• No results found

Human virus-specific T cells in peripheral blood and lymph nodes: Phenotype, function and clonal relationships - Chapter 9: General discussion

N/A
N/A
Protected

Academic year: 2021

Share "Human virus-specific T cells in peripheral blood and lymph nodes: Phenotype, function and clonal relationships - Chapter 9: General discussion"

Copied!
17
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

UvA-DARE (Digital Academic Repository)

Human virus-specific T cells in peripheral blood and lymph nodes: Phenotype,

function and clonal relationships

Remmerswaal, E.B.M.

Publication date

2014

Document Version

Final published version

Link to publication

Citation for published version (APA):

Remmerswaal, E. B. M. (2014). Human virus-specific T cells in peripheral blood and lymph

nodes: Phenotype, function and clonal relationships.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s)

and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open

content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please

let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material

inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter

to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You

will be contacted as soon as possible.

(2)
(3)
(4)
(5)

DISCUSSION

9

GENERAL DISCUSSION

This thesis focusses on the characterization of T cell responses against (persistent) viral infections in peripheral blood and lymph nodes. We studied the immune response against the persistent viral infections human cytomegalovirus (hCMV), Epstein Barr virus (EBV) and human immunodeficiency virus (HIV) and compared these with the transient viral infections influenza A and respiratory syncytial virus (RSV).

Differences in virus genome and virion composition, genetic variation of the host (polymorphisms in genes encoding cytokines, HLA-type, expression of chemokine receptors etc), immunocompetence of the host, route and dose of infection and even infection history can all influence the immune response. In mice, all these factors can be standardized and therefore data from these experimental models do provide an invaluable insight in the mechanisms involved in the mammalian immune system. However, the goal of our studies was the characterization of such responses in man. Thus, although inter-individual and inter-viral differences will without doubt be large, we try to find common denominators of anti-viral T cell responses in man.

Because it is usually unknown when an individual becomes infected with a virus, most of the studies performed in humans are performed on T cells in the memory (for transient viral infections) or latent phase (for persistent viral infections). The majority of the data on primary viral infections are derived from murine models. However, the possibility that a naïve renal transplant recipient can receive a graft from a latently infected donor, allows for a unique opportunity to study primary viral infection in humans. The renal transplant setting also provided the possibility to study paired lymph node (LN) and PB samples. These LNs could often be found in the residual tissue that was removed during the anastomosis of the arteria and vena renalis when the iliac artery and vein were dissected free.

At least two parameters might influence the outcome of anti-viral responses in renal transplant patients when compared to healthy individuals. First, these patients are treated with an immunosuppressive drug regimen to prevent allograft rejection. Second, the route and dose of infection in renal transplant patients differs from the way healthy individuals become infected. In healthy individuals epithelial cells will become infected first with cell free virions. Renal transplant patients become infected by virions produced by latently infected cells within the graft once these cells have entered the lytic cycle. These patients can also become infected by cell to cell transfer of virus from graft cells that have entered the lytic cycle. However, in spite of these differences, these patients are usually able to control the viral replication without serious clinical complications and they do produce virus-specific IgM and IgG antibodies upon primary infection (1). Also, although anti-viral CD4+ and CD8+ T cell

responses in renal transplant recipients have shown to be larger, the quality of the response closely resembles that of healthy individuals (2, 3).

HCMV, EBV and HIV all persist (latently) after the primary infection. Although there are similarities between the methods these herpesviridae employ to escape the

(6)

DISCUSSION

9

immune system, there are also a lot of differences. Herpesviridae have co-evolved with their vertebrate hosts for more than 100 million years (4), which has allowed the generation of many immune escape mechanisms by the double-stranded, relatively stable DNA virus. In contrast, HIV-1 has probably been introduced from chimpanzees into the human species some 100 years ago (5, 6). Because of the highly error-prone reverse transcription, the single stranded positive-sense RNA viruses changes upon every cycle and adaptations to its environment occur much faster than will ever be possible in the herpesviridae. Also the tropism of EBV, hCMV and HIV is completely different, which will influence the immune system.

Phenotype of latent virus-specific CD4

+

and CD8

+

T cells

HCMV

The long co-evolution of the herpesviridae with its host has also allowed for many adaptations of the host to the virus. In spite of, or perhaps due to, the HLA-I downregulation, NK evasion and other immune evasive mechanisms, up to half of the total CD8+ T cells in the peripheral blood (PB) of individuals who are latently infected

with hCMV, can be dedicated to hCMV (chapter 5). In fact both cytotoxic CD8+ and

CD4+ T cells, expressing granzyme B and perforin, can be found in significantly higher

proportions in individuals latently infected with hCMV ((7, 8) and chapters 3 and 7). The phenotype of the majority of the latent phase PB hCMV-specific CD4+ and

CD8+ T cells is very distinctive. These effector-type cells contain vast amounts of the

cytolytic molecules granzyme B and perforin and display direct ex vivo cytotoxicity and have lost the co-stimulatory markers CD27 and CD28. We have shown that hCMV reactivation leads to higher numbers of hCMV-specific CD8+ T cells and

CD27 downregulation (9). The CD27 downregulation may be caused by ligation to its ligand CD70, which can be expressed on activated T cells, B cells and dendritic cells. Expression of CD70 on activated T cells was shown to be dependent on the cytokine milieu. Where IL-2 and IL-15 enhanced CD70 upregulation, IL-21 was shown to prevent it (9). Thus, the cytokine environment in response to a viral infection and the repetitive reactivations of a persistent infection can influence the CD27-expression on a responding T cell. Other support for this theory can be found when analyzing hCMV-specific CD8+ T cells in untreated HIV infected individuals. The hCMV-pp65

specific CD8+ T cells in HIV infected individuals express less CD27 than the

hCMV-pp65-specific CD8+ T cells in uninfected individuals.

