viral interference with human class i major...

21
1 Viral interference with Human Class I Major Histocompatibility Complex Presentation. Adam Huys [email protected] Department of Microbiology and Immunology University of Saskatchewan Saskatchewan, Saskatoon, SK. Canada For: Advance Virology, VTMC 833 © Adam Huys Abstract Introduction Proper folding, loading, transportation and presentation of a functional MHC-I molecule Early Folding Peptide generation Peptide transport The Peptide Loading Complex Export to the Golgi and surface expression Viral interference Targeting MHC for degradation within the ER Preventing virus-derived peptides Interfering with peptide transport Disrupting the Peptide Loading Complex Inhibiting MHC-I transport from the ER to the cell surface Removal of MHC-I molecules from the cell surface Other Conclusion Abstract While most viruses capable of infecting humans fail to maintain their infection for a long period of time, others can maintain a life-long infection. Viruses capable of maintaining a long infection period have evolved a wide variety of mechanisms to avoid the human immune system. One of the ways these select viruses do this is by limiting the effectiveness of the class I major histocompatibility complex (MHC-I) presentation. This limitation is a result of viral interference

Upload: buiquynh

Post on 17-Apr-2018

226 views

Category:

Documents


2 download

TRANSCRIPT

Page 1: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

1

Viral interference with Human Class I Major Histocompatibility Complex Presentation.

Adam Huys

[email protected]

Department of Microbiology and Immunology University of Saskatchewan

Saskatchewan, Saskatoon, SK. Canada

For: Advance Virology, VTMC 833 © Adam Huys

Abstract Introduction Proper folding, loading, transportation and presentation of a functional MHC-I molecule

Early Folding Peptide generation Peptide transport

The Peptide Loading Complex Export to the Golgi and surface expression Viral interference

Targeting MHC for degradation within the ER Preventing virus-derived peptides Interfering with peptide transport Disrupting the Peptide Loading Complex Inhibiting MHC-I transport from the ER to the cell surface Removal of MHC-I molecules from the cell surface Other

Conclusion

Abstract

While most viruses capable of infecting humans fail to maintain their infection for a long

period of time, others can maintain a life-long infection. Viruses capable of maintaining a long

infection period have evolved a wide variety of mechanisms to avoid the human immune system.

One of the ways these select viruses do this is by limiting the effectiveness of the class I major

histocompatibility complex (MHC-I) presentation. This limitation is a result of viral interference

Page 2: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

2

in the MHC-I pathway at various points, such as MHC-I folding, peptide generation and

translocation, and MHC-I transport to and presentation on the cell surface. This review will

examine the viruses that have been identified to interfere with MHC-I presentation and where the

interference occurs along the pathway.

Introduction

Viruses and their human hosts have evolved together for millions of years. As a result the

host has evolved mechanisms for controlling virus spread and infection. Likewise, the viruses

have adapted mechanisms to avoid the host defenses. One of the important defenses employed

by humans against virus infection is the CD8+ cytotoxic T-lymphocyte (CTL), which is capable

of identifying and killing virally infected cells. Identification of infected cells by CTLs is

mediated by presentation of endogenously expressed peptides on the surface of infected cells.

These peptides are exported and presented on the cell surface via the class I major

histocompatibility complex (MHC-I) molecule.

MHC-I molecules and the peptides they present function as a communication device

between cells and the immune system. The peptides being carried on the MHC-I molecules

represent a mechanism by which the cell reports what is transpiring internally. In normal healthy

cells, the peptides being expressed are those which are considered “self” and under normal

conditions do not stimulate CTLs. When conditions within the cell change such as during a viral

infection, the viral proteins being translated become candidates for MHC-I presentation. These

proteins are viewed as foreign by the CTLs and therefore the MHC-I has delivered a message

that internal processes are not normal. Due to this mechanism, any virus having its protein

presented in the form of peptides on the MHC-I molecule is a great threat to the survival of the

virus, as CTL detection of foreign peptides leads to CTL-mediated cell killing to eliminate any

Page 3: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

3

threat. However, as viruses and humans have evolved together, viruses have developed

mechanisms to inhibit MHC-I molecules from delivering warnings to the immune system. These

mechanisms can affect almost every aspect of the MHC-I presentation pathway (pathway is

shown in Figure 1) from proper loading and folding, to transport to the cell surface. These

mechanisms of viral interference are shown in Figure 2 and summarized in Table 1.

Page 4: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

4

Page 5: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

5

Proper folding, loading, transportation and presentation of a functional MHC-I molecule

Early Folding

A functional MHC-I molecule found on the surface of a cell is composed of a heavy

chain (HC), a β2-microglubulin (β2m) and a peptide. The HC and β2m are independently

translated directly into the endoplasmic reticulum (ER) via the Sec61 translocon, an ER

membrane transport protein (6, 61). Upon entry into the ER the HC is immediately glycosylated.

This added sugar allows the recruitment of resident ER chaperones, of which the most strongly

implicated is calnexin (CNX). If a cell is deficient of CNX it is still able to make functional

MHC-I molecules, this is believed to be the consequence of immunoglobulin-binding protein

Page 6: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

6

(BiP) performing a similar task to that of CNX. The binding of the chaperone alters the HC

conformation to allow it to associate with β2m. This association releases the chaperone leaving

an “empty” MHC-I molecule ready for peptide loading (15, 56).

