral infections, persistent infections last for long periods, and occur when the primary infection is not cleared by the adaptive immune response. The viruses in this case are continually present in the body, and remain in specific cells of infected individuals.

Types of Viral Infections in Humans:

Page 3: VIRAL LATENCY

Persistent infections could be slow, latent, or chronic infections:

A. Slow infections:- Infections characterized by having a long incubational (preclinical) period (months or even years) followed by progressive disease, or having late complications following an acute infection.

- Examples include:

Subacute Sclerosing Panencephalitis that can follow an acute measles infection.

Progressive Encephalitis that can follow rubella infection. Another example of slow infections is those infections caused by

prions (although prions are not considered typical viruses).

Persistent infections:

Page 4: VIRAL LATENCY

B. Latent infections:These Infections are characterized with periodic reactivation, and are a common outcome after acute infection with the Herpes viruses.

Persistent infections:

Herpes family viruses (Herpesviridae):

The most common example of latent viruses is the Herpes virus family, Herpesviridae, all of which establish latent infections. Herpes viruses are the leading cause of human viral disease, second only to influenza and cold viruses, causing overt disease such as cold sores and chickenpox, or they may remain latent for many years to be reactivated in later life, as in shingles. Examples include: HSV-1, HSV-2 and VZV.

Page 5: VIRAL LATENCY
Page 6: VIRAL LATENCY
Page 7: VIRAL LATENCY

C. Chronic infections:In case of chronic virus infections, the virus can be demonstrated in the body at all times and the disease may be present or absent for an extended period of time. Examples include liver hepatitis, cirrhosis, and carcinoma caused by Hepatitis B and C viruses.

Latent vs chronic infections:

Page 8: VIRAL LATENCY

Natural history of acute and persistent human infectionsSolid line = shedding of infectious virus; dashed line = virus not readily demonstrable; +, disease episode.

Page 9: VIRAL LATENCY
Page 10: VIRAL LATENCY

Viral latency is the ability of a pathogenic virus to lie dormant within a cell, usually denoted as the lysogenic part of the viral life cycle.

A latent viral infection is known to be a type of persistent viral infection, and is distinguished from a chronic viral infection.

In a viral latent infection:

1. Virus production cease (despite that the viral genome is maintained intact inside the host cell).

2. No disease symptoms could be seen (asymptomatic).3. Virus remains in equilibrium with the host for long periods of time before

symptoms again appear. 4. Actual viruses could not be detected until reactivation of the disease occurs.

What is viral latency?

Page 11: VIRAL LATENCY

Since the viral genome is not fully eradicated from the body of the host, a latent virus can be reactivated and begin producing large amounts of viral progeny without the host being infected by new outside virus, (denoted as the lytic part of the viral life cycle), and stays within the host indefinitely.

During latency, productive cycle genes are repressed. But it is well known that many triggers can provoke the reactivation of a latent virus. These include:

Fever / common cold Physical trauma / fatigue Psychological stress / emotional disturbances Menstruation / pregnancy / hormonal changes Infection; viral (HIV) or bacterial (pneumococcal and meningococcal) Irradiation; including sunlight and UV Immunosuppression (as in CMV disease)

Latent and lytic cycles:

Page 12: VIRAL LATENCY

Latency is distinguished from lytic infection; in lytic infection many Herpes virus particles are produced and then burst or lyse the host cell.

Lytic infection is sometimes known as "productive" infection.

Latent cells harbor the virus for long time periods, and then occasionally convert to productive infection which may lead to a recurrence of Herpes symptoms.

Lytic cycle:

Page 13: VIRAL LATENCY

Virus latency should not be confused with clinical latency; Clinical latency occurs during the incubation period of an infection, in which the causative agent is present in the body and multiplying, but not causing symptoms. The virus involved in clinical latency is not dormant, as is the case with latent infections, but fully active and causing disease.

Virus latency vs clinical latency:

Page 14: VIRAL LATENCY

Latency in case of Bacteriophages is known as lysogeny.

The lysogenic cycle is one of two phases of viral reproduction (the lytic cycle is the other).

Lysogeny is characterized by integration of the bacteriophage nucleic acid into the host bacterium's genome.