HCMV can infect a large variety of different cell types like endothelial cells, epithelial cells and leukocytes. Since latency has been shown to occur in monocytes and CD34+

myeloid precursors, and reactivation has been shown to occur upon differentiation, hCMV infected monocyte-derived DCs can be expected to influence the immune response.

Upon T cell activation CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) can be upregulated. CD28 and CTLA4 compete for the same ligands CD80 (B7-1) and CD86

(7)

DISCUSSION

9

(B7-2) on antigen presenting cells. Ligation of CTLA4 leads to downregulation of the response and possibly tolerance by interference with TCR signals and the release of IDO from DCs (10-14). CTLA-4 has also been shown to enhance internalization and degradation of CD28 (15).Ligation of CD28 however leads to enhanced IL-2 production, proliferation and prevention of apoptosis (16, 17).

Latent phase PB hCMV-specific CD4+ and CD8+ T cells also express the chemokine

receptor CX3CR1 (12, 18), which allows them to migrate to fraktalkine (CX3CL1) expressing stressed (infected) endothelium and epithelium cells. Since hCMV-infected cells can also express vCX3CR1 (US28) (19, 20), the hCMV-specific T cells

might employ their CX3CR1 mediated migration to effectively cluster at places towards which infected leukocytes home.

EBV

As shown in chapter 2 and 3 the phenotype of EBNA3a-specific CD8+ T cells differs

from the BZLF1- and BMLF1-specific CD8+ T cells. Resting latent epitope-specific

CD8+ T cells usually still express both CD27 and CD28 and they contain granzyme

K. In contrast, half of the resting lytic cycle-specific CD8+ T cells have lost their CD28

expression and although a large proportion also contains granzyme K, a substantial amount does contain granzyme B.

No data have been presented about inhibition of EBV replication by granzyme K so far. However granzyme K has been shown to inhibit influenza virus replication via the disruption of the importin a1/b dimer, which is required for the transportation of viral NP to the nucleus (21). It has also been shown, just like all other granzymes, to target hnRNP K (heterogeneous nuclear ribonucleoprotein K), a multifunctional DNA/ RNA-binding protein involved in transcription, translation, splicing and mRNAstability. Granzyme M was shown to decrease hCMV-IE2 transcription and cell viability by cleavage of hnRNP K (22). It seems very likely that EBV-replication will be inhibited by similar mechanisms and because of the high percentage of EBV-specific CD8+ T cells

expressing granzyme K, this mechanism is most likely crucial in maintaining latency. In contrast to hCMV the tropism of EBV is rather limited. EBV infects epithelial cells and B cells, and latency can be established in memory B cells. Perhaps the lesser differentiated phenotype of EBV-specific CD8+ T cells can be explained by the fact

that EBV does not infect or affect professional APCs.

HIV

The main targets of HIV are CD4-expressing cells like T cells, dendritic cells, macrophages and monocytes. Although DCs are rather resistant to infection, depending on their C-type lectin expression they can affect the outcome of an HIV infection. DC-sign-expressing DCs can, by uptake of viral particles, infect CD4+ T cells

(8)

DISCUSSION

9

in Birbeck granules, if it was captured by langerin on Langerhans cells, thereby preventing further viral spread.

The individuals we studied in chapter 3 were untreated but had not yet progressed to AIDS. The loss of IL-7R

α

-expression on these cells probably reflects the very frequent engagement of the TCR, since we have shown that such an engagement causes prolonged downregulation of the IL-7R

α

(23). Downregulation due to binding of IL-7 to the IL-7R was shown to be transient.

Temporal context of hCMV and EBV proteins

and CD8

+

T cell responses

The above mentioned phenotype holds true for the latent phase CD8+ T cells against both

hCMV proteins studied: pp65 and IE1. This is quite remarkable since both proteins have different functions, different temporal contexts and they even differ in their presentation in HLA-I. Tegument protein pp65 is present in the virion and can therefore be processed and presented from the moment of infection. The transactivator IE1 however is the first protein to be produced after infection of a cell as the onset of the lytic cycle, but the already present pp65 can prevent presentation of IE1 derived peptides in HLA-I by phosphorylation of the IE1 protein. The balance between the amount of pp65 and IE1 is paramount in this blockade. With higher levels of another tegument protein, pp71, in the virion, IE transcription is enhanced and IE1 derived peptides are found in HLA-I (24). Indeed we have seen that the hCMV-pp65-specific CD8+ T cells often precede the

hCMV-IE1-specific CD8+ T cells (chapter 3 and data not shown) in primary hCMV.