Peptide generation

Peptides are small proteins that are either translated on their own or are derivatives of

protein degradation. The peptides that are most often associated with MHC-I molecules are

defective ribosomal translation products (DRIPs) (59), which are degraded into peptides in the

cytosol by the 26S proteasome (18). Because of the MHC-I preference to present DRIPs, it

ensures that the peptides being presented are a representative of what is currently being

transcribed within the cell as opposed to displaying old, used degraded proteins. This preference

leads to a reduction in the amount of time required for the cell to alert the immune system as they

represent proteins that are actively undergoing translation with no lag time for the protein to be

fully synthesized, used, and degraded.

Peptide transport

The “empty” MHC-I molecules are localized in the ER, while the peptides they require

are generated in the cytosol. In order for the MHC-I to be loaded, the peptides must be

transported into the ER. This transportation is facilitated by heterodimers composed of TAP1

and TAP2. Transporter associated with antigen presentation (TAP), is a member of the ATP-

binding cassette (ABC) transporter family. Peptide translocation to the ER is a form of active

transport as it requires energy supplied by ATP to function. TAP preferentially binds peptides

that are 8-16 amino acids (aa) long but is capable of binding both smaller and longer peptides.

The binding of a peptide to TAP produces a conformational change within the transporter to

allow the binding of ATP. The hydrolysis of ATP results in the transposition of the peptide in

Page 7: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

7

the cytosol to the lumen of the ER. TAP is most efficient at transporting peptides 8-12 aa long

(2, 3, 65). Once the peptide has been successfully transported across the ER membrane it is

trimmed by the aminopeptidases ERAAP (ER aminopeptidase associated with antigen

presentation) to make it more suitable for MHC-I loading (56, 70).

The Peptide Loading Complex

The peptide loading complex (PLC) is constructed for the loading of the peptide onto the

empty MHC-I molecule. The exact mechanism of PLC assembly is not yet fully understood, but

there are some aspects that are already known. Calreticulin (CRT), which is virtually a soluble

homologue of the CNX chaperone, described previously, binds to the empty MHC-I molecule.

The molecule then associates with tapasin and ERp57. Whether ERp57 associates with CRT or

tapasin first is unknown. Tapasin is bound to TAP, and its association with MHC-I molecules

therefore brings MHC-I molecules to the site of peptide translocation. Tapasin is also believed

to be involved in ensuring the binding of high affinity peptides with MHC-I by keeping the

MHC-I in an open form, this ensure that only high affinity peptides will be able to induce MHC-I

closure. Lower affinity peptides will come into the opening but will disassociate because they do

not interact strongly enough to close the binding pocket. When a high affinity peptide enters the

MHC-I opening, it is strong enough to close the MHC-I molecule, which results in the

disassociation of MHC-I from the PLC (15, 56, 70).

Tapasin knockout cell lines exhibit decreased peptide presentation and decreased CTL

response. This phenotype is attributed to a decrease in quality peptide binding (23, 58). Humans

encode four different MHC-I alleles also known as human leukocyte antigens (HLA) -A, -B, -C,

and –E. Interestingly it has been demonstrated that variation within the HLA alleles can lead to

Page 8: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

8

phenotypes which do not associate with tapasin, therefore negating its ability to associate with

the PLC (47, 57).

Export to the Golgi and surface expression

After dissociation from the PLC, the peptide-loaded MHC-I molecules interact with

cargo protein BAP31, which escorts the MHC-I molecule within the ER to exit sites. Once at an

exit site, the BAP31-MHC-I complex is secreted in a COP II decorated vesicle and is destined

for the Golgi apparatus (1, 45, 67). If a low-affinity peptide is loaded, tapasin is less likely to

detach from the MHC-I molecule when it exits the PLC. This allows tapasin to be transported

along with the MHC-I molecule to the Golgi apparatus. However, tapasin contains an ER

retrieval signal, so when it reaches the Golgi apparatus, this signal will promote the return of the

MHC-I molecule to the ER, where the MHC-I will be given another chance to associate with a

peptide with higher binding affinity (55). Successfully loaded MHC-I molecules will travel

through the Golgi and be expressed on the cell surface, where it can present its peptide to the

immune system.

Viral interference

Targeting MHC for degradation within the ER

Human cytomegalovirus (HCMV) encodes two glycoproteins, US2 and US11, which are

capable of removing MHC-I molecules from the ER and promoting their proteasome degradation

in the cytosol (40, 71). The ER has a maintenance system called ER associated degradation

(ERAD) for ensuring that improperly folded proteins are discarded. Proteins destined for

degradation by ERAD are polyubiquinated and digested by proteasomes in the cytosol (51).

US2 and US11 promote ERAD of MHC-I molecules in two unique fashions (21). US11 is able

to recruit Derlin-1, a known initiator of ERAD, to MHC-I molecules. This results in MHC-I

Page 9: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

9

entering the ERAD process to be ubiquinated and undergo proteolysis. US2 promotes

translocation to the cytosol and ubiquination of MHC-I in a yet to be determined mechanism that

does not involve Derlin-1 (48, 66). Although US2 and US11 perform a similar function, they

interact with MHC-I in different regions of the MHC-I molecule. This makes the likeliness of

the host avoiding both degradation systems much lower. Because they recognize MHC-I in

different locations of the protein, US2 and US11 have different binding domains and will vary in

the specificity to which they recognize the different human alleles (11, 12, 24, 25, 49).