The newly integrated genetic material, called a prophage can be transmitted to daughter cells at each subsequent cell division, and a later event (such as UV radiation) can release it, causing proliferation of new phages via the lytic cycle.

Lysogeny is common to all prokaryotes and may be associated with lysogenic conversion; new host phenotype because of the integrated viral DNA.

Latency in Bacteriophages:

Page 15: VIRAL LATENCY
Page 16: VIRAL LATENCY

An example of a latent virus is HSV which may reactivate from latency from time to time in the ganglia and may spread to peripheral tissues to cause reactivated disease or asymptomatic virus shedding in the cornea, lips, genitalia, or other sites.

Latent HSV-1 infection of the nervous system becomes increasingly prevalent among humans with age and most commonly involve the trigeminal ganglia, whereas involvement of other ganglia of the PNS is somewhat less frequent. HSV-2 primarily establishes latency in sacral ganglia.

Examples of latent viral infections:

Page 17: VIRAL LATENCY

Simian virus 40 (SV40) is a DNA virus that produces a lytic infection in the kidney cells of the African green monkey (these cells are used to cultivate viruses in the lab), but a latent infection in the cells of humans, mice, rats, and hamsters. Like lysogeny in bacteria, the SV40 genome becomes incorporated in the DNA of its host (in chromosome 7 in human cells). Although a human cell with harboring SV40 shows no outward sign of the virus, its presence can be detected by: )1) the appearance of viral-encoded antigens in the host cell, and (2) the ability of these cells to cause a lytic infection in African green monkey cells when fused with them.

In humans, lytic infections of plasma cells by the Epstein-Barr virus (EBV) occur in mononucleosis, whereas latent infections of B cells by EBV predispose the person to lymphoma.

Also lytic infections by the human papillomas virus (HPV) cause genital warts, whereas latent infections by some strains of HPV lead to cervical cancer.

Examples of latent viral infections:

Page 18: VIRAL LATENCY

The most important strategy HSV uses for immune evasion is the establishment of latent infection.

In order to achieve a long-term persistence in the infected host:

1. A virus must avoid killing too many host cells (which would lead to the early death of the host), and

2. It also must avoid elimination by the host immune system.

Latent infection of neurons allows HSV to escape elimination by the immune system and to persist indefinitely in the host.

Viral latency as a mechanism of immune evasion:

Page 19: VIRAL LATENCY

Until quite recently, there appeared to be simple explanation why HSV latency is a successful strategy for viral persistence: it could be argued that suppression of viral replication and a lack of expression of viral genes - associated with the lytic cycle during latency - limits viral cytotoxicity and allows for the survival of the infected neurons for decades, and that because no viral protein expression occurs during latency, the immune system doesn't recognize and therefore cannot eliminate the latently infected cells.

However, there is now accumulating evidence that mechanisms by which HSV achieves long-term persistence in neurons involve a more complex pattern of HSV gene expression than previously thought. Latency is generally maintained by viral genes expressed primarily during the latent period, known as the latency-associated genes.

Viral latency as a mechanism of immune evasion:

Page 20: VIRAL LATENCY

A. Episomal latency:Episomal latency refers to the use of genetic episomes during latency.

An episome is a portion of genetic material that can exist independent of the main body of genetic material (the chromosome).

In this type of latency, viral genes are floating in cytoplasm or nucleus as distinct objects, as linear structure.

Examples of viruses with episomal latency: The chicken-pox virus and herpes simplex viruses (HSV-1, HSV-2), which

establish episomal latency in neurons, and leave linear genetic material floating in the cytoplasm.

Mechanisms of viral latency:

Page 21: VIRAL LATENCY

The Gamma Herpesvirinae subfamily, which is associated with episomal latency established in cells of the immune system, such as B-cells in the case of Epstein-Barr Virus.

- Advantages of episomal latency include the fact that the virus may not need to enter the nucleus, and hence may avoid ND10 (nuclear domains 10) from activating interferon via that pathway.

- Disadvantages include more exposure to cellular defenses, leading to possible degradation of viral gene via cellular enzymes.