In latently infected cells the IE1 protein marks the onset of hCMV reactivation. The tegument pp65 is a late protein and as such will be produced late during the viral cycle. Prevention of presentation of IE1 derived peptides in HLA-I can therefore not be prevented from the beginning of the reactivation. It has been shown in mice that reactivation of mCMV in lungs occurs frequently in a patchwork-like way (25, 26). It has also been shown that IE1-derived peptides indeed were presented in HLA-I and that IE1-specific CD8+ T cells terminated the mCMV reactivation (27). Thus, the dynamics

of pp65 and IE-derived peptide presentation in HLA-I and the subsequent pp65 and IE-specific CD8+ T cells are different during primary hCMV and reactivation and should

be considered when studying hCMV-specific CD8+ T cells.

It is also crucial to separate the different epitopes the response is directed to, when analyzing the EBV-specific CD8+ T cells. Both the IE and the EBNA proteins are not

present in the virion. The IE protein transactivator BZLF1 and the E protein BMLF1 are produced early in the lytic cycle and will therefore be produced by infected epithelial cells early in the primary infection. The EBNA3 proteins however are only transcribed in some of the latently infected B cells. When studying primary EBV infections we did indeed observe that the lytic-epitope (BZLF1 and BMLF1) specific CD8+ T cells always

preceded the PB EBNA-specific CD8+ T cells (chapter 3 and 6).

(9)

DISCUSSION

9

Clonal breadth of virus-specific CD8

+

T cells

CD8+ T cell clones directed towards hCMV-IE- and pp65-derived epitopes are among

the largest clones in the CD8 compartment (chapter 3). IE-specific and pp65-specific CD8+ T cells however do differ significantly in their clonal breadth. In chapter 3 we show

that hCMV-pp65-specific CD8+ T cells generally consist of substantially more clones than

hCMV-IE specific CD8+ T cells. Exceptions to this rule do exist (chapter 3 and chapter 6) as

we have seen healthy individuals and renal transplant patients who mounted a monoclonal response against an hCMV-pp65 derived peptide and we have found one renal transplant patient who elicited a relatively broad response against an hCMV-IE epitope.

Indeed when hCMV-IE specific CD8+ T cells appear only late during the primary

hCMV, the cytokine milieu, availability of nutrients, co-stimulation, abundance of cognate peptide amongst others will differ considerably. For example, the pro-apoptotic protein Noxa has been shown to be upregulated at times of nutrient and cytokine deprivation and the absence of Noxa has been shown to result in enhanced diversity of the effector pool (28). Thus one might envision that during the onset of the immune response, when all nutrients and cytokines are abundant, Noxa will be low and lower affinity clones can expand too. In contrast at the height of the immune response competition for nutrients and cytokines will occur; Noxa will be upregulated and only higher affinity clones will expand. Thus, the early pp65-specific clones might be more diverse in their affinity and their clonal repertoire than the IE-specific clones. And when the hCMV-IE response is broad, it could be the result of an early hCMV-IE response, possibly caused by a pp71 favored balance of tegument proteins. Another reason for the difference in clonal breadth might be the fact that latently infected monocytes can differentiate into dendritic cells. Differentiation will lead to the onset of viral replication and the hCMV immune escape mechanism within these DCs are likely to influence both the clonal breadth and the resulting phenotype.

The difference in clonal breadth that was observed in the CD8 response towards hCMV-proteins is similar to that of EBNA-specific clones when compared to the BZLF-specific clones. CD8+ T cell responses towards EBNA3a and b derived peptides tend

to consist of fewer clones than those directed towards BZLF1 derived peptides. Once again, this might be related to the elapsed time after the onset of viral replication, but also on the cell type presenting the antigens.

Clonal evolution of CD8

+

T cell responses

Primary infection

We show in chapter 3 that the clonal repertoire of both CD8+ T cells directed against

EBV-lytic and latent epitopes as well as hCMV-IE and pp65-specific CD8+ T cells remains

rather stable for over more than 3 years after the primary infection. Others have shown that the acute phase response focusses early during the primary infection, leaving a more restricted repertoire in latency (29). For this last study, samples as early as two weeks after

(10)

DISCUSSION

9

infection were analyzed, whereas in chapter 3 we analyzed the samples once the tetramer positive population was big enough to detect the individual clones within the total CD8+

population. Thus, we might have missed this early focussing. On the other hand, when frequencies are so very low, the risk of impurities in the sorts are also more substantial and in the days before next generation sequencing, these impurities were harder to exclude in the analyses, due to the many separate PCR’s that had to be performed.

In chapter 6 we studied the primary hCMV-pp65 response in two additional patients. This time, we did split the IL-7R

α

-expressing and negative hCMV-specific CD8+ T cells. In mice it was shown that the IL-7R

α

-expressing population contained

the long-lived precursor cells. We sorted the first possible time point at which sufficient IL-7R

α

-expressing cells were present. These time points were earlier in the response than the time points taken in chapter 3 and the patients were different.