The p12 protein of Human T-cell leukemia virus (HTLV-I) has been shown to promote

the ER exit and degradation of MHC-I HC before it interacts with β2m (39). Unfortunately, at

this point in time there is no suggested mechanism of how p12 interacts with MHC-I and

promotes its early exit from the ER.

Preventing formation of virus-derived peptides

One of the mechanisms viruses can use to prevent being displayed on MHC-I molecules

is preventing the formation of peptides derived from their proteins. Two human viruses which

have displayed this ability come from the family Herpesviridae. Epstein-Barr virus (EBV) and

Karposi sarcoma-associated herpesvirus (KSHV) both infect and establish latency in human B

cells. During a latent infection of EBV, Epstein-Barr nuclear antigen 1 (EBNA-1) is expressed

to maintain the viral episome (72). As this is the only viral protein present during latency, it is

the only viral protein that could potentially trigger an immune response through CTL antigen

recognition during latent infection. EBNA-1 prevents itself from being displayed on the MHC-I

molecule in two fashions. The first mechanism encoded by EBNA-1 is limiting its translation

speed (73). By limiting its own translation speed, it is much more likely to be synthesized

correctly, decreasing the likelihood that it will become a DRIP. The second method EBNA-1

Page 10: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

10

employs to avoid being presented on MHC-I is its stability and ability to avoid being degraded

by the proteasome (46). Both of these mechanisms are attributed to the glycine-alanine repeat

region (GAr) found within the EBNA-1. Latency-associated nuclear antigen 1 (LANA-1) is

synthesized by KSHV during latency, and carries out the same functions as EBNA-1, by

preventing the formation of LANA-1 derived peptides in an identical manner. The sole

exception to the similarities between LANA-1 and EBNA-1 is that LANA-1 contains a GZ

(glycine, glutamic acid/aspartic acid) repeat region as opposed to a GAr (42, 74). Overall

EBNA-1 and LANA-1 both decrease the amount of their specific peptides being synthesized,

therefore decreasing the probability of being displayed on the MHC-I molecule and having its’

host cell destroyed.

Interfering with peptide transport

As discussed earlier, peptides must be transported across the ER to be loaded onto MHC-

I molecules. Thus viruses have evolved to target this step to prevent MHC-I presentation.

Herpes simplex virus type 1 (HSV-1) translates a protein, ICP47 which has high binding affinity

for TAP. This results in ICP47 competing with cellular-derived peptides for TAP binding (5, 31,

68). ICP47 binding to TAP also interferes with the stabilization of the heterodimer TAP1/TAP2

(44). When ICP47 binds TAP it prevents other peptides from binding to the transporter, therefore

preventing the transportation of any peptides from the cytosol into the ER.

HCMV encodes the US6 protein, which when translated is located in the lumen of the

ER, but is embedded in the ER membrane through its trans-membrane domain (28). US6

interferes with TAP on the ER side. The interference leads to a conformational change in TAP

that prevents it from binding ATP. TAP is still able to bind peptides, but is unable to receive the

Page 11: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

11

energy from ATP to transport the peptide into the ER (30, 43). The inability of TAP to access its

energy source leads to inhibition of peptides being transported into the ER for MHC-I loading.

BNLF2α is a protein expressed during the lytic phase of EBV infection and has recently

been shown to decrease MHC-I surface expression. It has been demonstrated that BNLF2α

interacts with TAP and inhibits the ability of TAP to bind both ATP and peptides (33, 60). It is

predicted to be able to accomplish this by inserting itself into the ER membrane, and then

interacting with TAP1, which stabilizes BNLF2α. This interaction directly interferes with ATP

and peptide binding on the cytosol side of the ER membrane. Although BNLF2α structure has

not been determined, it is hypothesized that this protein contains a trans-membrane domain in its

C-terminal end, because the C-terminal domain is hydrophobic. BNLF2α localization assays

also visualize the protein at the ER membrane (34), providing further support to the possibility

that BNLF2α inhibits MHC-I surface expression by interacting with TAP resulting in the

inhibition of peptide transport into the ER.

The E7 protein encoded by Human papillomavirus-11 has been demonstrated to interact

with TAP1. This interaction inhibits the ability of TAP to transport peptides across the ER. The

lack of available peptides for MHC-I loading decreases the surface expression of MHC-I

molecules (69).

Disrupting the Peptide Loading Complex

Disabling the formation of the PLC results in a decrease in MHC-I surface expression,

and this represents targets which viruses have evolved to disrupt. The adenoviruses translate the

early protein E19, which has been demonstrated to reduce the surface expression of MHC-I (7,

13). E19 has the ability to bind to empty MHC-I molecules and to TAP (14). This ability is a

mimic of the role tapasin plays in the PLC. Another mechanism employed by E19 to diminish

Page 12: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

12

MHC-I surface expression is encoded in its cytosolic tail, which contains a dilysine motif

(KKXX). This motif functions as an ER retrieval signal. If the MHC-I molecule is bound by

E19 when it is transported to the Golgi, the coatamer protein complex (COPI) will recognize the

motif and return it to the ER (22, 38).