Episomal latency is more vulnerable to ribozymes or host foreign gene degradation than provirus latency.

Mechanisms of viral latency:

Page 22: VIRAL LATENCY

B. Proviral latency:Proviral latency occurs when the virus genome integrates into the host genome, effectively becomes a provirus. This requires that the viral gene get into the nucleus and insert itself into the host genome.

The family of which exemplifies this behavior being the retroviruses.

For example, when a retrovirus HIV invades a host cell:

1. The genomic RNA of the retrovirus is transcribed into DNA by the reverse transcriptase, then 2. It become inserted into the host genome by an integrase,

and remains within the hosts own gene.

Mechanisms of viral latency:

Page 23: VIRAL LATENCY

A provirus not only refers to a retrovirus, but is also used to describe other viruses that can integrate into the host chromosomes, another example being adeno-associated virus.

In many cases, retroviruses make a DNA copy of their own viral genome and insert it into their host’s genome.

If this happens to a germ line cell (i.e. the sperm or egg cells), the retroviral DNA will be inherited by descendants of the host.

Mechanisms of viral latency:

It is thought that proviruses may account for approximately 8% of the human genome in the form of inherited endogenous retroviruses.

Page 24: VIRAL LATENCY

- Advantages include an automatic host cell division that results in replication of the viruses' gene.

The provirus does not directly make new DNA copies of itself while integrated into a host genome in this way. Instead, it is passively replicated along with the host genome and passed on to the original cell's offspring; all descendants of the infected cell will also bear proviruses in their genomes.

Another advantage is the fact that it is near impossible to remove an integrated provirus from an infected cell without killing the cell.

- Disadvantages include the need to enter the nucleus (and the need for packaging proteins that will allow for that) and increased difficulty in maintaining the latency.

Mechanisms of viral latency:

Page 25: VIRAL LATENCY

In the field of HIV research, proviral latency in specific long-lived cell types is the basis for the concept of one or more viral reservoirs, referring to locations (cell types or tissues) characterized by persistence of latent virus.

Specifically, the presence of replication-competent HIV in resting CD4-positive T cells, allows this virus to persist for years without evolving despite prolonged exposure to antiretroviral drugs.

This latent reservoir of HIV may explain the inability of anti-retroviral treatment to cure HIV infection.

Latent reservoirs of HIV;An obstacle to the eradication of virus:

Page 26: VIRAL LATENCY

A certain type of latency could be ascribed to the endogenous retroviruses. These viruses have infected human germline cells and have been incorporated into the human genome in the distant past, and are now passed through reproduction.

Endogenous retroviruses can persist in the genome of their host for long periods. Generally these types of viruses have become highly evolved, and have lost the expression of many gene products. Some of the proteins expressed by these viruses have co-evolved with host cells to play important roles in normal processes.

Endogenous retroviruses, however, could be "infectious" after integration if they acquire 'knockout' mutations during host DNA replication. They can also be partially excised from the genome by a process known as recombinational deletion. Some human ERVs have been implicated in certain autoimmune diseases and cancers.

Endogenous Retroviruses (ERVs):

Page 27: VIRAL LATENCY

How do latent viruses maintain their latency?

Both proviral and episomal latency may require maintenance for continued infection and fidelity of viral genes. Latency is generally maintained by viral genes expressed primarily during latency.

Expression of these latency-associated genes may function to keep the viral genome from:

(1) being digested by cellular ribozymes, or (2) being found out by the immune system.

Certain viral gene products (RNA transcripts such as non-coding RNAs and proteins) may also inhibit apoptosis or induce cell growth and division to allow more copies of the infected cell to be produced.

Maintaining Viral Latency:

Page 28: VIRAL LATENCY

In vivo, HSV-1 and HSV-2 infections can be divided into 4 stages:

1. Acute infection2. Establishment of latency3. Maintenance of the latent state, and 4. Reactivation of latent virus

During acute infection, virus replicates at the site of inoculation on epithelial surfaces resulting in primary lesions.

Virus then enters into nerve terminals where it travels by axonal transport to associated sensory ganglia. At this stage, in some animal models there is a short period of virus replication in the ganglia, which can be detected by virus isolation from cell-free homogenates of the ganglia. It is unknown whether this also occurs in humans and it may be an artifact due to the animal species, route of inoculation or the large virus inoculum used in experimental models.