In chapter 6 we showed that IL-7R

α

expressing hCMV-pp65-specific CD8+ T cells

from the acute phase of the response are not the precursors for the latent phase T cells. Intuitively this seems logical. Circulating hCMV-specific cells would be exposed to their antigen with every reactivation that results in viral replication, leading to depletion or exhaustion of the precursor pool. The buffered memory theory, suggesting a location where such a precursor pool would be hidden from antigenic challenges unless the antigenic burden was large enough to reach such a site offers a reasonable theory (30).

Reactivation / reinfection

Endogenous reactivation of hCMV can occur because of the immunosuppressive treatment of the renal transplant patient, which requires adjustments of the immune response to maintain control of virus. However, when the donor also is latently infected with hCMV, infection with the donor derived virus can occur too.

As explained above, the IE1 protein is the first to be produced upon viral reactivation and generally these reactivations is effectively controlled by T cells before the actual production of new virions. However, such reactivations will be hard to study, since one can not detect a viral load. We have studied renal transplant recipients who were latently infected before transplantation and in whom we could measure hCMV-DNA in the peripheral blood compartment (chapter 6).

In cases where the donor was latently infected with hCMV too, reactivation of hCMV in donor derived cells would require the recognition of the HLA-epitope combination on the donor derived cells. It will therefore depend on the matching between recipient and donor HLA and the HLA-peptide combinations against which the recipient has mounted a response. If the recipient’s hCMV-IE-specific CD8+ T cells can not recognize the

reactivating donor derived cells, they will be unable to eliminate them before the release of new virions. In such cases, the interplay between pp65 and pp71 with IE1 in the newly infected recipients cells, will once again determine the time of response of CD8+ T cells.

This might account for the lack of expansion of IE1-specific CD8+ T cell responses at the

(11)

DISCUSSION

9

moment of hCMV replication when compared to the pp65-directed responses. However, the IE1-specific CD8+ T cells in these patients can respond to suppress the endogenous

reactivation of their own cells even before viral replication occurs.

We showed that upon viral reactivation or reinfection, apparently new clones can be recruited into the PB hCMV-IE- and pp65-specific CD8+ T cell pool, depending

on the breadth of the pre-transplantation pool. If a hCMV-specific response was highly restricted before reactivation it remained monoclonal, suggesting that these individuals either mounted a very adequate response during primary infection or that the T cells directed against this epitope are actually not involved (epitope is not (yet) presented, or T cells are exhausted). We have shown that when hCMV-specific CD8+ T cells are stimulated with their cognate antigen in the optimal milieu, they

can proliferate, produce cytokines and effectively kill the loaded target (31). Thus, exhaustion of these cells seems an unlikely option.

The fact that the monoclonal pp65-specific CD8+ T cells did become activated and

expanded in the PB suggests that these cells did see their antigen. That leaves the possibility of the optimal monoclonal response during primary infection, implying that a broad repertoire might not be optimal. The recruitment of new clones as a result of the hCMV-reactivation in patients who had an oligoclonal CD8+ T cell response

before reactivation also suggest that the pre transplantation clones were not optimal. However, many healthy donors do have oligoclonal CD8+ T cell responses, and they

do not experience major adverse events of the hCMV infection.

It has been shown that a higher percentage of the CD45RA re-expressing effector-type hCMV-pp65-specific CD8+ T cells when compared to their CD45R0-expressing

counterparts had a lower antigen binding affinity (32). This was especially seen in elderly individuals and depended on the size of the hCMV-specific CD8+ T cell pool

(32). It was also shown that this CD45RA re-expression could occur (at least in vitro) by exposure to IL-15 possibly produced by monocytes (32).

We have shown that hCMV reactivation caused both downregulation of CD27 and expansion of the pre-existing pool of hCMV-pp65-specific CD8+ T cells (9). Moreover

the size of the effector-type cell pool within the total hCMV-pp65 CD8+ T cells has

been shown to correlate with the total size of this pool (9). Thus it seems likely that upon every reactivation either more lower antigen binding affinity cells are recruited into the PB hCMV-pp65 specific CD8+ T cell pool, or the lower affinity hCMV-specific

CD8+ T cells are expanding or surviving relatively more than the higher affinity

hCMV-pp65-specific CD8+ T cells.

The first option would confirm the recruitment of new clones into the PB especially in individuals who already have a broader clonal repertoire. The fact that we sometimes see some small new clones appearing in individuals with a monoclonal response before reactivation might be the onset of such a broader repertoire.

The second option would suggest that the high affinity clones would be lost over time, either due to the fact that get outgrown or because they die after being activated.

(12)

DISCUSSION

9

The affinity data need to be confirmed for other hCMV-specific CD8+ T cells like

IE1-specific CD8+ T cells.

Virus-specific CD8

+

T cells in lymph nodes

Where virus-specific CD8+ T cells in PB can be strikingly different depending on the virus

or even the protein against which they have been generated, hCMV-, EBV and influenza A-specific CD8+ T cells in LNs actually quite resemble each other in number, phenotype

and function (chapter 5). We have also shown that LNs actually contained unique hCMV-, EBV and influenza A-specific CD8+ T clones that could not be found in the PB.