The US3 protein of HCMV has also been demonstrated to interfere with the PLC. US3

functions similarly to E19 in that it too has the ability to bind TAP and MHC-I molecules

reducing the amount of MHC-I being successfully loaded with peptides (54). However unlike

E19, US3 does not contain an ER retrieval motif, therefore the protein does not force MHC-I

molecules to return to the ER after transportation to the Golgi.

Inhibiting MHC-I transport from the ER to the cell surface

All MHC-I molecules must make the journey from the ER to the Golgi apparatus. This

step in the MHC-I presentation pathway is the limiting factor for how much MHC-I is expressed

on the cell surface (67). As it is a limiting step viruses have evolved mechanisms to reduce its

speed. As discussed above, adenoviruses have a mechanism to inhibit MHC-I molecules by

perturbing the transportation of the MHC-I from the ER to the Golgi. Another human

pathogenic virus which inhibits this process is HCMV, which encodes US10. US10 is capable of

interacting with MHC-I and retaining the molecule in the ER (20). This alone does not decrease

MHC-I presentation, but allows the MHC-I molecules to be exposed to other HCMV proteins,

such as US2 and US11, for a greater period of time (52). If US2 and US11 have more time to

interact with MHC-I it will further decrease MHC-I surface expression. This additive affect is

seen with US3 also (53).

Human herpesvirus 7 (HHV-7) and human herpesvirus 6 (HHV-6) encode the protein

U21. Although this protein demonstrates only 30% similarity between the two viruses, it has

Page 13: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

13

been shown to downregulate MHC-I presentation (32, 36). U21 has the ability to bind properly

folded MHC-I molecules in the ER and accompany it to the Golgi apparatus. Once the MHC-

I/U21 complex arrives at the Golgi, the U21 dictates their transport to lysosomal compartments

where the two proteins are degraded (26, 35).

The Nef protein encoded by HIV-1 has been shown to downregulate several surface-

expressed receptors, one of these being the MHC-I molecule (8). Nef associates with MHC-I

and accompanies it to the Golgi (41). The Nef/MHC-I complex travels through the trans Golgi

network (TGN) until it comes into contact with adaptor protein 1 (AP-1). AP-1 is capable of

retaining proteins in TGN and initiating its re-direction to lysosomes (63). Nef, when associated

with MHC-I, is capable of recruiting AP-1 to MHC-I cytosolic tail. Whether AP-1 interacts with

Nef/MHC-I on its way to the cell surface or whether it interacts with the complex upon being

internalized from the cell surface, which would also be induced by Nef, is still not understood,

but there is evidence supporting both models (50, 62).

Human papillomavirus-16 encodes the protein E5. This protein has been demonstrated to

interfere with MHC-I presentation pathway. E5 has been shown to interact with both the heavy

chain of the MHC-I and the ER chaperone CNX. This interaction is hypothesized to form a

ternary protein complex consisting of MHC-I molecule, CNX and E5 resulting in the MHC-I

molecule being accumulated in the ER and Golgi apparatus preventing its expression on the cell

surface (9, 10, 27).

Removal of MHC-I molecules from the cell surface

The proteins K3 and K5, also called modulator of immune recognition 1 and 2 (MIR-1,-

2), are expressed by KSHV and promote internalization of surface-expressed MHC-I molecules

(37), thus inhibiting the final stages of MHC-I presentation: the ability to interact with CTLs. K3

Page 14: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

14

and K5 are able to recruit enzymes capable of initiating ubiquination of surface-expressed MHC-

I. The KSHV proteins are able to promote this recruitment through their variant really

interesting new gene (RING-CH) motif located in the N-terminal domain (17). The ubiquination

results in the MHC-I being internalized from the cell surface and shipped to a lysosome for

degradation (16). K3 and K5 association with MHC-I is predicted to occur in the medial-Golgi

compartment (29). K3 and K5 have identical functions, but their effectiveness for each HLA

allele differs: K3 is capable of down regulating all of the HLA alleles, while K5 is only able to

decrease the expression of HLA-A, and –B (64).

During the lytic stage of an EBV infection, EBV produces BILF1. This protein has

recently been demonstrated to reduce the amount of MHC-I surface expression. BILF1 has the

ability to directly interact with MHC-I molecules. This interaction results in an increase of

MHC-I surface turnover and presumably their degradation in lysosomal compartments. This

interaction between MHC-I and BILF1 is hypothesized to occur early in the MHC-I expression

pathway as immunoprecipitation indicate that there is association between the two proteins

within 15 minutes of MHC-I expression(76).

Other

Varicella-zoster virus (VZV) encodes the protein ORF66 which has been shown to

decrease surface expression of MHC-I molecule, however the mechanism by which ORF66

function is still unknown (4, 19).

Core protein of hepatitis C has also been demonstrated to down regulate surface

expression of MHC-I molecules. However at this time there has been no published suggestion of

mechanism of how core protein achieves this feat (75).

Conclusion

Page 15: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

15

By examining all the viruses that interfere with MHC-I presentation, it is possible to

identify a common theme amongst them. These viruses are all capable of becoming latent or

establishing long term chronic infections in humans. This is an interesting common theme

because if a virus wishes to become latent or cause a chronic infection, it must remain unnoticed

by the adaptive immune response. MHC-I presentation of peptides and the recognition of the

peptides by CTLs is a form of adaptive immunity. Viruses which infect their host and rapidly

replicate and spread to a new host do not appear to inhibit MHC-I presentation because they do

not benefit, or are not capable of becoming latent. The theme also demonstrates how important

the MHC-I presentation is to immune protection. Viruses that can successfully establish a long

infection period interfere with it, while those that do not interfere fail to establish an infection for

a long period of time.