HSV Gene Expression and Latency:

Page 29: VIRAL LATENCY

In the second stage, occurring at approximately 2-4 weeks p.i., a latent infection is established in which viral DNA is maintained in the neurons as an episome and there is limited HSV gene expression. Infectious virus is no longer detected in cell-free homogenates, but it can be detected by explantation and organ culture of the ganglia.

In the last stage, certain stimuli may result in activation of virus replication with concomitant axonal transport of virus progeny to a peripheral site, at or near the portal of entry. Such stimuli include physical or emotional stress or peripheral tissue damage.

In some animal models (guinea pigs), latent virus reactivates spontaneously. In others it can be experimentally reactivated by neurectomy, trauma to the ganglia, electrical stimulation, epinephrine iontophoresis or cadmium treatment.

HSV Gene Expression and Latency:

Page 30: VIRAL LATENCY
Page 31: VIRAL LATENCY

The most common example of latency-associated gene products is the Latency Associated Transcripts (LAT) of the HSV.

LAT is a length of RNA which accumulates in cells hosting long-term, or latent, HHV infections. While these infected host cells would ordinarily undergo an organized death or be removed by the immune system, the consequences of LAT production interfere with these normal processes. LAT is expressed by the latent virus, and functions to:

1. Regulate viral genome.2. Maintain viral latency by interfering with apoptosis through the down regulation of MHC, thus inhibiting the apoptotic pathways.3. LAT are also required for the virus to be reactivated from latency.

A. Maintaining latency in HSV infections; Latency Associated Transcript (LAT):

Page 32: VIRAL LATENCY

In order to maintain a reservoir of latently infected host cells, Herpes virus interferes with apoptosis.

During latency, most of the Herpes DNA is inactive, with the exception of LAT, which accumulates within infected cells. The region of HHV DNA which encodes LAT is known as LAT-DNA. After splicing, LAT is a 2.0-kilobase transcript (or intron) produced from the 8.3-kb LAT-DNA. The DNA region containing LAT-DNA is known as the Latency Associated Transcript Region.

Expression of LAT: (1) reduces the production of proteins involved in the apoptosis mechanism, including proteins caspase-8 and caspase-9, and also (2) results in the suppression of herpes lytic genes.

In rabbit trigeminal ganglia, extensive apoptosis occurred with LAT (-) virus but not with LAT (+) viruses. In addition, a plasmid expressing LAT blocked apoptosis in cultured cells. Thus, LAT promotes neuronal survival after HSV-1 infection by reducing apoptosis.

Maintaining latency through inhibition of apoptosis:

Page 33: VIRAL LATENCY

For a virus to survive, it must elude the ever vigilant immune sentinels of its host. A latent virus can escape immune detection if it resides in non-dividing cells and doesn’t produce any proteins. No viral proteins means no red flags for immune cells. If the virus targets one of the many cell types that rarely divide, it’s relatively safe while latent.

But some viruses, like the gamma-herpes virus, infect B cells of the immune system, which occasionally divide. The gamma-herpes virus genome persists as circular pieces of DNA called episomes. When an infected B cell divides, the latent gamma-herpes virus episome must replicate and segregate into daughter cells along with the cell’s genome. Viral replication and segregation requires the services of a protein called the episome maintenance protein.

B. Maintaining latency in EBV infections; Epstein-Barr nuclear antigen 1 (EBNA-1):

Page 34: VIRAL LATENCY

The episome maintenance protein produced by EBV is known as the Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA-1).

This viral protein is essential for:1. Replication of the episomal EBV DNAs, and2. Maintenance of latency.

Role of EBNA-1 in viral replication: EBNA-1 plays an essential role in replication and partitioning of viral genomic DNA during latent viral infection. During this phase, the circular double-stranded viral DNA undergoes replication once per cell cycle and is efficiently partitioned to the daughter cells. EBNA1 activates the initiation of viral DNA replication through binding to specific sites in the viral latent origin of replication, oriP. Additionally, it governs the segregation of viral episomes by mediating their attachment to host cell metaphase chromosomes.