LN are required for the priming of naïve T cells, but lesser so during memory responses and reactivations as antigen experienced T cells do not need professional APCs to become activated. These data might suggest that virus-specific CD8+ T

cells in LN either form a reservoir for memory cells, shielded from constant antigenic stimulation, or that they have a special function within the lymph node. In chapter 7 we showed that the LN probably may not function as such a memory cell reservoir, which suggests that these cells indeed are tissue resident T cells, with a possible local function. One such function could be the elimination of dendritic cells that present epitopes that are already efficiently recognized by the existing pool of antigen experienced cells. Regardless of the phenotype of a resting antigen-experienced cell one can expect that upon reengagement of the T cell with its antigen, cytotoxic molecules, like granzyme B and perforin, will be upregulated, allowing them to effectively kill the antigen expressing cell. It has been suggested before that homing of CXCR3-expressing T cells to draining lymph nodes could indeed abrogate the ongoing immune response after a primary infection by eliminating the antigen presenting DCs (33). Thus, it seems possible that these cells remain and prevent a second round of activation of naive T cells upon reactivation.

Of course the question remains if a memory cell pool shielded from the PB does exist and if so, where this buffered memory pool would be located. Based on previous publications, two locations could harbor such a pool. The first one is the spleen and the second the bone marrow. HCMV-specific CD8+ T cells have been found in both

tissues (34-36). However, the phenotype and functions of the hCMV-specific CD8+ T

cells in these tissues still need thorough investigation. In mice it was shown that CCR7-expressing memory cells were able to home to LN and the white pulpa of the spleen, whereas CCR7-negative cells occupied the red pulpa (37, 38). An interesting feature of the hCMV-specific CD8+ T cells in the bone marrow is the possible presence of the

latently infected CD34+ myeloid precursors which might actually reside there. Further

research on the phenotype, function and the clonal relationships of the hCMV and EBV-specific CD8+ T cells compared to their PB counterparts will give further insight

into the sustenance of immune responses against persistent viral infections.

(13)

DISCUSSION

9

Cytotoxic hCMV-specific CD4

+

T cells

Recently it has been shown that cytotoxic CD4+ T cells differ from the CD4+ T helper

cells with respect to the expression of the T helper (Th) lineage master regulator, the Th-inducing BTB/POZ domain-containing Kruppel-like zinc-finger transcription factor, ThPOK (39). In CD8+ T cells ThPOK expression is repressed in the thymus by the

expression of RUNX3 (runt-related transcription factor 3). In cytotoxic CD4+ T cells

however the loss of ThPOK expression occurs post thymically as a result of repeated stimulation with their cognate antigen in vivo (39). RUNX3 is upregulated in activated CD4+ T cells by T-Bet and promotes the Th1 program and subsequent IFN

γ

production

by CD4+ T cells. The loss of ThPOK expression leads to the de-repression of the

cytolytic-gene expression program, which causes the cytolytic protein expression (40). Other indications that cytotoxic hCMV-specific CD4+ T cells emerge as a result

of repetitive antigenic stimulation are the loss of CD27 and the fact that we observe significantly higher percentages of these cells in immunocompromised patients when compared to healthy donors.

In chapter 7 and 8 we showed that CD28

CD4+ T cells are hCMV-specific CD4+

T cells (chapter 7 and 8). They could only be found in individuals latently infected with hCMV, appeared after primary hCMV infection, produced IFN

γ

and proliferated after stimulation with antigen, and displayed ex vivo cytotoxicity against hCMV-antigen or pp65-derived peptide-loaded target cells. Phenotypically and functionally they resembled their hCMV-specific CD8+ T cell counterparts. They contained

granzyme B and perforin, lost their expression of CD27 and CD28, and do express CX3CR1 (18). They also produced IFN

γ

, TNF

α

, MIP-1

β

, but no IL-2 and expressed CD107a after stimulation with their cognate peptide (41). Lastly, the cytotoxic CD4+

T cells also had a very limited clonal repertoire, which overlapped minimally with the acute phase IFN

γ

-producing CD4+ T cells (chapter 8).

We have shown that at least part of these cells recognize a pp65 derived epitope. It has also been shown that gB (Glycoprotein B, involved in cell entry)-specific CD4+

T cells display direct ex-vivo cytotoxicity towards endogenous gB but not exogenous gB presented by epithelial cells, endothelial cells and glial cells via autophagy (42). GB-specific CD4+ T cells can lyse lytically infected monocytes, but not latently infected

monocytes (43). Also HCMV-IE specific CD4+ T cells have been shown to produce IFN

γ

.

We tested the specificity of the CD28

CD4+ T cell by stimulation with hCMV-Ag,

and showed that not all cells proliferated or produced antigen. A drawback of the use of antigen is that it will enter the exogenous presentation route, and since it was shown that only endogenous gB was recognized by gB-specific CD4+ T cells, the

gB-specific CD4+ T cells will not recognize the hCMV-antigen loaded target cells and thus

not proliferate, produce cytokine and lyse the loaded target cell. Responses against other hCMV-proteins might be affected in a similar fashion.