Viral interference with MHC-I presentation also demonstrates how humans and viruses

have evolved together. The viruses that are most effective at evading the human immune system

are also those that are believed to have infected humans the longest. Some viruses affecting

MHC-I presentation encode multiple proteins which affect MHC-I, while other viruses encode

only one protein. Perhaps the redundancy and multiple modes of MHC-I inhibition can act as

markers for how long humans have been evolving with infection to these viruses. For example

HCMV has multiple proteins which are involved in preventing MHC-I presentation. Perhaps this

apparent redundancy has occurred as a result of an arms race between the human immune system

and HCMV, which has been occurring since the dawn of humans. Humans evolved MHC-I

presentation, so under selective pressure HCMV evolved a protein to inhibit MHC-I formation.

Humans then evolved different alleles, so in turn HCMV evolved a second protein that would

better recognize that allele. As discussed above, HSV only encodes one protein which inhibits

Page 16: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

16

MHC-I presentation. This protein, ICP47, inhibits all MHC-I alleles by affecting peptide supply

in the ER. This illustrates that perhaps HCMV evolved with us, maybe even before we were

humans, where as HSV may have become a pathogen to us more recently in our and its

evolution.

Acknowledgements

I would like to extend my gratitude to Jennifer Zaba and Patricia McGee for their help and time

with proof reading and editing, without it this paper would have suffered grammatically.

References 1. Abe, F., N. Van Prooyen, J. J. Ladasky, and M. Edidin. 2009. Interaction of Bap31

and MHC class I molecules and their traffic out of the endoplasmic reticulum. J Immunol 182:4776-83.

2. Abele, R., and R. Tampe. 2004. The ABCs of immunology: structure and function of TAP, the transporter associated with antigen processing. Physiology (Bethesda) 19:216-24.

3. Abele, R., and R. Tampe. 2006. Modulation of the antigen transport machinery TAP by friends and enemies. FEBS Lett 580:1156-63.

4. Abendroth, A., I. Lin, B. Slobedman, H. Ploegh, and A. M. Arvin. 2001. Varicella-zoster virus retains major histocompatibility complex class I proteins in the Golgi compartment of infected cells. J Virol 75:4878-88.

5. Ahn, K., T. H. Meyer, S. Uebel, P. Sempe, H. Djaballah, Y. Yang, P. A. Peterson, K. Fruh, and R. Tampe. 1996. Molecular mechanism and species specificity of TAP inhibition by herpes simplex virus ICP47. EMBO J 15:3247-55.

6. Ambagala, A. P., J. C. Solheim, and S. Srikumaran. 2005. Viral interference with MHC class I antigen presentation pathway: the battle continues. Vet Immunol Immunopathol 107:1-15.

7. Andersson, M., S. Paabo, T. Nilsson, and P. A. Peterson. 1985. Impaired intracellular transport of class I MHC antigens as a possible means for adenoviruses to evade immune surveillance. Cell 43:215-22.

8. Arora, V. K., B. L. Fredericksen, and J. V. Garcia. 2002. Nef: agent of cell subversion. Microbes Infect 4:189-99.

9. Ashrafi, G. H., M. Haghshenas, B. Marchetti, and M. S. Campo. 2006. E5 protein of human papillomavirus 16 downregulates HLA class I and interacts with the heavy chain via its first hydrophobic domain. Int J Cancer 119:2105-12.

10. Ashrafi, G. H., M. R. Haghshenas, B. Marchetti, P. M. O'Brien, and M. S. Campo. 2005. E5 protein of human papillomavirus type 16 selectively downregulates surface HLA class I. Int J Cancer 113:276-83.

Page 17: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

17

11. Barel, M. T., N. Pizzato, D. van Leeuwen, P. L. Bouteiller, E. J. Wiertz, and F. Lenfant. 2003. Amino acid composition of alpha1/alpha2 domains and cytoplasmic tail of MHC class I molecules determine their susceptibility to human cytomegalovirus US11-mediated down-regulation. Eur J Immunol 33:1707-16.

12. Barel, M. T., M. Ressing, N. Pizzato, D. van Leeuwen, P. Le Bouteiller, F. Lenfant, and E. J. Wiertz. 2003. Human cytomegalovirus-encoded US2 differentially affects surface expression of MHC class I locus products and targets membrane-bound, but not soluble HLA-G1 for degradation. J Immunol 171:6757-65.

13. Beier, D. C., J. H. Cox, D. R. Vining, P. Cresswell, and V. H. Engelhard. 1994. Association of human class I MHC alleles with the adenovirus E3/19K protein. J Immunol 152:3862-72.

14. Bennett, E. M., J. R. Bennink, J. W. Yewdell, and F. M. Brodsky. 1999. Cutting edge: adenovirus E19 has two mechanisms for affecting class I MHC expression. J Immunol 162:5049-52.

15. Bouvier, M. 2003. Accessory proteins and the assembly of human class I MHC molecules: a molecular and structural perspective. Mol Immunol 39:697-706.