EBNA-1 and its role in viral replication:

Page 35: VIRAL LATENCY

Although episome maintenance proteins might be a potentially recognizable target for immune cells, it was discovered that EBNA-1 harbors an amino acid element in its epitope (the region that binds to a T cell and triggers an immune response) that helps the viral protein evade the killer T cells that could destroy it. As a result of this, EBNA-1 is not recognized by the cellular immune system.

Lab studies show that the amino acid element limits EBNA-1’s interaction with T cells by inhibiting synthesis and, to a lesser degree, degradation of the protein. This results in:

A. Impairing antigen processing, and B. MHC class I-restricted antigen presentation, thereby inhibiting the CD8-restricted cytotoxic T cell response against virus-infected cells.

How this evasive action works or helps the virus in a living organism is not entirely clear. But if T cells aren’t presented with bits of viral protein, they have no way of knowing the virus is present.

Maintaining latency through evading immune response:

Page 36: VIRAL LATENCY

A- Viral Reactivation:

While viral latency exhibits no active viral shedding nor causes any pathologies or symptoms, the virus is still able to reactivate via external activators (i.e. sunlight, stress) to cause an acute infection.

In case of Herpes simplex virus, which generally infects an individual for life, a serotype of the virus reactivates occasionally to cause cold sores.

The sores are quickly resolved by the immune system, however may be a minor annoyance from time to time.

Latent infections can also become a serious problem when a patient becomes immunocompromised, as the latent infection may manifest when the patient's immune system passes a critical point.

Outcomes of viral latency:

Page 37: VIRAL LATENCY
Page 38: VIRAL LATENCY

A | Infection of epithelial cells in the mucosal surface gives rise to productive replication, resulting in the production of progeny virions, which can spread to infect additional epithelial cells. Virus enters innervating sensory neurons, and nucleocapsids are transported to the neuronal cell body. The viral DNA is released into the neuronal nucleus and circularizes. Circular viral DNA persists in the neuronal cell nucleus, and the latency-associated transcript is expressed.

B | Upon reactivation, viral lytic gene expression is initiated, and newly formed capsids are transported to the axonal termini. Infectious virus is released from the axon and infects epithelial cells, resulting in recurrent infection and virus shedding.

HSV latent and lytic cycle:

Page 39: VIRAL LATENCY
Page 40: VIRAL LATENCY

B- Host Cell Transformation:

A more serious outcome of a latent infection could be the possibility of transforming the host cell, and forcing it into an uncontrolled cell division. This is usually seen in infections with human papilloma virus, in which persistent infection may lead to cervical cancer as a result of cellular transformation.

Although it is unclear how high-risk HPV types cause cancer, studies indicate that malignant transformation involves the viral E6 and E7 gene products, which may exert their effect by interfering with the cellular proteins that regulate cell growth.

Outcomes of viral latency:

Page 41: VIRAL LATENCY

In vitro, cervical epithelial cells with integrated HPV-16 genes multiply faster than those with extra chromosomal (episomal) HPV-16.

An explanation for this is that the expression of viral genes E6 and E7 is increased in cells with where the HPV-16 genome is integrated, and these gene products, oncoproteins E6 and E7, respectively bind and inactivate cell tumor suppressor proteins p53 and pRB.

Outcomes of viral latency:

Page 42: VIRAL LATENCY

Another example is the Epstein-Barr virus (EBV), which is a human gamma herpes virus that is best known for being the causative agent of infectious mononucleosis in man.

A fascinating feature of this virus is its ability to persist in the host and it is estimated that more than 95% of adults are carriers of the virus.

Importantly, EBV can transform latently infected primary cells from healthy individuals into cancerous ones, thereby causing important human cancers such as:

1. B-cell neoplasms (Burkitt's lymphoma and post-transplant lymphomas)2. Certain forms of T-cell lymphoma3. Some epithelial tumors (e.g. gastric carcinomas)

Outcomes of viral latency:

Page 43: VIRAL LATENCY

Epstein-Barr virus (EBV) is the causative agent of Burkitt's lymphoma in Africa (as shown in figure), nasopharyngeal cancer in the orient and infectious mononucleosis in the west.