In addition, the hCMV-antigen we used is derived from the MRC-5 cells infected with the hCMV AD169 strain (Microbix Biosystems Inc). This strain has been shown to have

(14)

DISCUSSION

9

a 1.5kb deletion in its genome (UL133-151), when compared to the clinical strains. The deleted genes encode proteins that are involved in immune evasion, viral chemokines and proteins that determine cell type specific outcome of infection. This implies that at least part of the CD28

CD4+ T cells might be directed against peptides derived from one or

more of these, in AD169 absent, proteins. Indeed it has been shown that UL138 (a protein that mediates suppression of viral replication (43))-specific CD4+ T cells do exist in healthy

hCMV seropositive subjects (44). These UL138-specific CD4+ T cells were shown to indeed

display HLA-II restricted cytotoxicity and produced IFN

γ

upon stimulation.

Thus, some of the acute phase lytic epitope (like pp65, gB and IE)-specific hCMV-specific CD28+CD4+ T clones and the somewhat later appearing latent epitope (like

UL138) specific hCMV-specific CD4+ T cell clones might give rise to the cytotoxic

CD28

CD4+ T cells upon repetitive exposure to their cognate antigen.

OVERALL CONCLUSION

The combination of multifluorochrome flowcytometry and next generation sequencing used in this thesis shows that it is possible to characterize many aspects of the human anti-viral immune response.

Thus, this thesis paves the way for a better understanding of the phenotype, function and clonal relationships of virus-specific CD4+ and CD8+ T cells directed against many more

proteins derived from many more viruses in a wide variety of tissues. The fact that an ever smaller sample can be used to perform such an analysis without loss of information, will allow for the study of virus-specific CD4+ and CD8+ T cells with extremely low frequencies

and the analysis of anti-viral responses in small samples such as biopsies.

(15)

DISCUSSION

9

1. Rentenaar RJ, Gamadia LE, van der Hoek N, van Diepen FN, Boom R, Weel JF, Wertheim-van Dillen PM, van Lier RA, and ten Berge IJ. Development of virus-specific CD4(+) T cells during primary cytomegalovirus infection. The Journal of

clinical investigation. 2000;105(4):541-8.

2. Gamadia LE, Rentenaar RJ, Baars PA, Remmerswaal EB, Surachno S, Weel JF, Toebes M, Schumacher TN, ten Berge IJ, and van Lier RA. Differentiation of cytomegalovirus-specific CD8(+) T cells in healthy and immunosuppressed virus carriers. Blood. 2001;98(3):754-61.

3. van Leeuwen EM, Remmerswaal EB, Vossen MT, Rowshani AT, Wertheim-van Dillen PM, van Lier RA, and ten Berge IJ. Emergence of a CD4+CD28- granzyme B+, cytomegalovirus-specific T cell subset after recovery of primary cytomegalovirus infection. Journal of immunology (Baltimore,

Md : 1950). 2004;173(3):1834-41.

4. McGeoch DJ, Dolan A, and Ralph AC. Toward a comprehensive phylogeny for mammalian and avian herpesviruses.

Journal of virology. 2000;74(22):10401-6.

5. Worobey M, Gemmel M, Teuwen DE, Haselkorn T, Kunstman K, Bunce M, Muyembe JJ, Kabongo JM, Kalengayi RM, Van Marck E, et al. Direct evidence of extensive diversity of HIV-1 in Kinshasa by 1960. Nature. 2008;455(7213):661-4. 6. Sharp PM, and Hahn BH. Origins of HIV

and the AIDS pandemic. Cold Spring

Harbor perspectives in medicine.

2011;1(1):a006841.

7. Bekker V, Bronke C, Scherpbier HJ, Weel JF, Jurriaans S, Wertheim-van Dillen PM, van Leth F, Lange JM, Tesselaar K, van Baarle D, et al. Cytomegalovirus rather than HIV triggers the outgrowth of effector CD8+CD45RA+CD27- T cells in HIV-1-infected children. AIDS (London,

England). 2005;19(10):1025-34.

8. Kuijpers TW, Vossen MT, Gent MR, Davin JC, Roos MT, Wertheim-van Dillen PM, Weel JF, Baars PA, and van Lier RA. Frequencies of circulating cytolytic, CD45RA+CD27-, CD8+ T lymphocytes depend on infection with CMV. Journal

of immunology (Baltimore, Md : 1950).

2003;170(8):4342-8.

9. Gamadia LE, van Leeuwen EM, Remmerswaal EB, Yong SL, Surachno S,

Wertheim-van Dillen PM, ten Berge IJ, and van Lier RA. The size and phenotype of virus-specific T cell populations is determined by repetitive antigenic stimulation and environmental cytokines.

JImmunol. 2004;172(10):6107-14.

10. Thompson CB, and Allison JP. The emerging role of CTLA-4 as an immune attenuator. Immunity. 1997;7(4):445-50. 11. Munn DH, Sharma MD, and Mellor

AL. Ligation of B7-1/B7-2 by human CD4+ T cells triggers indoleamine 2,3-dioxygenase activity in dendritic cells.

Journal of immunology (Baltimore, Md : 1950). 2004;172(7):4100-10.

12. Hertoghs KM, Moerland PD, van Stijn A, Remmerswaal EB, Yong SL, van de Berg PJ, van Ham SM, Baas F, ten Berge IJ, and van Lier RA. Molecular profiling of cytomegalovirus-induced human CD8+ T cell differentiation. The Journal of clinical

investigation. 2010;120(11):4077-90.