16. Coscoy, L. 2007. Immune evasion by Kaposi's sarcoma-associated herpesvirus. Nat Rev Immunol 7:391-401.

17. Coscoy, L., D. J. Sanchez, and D. Ganem. 2001. A novel class of herpesvirus-encoded membrane-bound E3 ubiquitin ligases regulates endocytosis of proteins involved in immune recognition. J Cell Biol 155:1265-73.

18. Coux, O., K. Tanaka, and A. L. Goldberg. 1996. Structure and functions of the 20S and 26S proteasomes. Annu Rev Biochem 65:801-47.

19. Eisfeld, A. J., M. B. Yee, A. Erazo, A. Abendroth, and P. R. Kinchington. 2007. Downregulation of class I major histocompatibility complex surface expression by varicella-zoster virus involves open reading frame 66 protein kinase-dependent and -independent mechanisms. J Virol 81:9034-49.

20. Furman, M. H., N. Dey, D. Tortorella, and H. L. Ploegh. 2002. The human cytomegalovirus US10 gene product delays trafficking of major histocompatibility complex class I molecules. J Virol 76:11753-6.

21. Furman, M. H., H. L. Ploegh, and D. Tortorella. 2002. Membrane-specific, host-derived factors are required for US2- and US11-mediated degradation of major histocompatibility complex class I molecules. J Biol Chem 277:3258-67.

22. Gabathuler, R., and S. Kvist. 1990. The endoplasmic reticulum retention signal of the E3/19K protein of adenovirus type 2 consists of three separate amino acid segments at the carboxy terminus. J Cell Biol 111:1803-10.

23. Garbi, N., P. Tan, A. D. Diehl, B. J. Chambers, H. G. Ljunggren, F. Momburg, and G. J. Hammerling. 2000. Impaired immune responses and altered peptide repertoire in tapasin-deficient mice. Nat Immunol 1:234-8.

24. Gewurz, B. E., R. Gaudet, D. Tortorella, E. W. Wang, H. L. Ploegh, and D. C. Wiley. 2001. Antigen presentation subverted: Structure of the human cytomegalovirus protein US2 bound to the class I molecule HLA-A2. Proc Natl Acad Sci U S A 98:6794-9.

25. Gewurz, B. E., E. W. Wang, D. Tortorella, D. J. Schust, and H. L. Ploegh. 2001. Human cytomegalovirus US2 endoplasmic reticulum-lumenal domain dictates

Page 18: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

18

association with major histocompatibility complex class I in a locus-specific manner. J Virol 75:5197-204.

26. Glosson, N. L., and A. W. Hudson. 2007. Human herpesvirus-6A and -6B encode viral immunoevasins that downregulate class I MHC molecules. Virology 365:125-35.

27. Gruener, M., I. G. Bravo, F. Momburg, A. Alonso, and P. Tomakidi. 2007. The E5 protein of the human papillomavirus type 16 down-regulates HLA-I surface expression in calnexin-expressing but not in calnexin-deficient cells. Virol J 4:116.

28. Halenius, A., F. Momburg, H. Reinhard, D. Bauer, M. Lobigs, and H. Hengel. 2006. Physical and functional interactions of the cytomegalovirus US6 glycoprotein with the transporter associated with antigen processing. J Biol Chem 281:5383-90.

29. Hewitt, E. W., L. Duncan, D. Mufti, J. Baker, P. G. Stevenson, and P. J. Lehner. 2002. Ubiquitylation of MHC class I by the K3 viral protein signals internalization and TSG101-dependent degradation. EMBO J 21:2418-29.

30. Hewitt, E. W., S. S. Gupta, and P. J. Lehner. 2001. The human cytomegalovirus gene product US6 inhibits ATP binding by TAP. EMBO J 20:387-96.

31. Hill, A. B., B. C. Barnett, A. J. McMichael, and D. J. McGeoch. 1994. HLA class I molecules are not transported to the cell surface in cells infected with herpes simplex virus types 1 and 2. J Immunol 152:2736-41.

32. Hirata, Y., K. Kondo, and K. Yamanishi. 2001. Human herpesvirus 6 downregulates major histocompatibility complex class I in dendritic cells. J Med Virol 65:576-83.

33. Hislop, A. D., M. E. Ressing, D. van Leeuwen, V. A. Pudney, D. Horst, D. Koppers-Lalic, N. P. Croft, J. J. Neefjes, A. B. Rickinson, and E. J. Wiertz. 2007. A CD8+ T cell immune evasion protein specific to Epstein-Barr virus and its close relatives in Old World primates. J Exp Med 204:1863-73.

34. Horst, D., D. van Leeuwen, N. P. Croft, M. A. Garstka, A. D. Hislop, E. Kremmer, A. B. Rickinson, E. J. Wiertz, and M. E. Ressing. 2009. Specific targeting of the EBV lytic phase protein BNLF2a to the transporter associated with antigen processing results in impairment of HLA class I-restricted antigen presentation. J Immunol 182:2313-24.

35. Hudson, A. W., D. Blom, P. M. Howley, and H. L. Ploegh. 2003. The ER-lumenal domain of the HHV-7 immunoevasin U21 directs class I MHC molecules to lysosomes. Traffic 4:824-37.