It was first discovered as the causative agent of Burkitt's lymphoma and it was later found that patients with infectious mononucleosis have antibodies that react with Burkitt's lymphoma cells.

Epstein - Barr virus:

Page 44: VIRAL LATENCY

Understanding viral latency, what triggers viral reactivation and the mechanism of transformation of normal host cells into malignant cells are critical for the development of strategies for the prevention and control of this intriguing virus and related cancers.

A. Epidemiology:

A large proportion of the population (90-95%) is infected with Epstein-Barr virus, and these people, although usually asymptomatic, will shed the virus from time to time throughout life.

B. Mode of Transmission: Spread by close contact Also can be spread by blood transfusion

Epstein - Barr virus:

Page 45: VIRAL LATENCY

C. Viral cell targets:

EBV only infects a small number of cell types that express the receptor for complement C3d component (CR2 or CD21). These cells may be: Certain epithelial cells (oro- and naso-pharynx), and B lymphocytes

D. Viral life cycle:

Virus infection involves two types of cells: 1- B cells, where infection is predominantly latent and has the potential to induce growth-transformation of infected cells; and2- Epithelial cells, where infection is predominantly replicative. EBV latent infection of B-lymphocytes is necessary for virus persistence, subsequent replication in epithelial cells, and release of infectious virus into saliva.

Epstein - Barr virus:

Page 46: VIRAL LATENCY
Page 47: VIRAL LATENCY

Following primary infection of B cells, a chronic virus carrier state is established in which the outgrowth of EBV-transformed B cells is controlled by an EBV-specific cytotoxic T-lymphocytes.

Because of the marked CD4+ and CD8+ T-cell response to EBV nuclear proteins in Latency III infected B-lymphocytes, EBV associated lymphoid malignancies are most common in immune compromised people.

Latently infected B cells can become permissive for lytic EBV infection. Infectious virus released from these cells can be shed directly into the saliva or might infect epithelial cells and other B cells. In this way a virus-carrier state is established that is characterized by:

1. Persistent, latent infection in circulating B cells, and2. Occasional EBV replication in B cells and epithelial cells.

Epstein - Barr virus:

Page 48: VIRAL LATENCY

Latency Strategies of Herpes viruses: http://books.google.com.eg/books?id=wHrTa1E-r-AC&hl=en&source=gbs_navlinks_s http://en.wikipedia.org/wiki/Virus_latency http://student.ccbcmd.edu/courses/bio141/lecguide/unit3/viruses/virinf.html http://textbookofbacteriology.net/themicrobialworld/Herpes.html http://en.wikipedia.org/wiki/Lysogenic http://en.wikipedia.org/wiki/Provirus http://science.jrank.org/pages/2548/Episomes.html http://www.answers.com/topic/slow-virus-infection http://www.bio.miami.edu/~cmallery/150/gene/18x5.jpg http://lib.bioinfo.pl/pmid:10688801 http://www.horizonpress.com/ebv2 http://en.wikipedia.org/wiki/HHV_Latency_Associated_Transcript http://www.sciencedaily.com/releases/2005/03/050328173659.htm http://www.wikigenes.org/e/gene/e/3783709.html http://en.wikipedia.org/wiki/Epstein-Barr_nuclear_antigen_1 http://www.ncbi.nlm.nih.gov/pubmed/7957053 http://www.uniprot.org/uniprot/P03211 http://en.wikipedia.org/wiki/Endogenous_retrovirus http://www.nature.com/nrmicro/journal/v6/n3/fig_tab/nrmicro1794_F1.html http://en.wikipedia.org/wiki/File:Large_facial_Burkitt's_Lymphoma.JPG http://users.rcn.com/jkimball.ma.ultranet/BiologyPages/V/Viruses.html#LatentViruses http://users.rcn.com/jkimball.ma.ultranet/BiologyPages/T/TumorSuppressorGenes.html#hpv http://www.biologynews.net/archives/2005/04/10/how_a_latent_virus_eludes_immune_defenses.html

Reference web pages:

Page 49: VIRAL LATENCY

THANK YOU


Recommended