13. Linsley PS, Bradshaw J, Greene J, Peach R, Bennett KL, and Mittler RS. Intracellular trafficking of CTLA-4 and focal localization towards sites of TCR engagement.

Immunity. 1996;4(6):535-43.

14. Yao S, Zhu Y, Zhu G, Augustine M, Zheng L, Goode DJ, Broadwater M, Ruff W, Flies S, Xu H, et al. B7-h2 is a costimulatory ligand for CD28 in human. Immunity. 2011;34(5):729-40.

15. Berg M, and Zavazava N. Regulation of CD28 expression on CD8+ T cells by CTLA-4. Journal of leukocyte biology. 2008;83(4):853-63.

16. Radvanyi LG, Shi Y, Vaziri H, Sharma A, Dhala R, Mills GB, and Miller RG. CD28 costimulation inhibits TCR-induced apoptosis during a primary T cell response.

Journal of immunology (Baltimore, Md : 1950). 1996;156(5):1788-98.

17. Bluestone JA. New perspectives of CD28-B7-mediated T cell costimulation.

Immunity. 1995;2(6):555-9.

18. van de Berg PJ, Yong SL, Remmerswaal EB, van Lier RA, and ten Berge IJ. Cytomegalovirus-induced effector T cells cause endothelial cell damage.

ClinVaccine Immunol. 2012;19(5):772-9.

19. Noriega VM, Gardner TJ, Redmann V, Bongers G, Lira SA, and Tortorella D. Human cytomegalovirus US28 facilitates

(16)

DISCUSSION

9

cell-to-cell viral dissemination. Viruses. 2014;6(3):1202-18.

20. Hjorto GM, Kiilerich-Pedersen K, Selmeczi D, Kledal TN, and Larsen NB. Human cytomegalovirus chemokine receptor US28 induces migration of cells on a CX3CL1-presenting surface. The Journal of general

virology. 2013;94(Pt 5):1111-20.

21. Zhong C, Li C, Wang X, Toyoda T, Gao G, and Fan Z. Granzyme K inhibits replication of influenza virus through cleaving the nuclear transport complex importin alpha1/ beta dimer of infected host cells. Cell death

and differentiation. 2012;19(5):882-90.

22. van Domselaar R, de Poot SA, Remmerswaal EB, Lai KW, ten Berge IJ, and Bovenschen N. Granzyme M targets host cell hnRNP K that is essential for human cytomegalovirus replication. Cell death and differentiation. 2013;20(3):419-29.

23. Alves NL, van Leeuwen EM, Derks IA, and van Lier RA. Differential regulation of human IL-7 receptor alpha expression by IL-7 and TCR signaling. JImmunol. 2008;180(8):5201-10.

24. Hesse J, Reyda S, Tenzer S, Besold K, Reuter N, Krauter S, Buscher N, Stamminger T, and Plachter B. Human cytomegalovirus pp71 stimulates major histocompatibility complex class i presentation of IE1-derived peptides at immediate early times of infection. Journal of virology. 2013;87(9):5229-38.

25. Kurz SK, Rapp M, Steffens HP, Grzimek NK, Schmalz S, and Reddehase MJ. Focal transcriptional activity of murine cytomegalovirus during latency in the lungs.

Journal of virology. 1999;73(1):482-94.

26. Kurz SK, and Reddehase MJ. Patchwork pattern of transcriptional reactivation in the lungs indicates sequential checkpoints in the transition from murine cytomegalovirus latency to recurrence.

Journal of virology. 1999;73(10):8612-22.

27. Simon CO, Holtappels R, Tervo HM, Bohm V, Daubner T, Oehrlein-Karpi SA, Kuhnapfel B, Renzaho A, Strand D, Podlech J, et al. CD8 T cells control cytomegalovirus latency by epitope-specific sensing of transcriptional reactivation. Journal of

virology. 2006;80(21):10436-56.

28. Wensveen FM, Klarenbeek PL, van Gisbergen KP, Pascutti MF, Derks IA, van Schaik BD, Ten

Brinke A, de Vries N, Cekinovic D, Jonjic S, et al. Pro-apoptotic protein Noxa regulates memory T cell population size and protects against lethal immunopathology. Journal

of immunology (Baltimore, Md : 1950).

2013;190(3):1180-91.

29. Day EK, Carmichael AJ, ten Berge IJ, Waller EC, Sissons JG, and Wills MR. Rapid CD8+ T cell repertoire focusing and selection of high-affinity clones into memory following primary infection with a persistent human virus: human cytomegalovirus. Journal

of immunology (Baltimore, Md : 1950).

2007;179(5):3203-13.

30. Snyder CM. Buffered memory: a hypothesis for the maintenance of functional, virus-specific CD8(+) T cells during cytomegalovirus infection.

ImmunolRes. 2011;51(2-3):195-204.

31. van Leeuwen EM, Gamadia LE, Baars PA, Remmerswaal EB, ten Berge IJ, and van Lier RA. Proliferation requirements of cytomegalovirus-specific, effector-type human CD8+ T cells. JImmunol. 2002;169(10):5838-43.