36. Hudson, A. W., P. M. Howley, and H. L. Ploegh. 2001. A human herpesvirus 7 glycoprotein, U21, diverts major histocompatibility complex class I molecules to lysosomes. J Virol 75:12347-58.

37. Ishido, S., C. Wang, B. S. Lee, G. B. Cohen, and J. U. Jung. 2000. Downregulation of major histocompatibility complex class I molecules by Kaposi's sarcoma-associated herpesvirus K3 and K5 proteins. J Virol 74:5300-9.

38. Jackson, M. R., T. Nilsson, and P. A. Peterson. 1990. Identification of a consensus motif for retention of transmembrane proteins in the endoplasmic reticulum. EMBO J 9:3153-62.

39. Johnson, J. M., C. Nicot, J. Fullen, V. Ciminale, L. Casareto, J. C. Mulloy, S. Jacobson, and G. Franchini. 2001. Free major histocompatibility complex class I heavy chain is preferentially targeted for degradation by human T-cell leukemia/lymphotropic virus type 1 p12(I) protein. J Virol 75:6086-94.

40. Jones, T. R., and L. Sun. 1997. Human cytomegalovirus US2 destabilizes major histocompatibility complex class I heavy chains. J Virol 71:2970-9.

Page 19: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

19

41. Kasper, M. R., J. F. Roeth, M. Williams, T. M. Filzen, R. I. Fleis, and K. L. Collins. 2005. HIV-1 Nef disrupts antigen presentation early in the secretory pathway. J Biol Chem 280:12840-8.

42. Kwun, H. J., S. R. da Silva, I. M. Shah, N. Blake, P. S. Moore, and Y. Chang. 2007. Kaposi's sarcoma-associated herpesvirus latency-associated nuclear antigen 1 mimics Epstein-Barr virus EBNA1 immune evasion through central repeat domain effects on protein processing. J Virol 81:8225-35.

43. Kyritsis, C., S. Gorbulev, S. Hutschenreiter, K. Pawlitschko, R. Abele, and R. Tampe. 2001. Molecular mechanism and structural aspects of transporter associated with antigen processing inhibition by the cytomegalovirus protein US6. J Biol Chem 276:48031-9.

44. Lacaille, V. G., and M. J. Androlewicz. 1998. Herpes simplex virus inhibitor ICP47 destabilizes the transporter associated with antigen processing (TAP) heterodimer. J Biol Chem 273:17386-90.

45. Ladasky, J. J., S. Boyle, M. Seth, H. Li, T. Pentcheva, F. Abe, S. J. Steinberg, and M. Edidin. 2006. Bap31 enhances the endoplasmic reticulum export and quality control of human class I MHC molecules. J Immunol 177:6172-81.

46. Levitskaya, J., A. Sharipo, A. Leonchiks, A. Ciechanover, and M. G. Masucci. 1997. Inhibition of ubiquitin/proteasome-dependent protein degradation by the Gly-Ala repeat domain of the Epstein-Barr virus nuclear antigen 1. Proc Natl Acad Sci U S A 94:12616-21.

47. Lewis, J. W., A. Sewell, D. Price, and T. Elliott. 1998. HLA-A*0201 presents TAP-dependent peptide epitopes to cytotoxic T lymphocytes in the absence of tapasin. Eur J Immunol 28:3214-20.

48. Lilley, B. N., and H. L. Ploegh. 2005. Viral modulation of antigen presentation: manipulation of cellular targets in the ER and beyond. Immunol Rev 207:126-44.

49. Llano, M., M. Guma, M. Ortega, A. Angulo, and M. Lopez-Botet. 2003. Differential effects of US2, US6 and US11 human cytomegalovirus proteins on HLA class Ia and HLA-E expression: impact on target susceptibility to NK cell subsets. Eur J Immunol 33:2744-54.

50. Lubben, N. B., D. A. Sahlender, A. M. Motley, P. J. Lehner, P. Benaroch, and M. S. Robinson. 2007. HIV-1 Nef-induced down-regulation of MHC class I requires AP-1 and clathrin but not PACS-1 and is impeded by AP-2. Mol Biol Cell 18:3351-65.

51. Meusser, B., C. Hirsch, E. Jarosch, and T. Sommer. 2005. ERAD: the long road to destruction. Nat Cell Biol 7:766-72.

52. Miller-Kittrell, M., and T. E. Sparer. 2009. Feeling manipulated: cytomegalovirus immune manipulation. Virol J 6:4.

53. Noriega, V. M., and D. Tortorella. 2009. Human cytomegalovirus-encoded immune modulators partner to downregulate major histocompatibility complex class I molecules. J Virol 83:1359-67.

54. Park, B., Y. Kim, J. Shin, S. Lee, K. Cho, K. Fruh, and K. Ahn. 2004. Human cytomegalovirus inhibits tapasin-dependent peptide loading and optimization of the MHC class I peptide cargo for immune evasion. Immunity 20:71-85.

55. Paulsson, K. M., M. Jevon, J. W. Wang, S. Li, and P. Wang. 2006. The double lysine motif of tapasin is a retrieval signal for retention of unstable MHC class I molecules in the endoplasmic reticulum. J Immunol 176:7482-8.

Page 20: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

20

56. Peaper, D. R., and P. Cresswell. 2008. Regulation of MHC class I assembly and peptide binding. Annu Rev Cell Dev Biol 24:343-68.