32. Griffiths SJ, Riddell NE, Masters J, Libri V, Henson SM, Wertheimer A, Wallace D, Sims S, Rivino L, Larbi A, et al. Age-associated increase of low-avidity cytomegalovirus-specific CD8+ T cells that re-express CD45RA. JImmunol. 2013;190(11):5363-72. 33. Guarda G, Hons M, Soriano SF, Huang

AY, Polley R, Martin-Fontecha A, Stein JV, Germain RN, Lanzavecchia A, and Sallusto F. L-selectin-negative CCR7- effector and memory CD8+ T cells enter reactive lymph nodes and kill dendritic cells. Nature

immunology. 2007;8(7):743-52.

34. Langeveld M, Gamadia LE, and ten Berge IJ. T-lymphocyte subset distribution in human spleen. EurJClinInvest. 2006;36(4):250-6. 35. Letsch A, Knoedler M, Na IK, Kern F,

Asemissen AM, Keilholz U, Loesch M, Thiel E, Volk HD, and Scheibenbogen C. CMV-specific central memory T cells reside in bone marrow. European journal

of immunology. 2007;37(11):3063-8.

36. Palendira U, Chinn R, Raza W, Piper K, Pratt G, Machado L, Bell A, Khan N, Hislop AD, Steyn R, et al. Selective accumulation of virus-specific CD8+ T cells with unique homing phenotype within the human bone marrow. Blood. 2008;112(8):3293-302.

(17)

DISCUSSION

9

37. Potsch C, Vohringer D, and Pircher H. Distinct migration patterns of naive and effector CD8 T cells in the spleen: correlation with CCR7 receptor expression and chemokine reactivity. European journal

of immunology. 1999;29(11):3562-70.

38. Jung YW, Rutishauser RL, Joshi NS, Haberman AM, and Kaech SM. Differential localization of effector and memory CD8 T cell subsets in lymphoid organs during acute viral infection. Journal of

immunology (Baltimore, Md : 1950).

2010;185(9):5315-25.

39. Mucida D, Husain MM, Muroi S, van Wijk F, Shinnakasu R, Naoe Y, Reis BS, Huang Y, Lambolez F, Docherty M, et al. Transcriptional reprogramming of mature CD4(+) helper T cells generates distinct MHC class II-restricted cytotoxic T lymphocytes. Nature immunology. 2013;14(3):281-9.

40. Djuretic IM, Levanon D, Negreanu V, Groner Y, Rao A, and Ansel KM. Transcription factors T-bet and Runx3 cooperate to activate Ifng and silence Il4 in T helper type 1 cells. Nature

immunology. 2007;8(2):145-53.

41. Casazza JP, Betts MR, Price DA, Precopio ML, Ruff LE, Brenchley JM, Hill BJ, Roederer M, Douek DC, and Koup RA. Acquisition of direct antiviral effector functions by CMV-specific CD4+ T lymphocytes with cellular maturation. The Journal of experimental

medicine. 2006;203(13):2865-77.

42. Hegde NR, Dunn C, Lewinsohn DM, Jarvis MA, Nelson JA, and Johnson DC. Endogenous human cytomegalovirus gB is presented efficiently by MHC class II molecules to CD4+ CTL. The Journal of

experimental medicine.

2005;202(8):1109-19.

43. Umashankar M, Rak M, Bughio F, Zagallo P, Caviness K, and Goodrum FD. Antagonistic determinants controlling replicative and latent states of human cytomegalovirus infection. Journal of

virology. 2014;88(11):5987-6002.

44. Mason GM, Jackson S, Okecha G, Poole E, Sissons JG, Sinclair J, and Wills MR. Human cytomegalovirus latency-associated proteins elicit immune-suppressive IL-10 producing CD4(+) T cells. PLoS pathogens. 2013;9(10):e1003635.

Referenties

GERELATEERDE DOCUMENTEN

T Y P EE I: Target-- compound d analysis s Typee II: Group-type e analysis s T Y P EE III: Fingerprinting g Applicationn requirements Highh peak capacity Reliablee component

This standard was treated as a sample and thee concentrations were calculated for each component with integration, Parafac,, Parafac2, and NPLS.. Ideally, the

Quantification of such component groups,, which is a very important aspect of group-type analysis, requires quantificationn tools different from those typically used for

Although thee score plots obtained by the two techniques showed much resemblance, thee percentage of variance captured in the first PC (from PCA) or factor (fromm Parafac) was

Small differences between the samples are blown out off proportion.Projection pursuit yielded a somewhat improved clustering results,, as shown in Figure 6.9.The observation that

Op deze manier kunnen verhingen die zorgen voor het slechtt presteren van een product worden achterhaald.. Hett classificatie-schema moet zowel de ontwikkelaars van nieuwe

Jouw deur stond altijd open en jij had eigenlijkk altijd wel tijd voor mij.. Ik moet wel toegeven dat werkbesprekingen omm acht uur 's morgens zonder koffie geen eenvoudige

Trilinear chemometric analysiss of two-dimensional comprehensive gas chromatography coupled to time-of- flightflight mass spectrometry data.. Johnson