57. Peh, C. A., S. R. Burrows, M. Barnden, R. Khanna, P. Cresswell, D. J. Moss, and J. McCluskey. 1998. HLA-B27-restricted antigen presentation in the absence of tapasin reveals polymorphism in mechanisms of HLA class I peptide loading. Immunity 8:531-42.

58. Purcell, A. W., J. J. Gorman, M. Garcia-Peydro, A. Paradela, S. R. Burrows, G. H. Talbo, N. Laham, C. A. Peh, E. C. Reynolds, J. A. Lopez De Castro, and J. McCluskey. 2001. Quantitative and qualitative influences of tapasin on the class I peptide repertoire. J Immunol 166:1016-27.

59. Reits, E. A., J. C. Vos, M. Gromme, and J. Neefjes. 2000. The major substrates for TAP in vivo are derived from newly synthesized proteins. Nature 404:774-8.

60. Ressing, M. E., S. E. Keating, D. van Leeuwen, D. Koppers-Lalic, I. Y. Pappworth, E. J. Wiertz, and M. Rowe. 2005. Impaired transporter associated with antigen processing-dependent peptide transport during productive EBV infection. J Immunol 174:6829-38.

61. Roder, G., L. Geironson, I. Bressendorff, and K. Paulsson. 2008. Viral proteins interfering with antigen presentation target the major histocompatibility complex class I peptide-loading complex. J Virol 82:8246-52.

62. Roeth, J. F., and K. L. Collins. 2006. Human immunodeficiency virus type 1 Nef: adapting to intracellular trafficking pathways. Microbiol Mol Biol Rev 70:548-63.

63. Roeth, J. F., M. Williams, M. R. Kasper, T. M. Filzen, and K. L. Collins. 2004. HIV-1 Nef disrupts MHC-I trafficking by recruiting AP-1 to the MHC-I cytoplasmic tail. J Cell Biol 167:903-13.

64. Sanchez, D. J., L. Coscoy, and D. Ganem. 2002. Functional organization of MIR2, a novel viral regulator of selective endocytosis. J Biol Chem 277:6124-30.

65. Scholz, C., and R. Tampe. 2005. The intracellular antigen transport machinery TAP in adaptive immunity and virus escape mechanisms. J Bioenerg Biomembr 37:509-15.

66. Shamu, C. E., C. M. Story, T. A. Rapoport, and H. L. Ploegh. 1999. The pathway of US11-dependent degradation of MHC class I heavy chains involves a ubiquitin-conjugated intermediate. J Cell Biol 147:45-58.

67. Spiliotis, E. T., H. Manley, M. Osorio, M. C. Zuniga, and M. Edidin. 2000. Selective export of MHC class I molecules from the ER after their dissociation from TAP. Immunity 13:841-51.

68. Tomazin, R., A. B. Hill, P. Jugovic, I. York, P. van Endert, H. L. Ploegh, D. W. Andrews, and D. C. Johnson. 1996. Stable binding of the herpes simplex virus ICP47 protein to the peptide binding site of TAP. EMBO J 15:3256-66.

69. Vambutas, A., J. DeVoti, W. Pinn, B. M. Steinberg, and V. R. Bonagura. 2001. Interaction of human papillomavirus type 11 E7 protein with TAP-1 results in the reduction of ATP-dependent peptide transport. Clin Immunol 101:94-9.

70. Wearsch, P. A., and P. Cresswell. 2008. The quality control of MHC class I peptide loading. Curr Opin Cell Biol 20:624-31.

71. Wiertz, E. J., T. R. Jones, L. Sun, M. Bogyo, H. J. Geuze, and H. L. Ploegh. 1996. The human cytomegalovirus US11 gene product dislocates MHC class I heavy chains from the endoplasmic reticulum to the cytosol. Cell 84:769-79.

Page 21: Viral interference with Human Class I Major …homepage.usask.ca/~vim458/advirol/2010/huys/viral... ·  · 2010-01-03Viral interference with Human Class I Major Histocompatibility

21

72. Wu, H., P. Kapoor, and L. Frappier. 2002. Separation of the DNA replication, segregation, and transcriptional activation functions of Epstein-Barr nuclear antigen 1. J Virol 76:2480-90.

73. Yin, Y., B. Manoury, and R. Fahraeus. 2003. Self-inhibition of synthesis and antigen presentation by Epstein-Barr virus-encoded EBNA1. Science 301:1371-4.

74. Zaldumbide, A., M. Ossevoort, E. J. Wiertz, and R. C. Hoeben. 2007. In cis inhibition of antigen processing by the latency-associated nuclear antigen I of Kaposi sarcoma herpes virus. Mol Immunol 44:1352-60.

75. Zimmermann, M., C. Flechsig, N. La Monica, M. Tripodi, G. Adler, and N. Dikopoulos. 2008. Hepatitis C virus core protein impairs in vitro priming of specific T cell responses by dendritic cells and hepatocytes. J Hepatol 48:51-60.

76. Zuo, J., A. Currin, B. D. Griffin, C. Shannon-Lowe, W. A. Thomas, M. E. Ressing, E. J. Wiertz, and M. Rowe. 2009. The Epstein-Barr virus G-protein-coupled receptor contributes to immune evasion by targeting MHC class I molecules for degradation. PLoS Pathog 5:e1000255.