ArticlePDF Available

Functional Impairment of Simian Immunodeficiency Virus-Specific CD8+ T Cells during the Chronic Phase of Infection

American Society for Microbiology
Journal of Virology
Authors:

Abstract and Figures

In an attempt to determine why high frequencies of circulating virus-specific CD8+ T cells are unable to control human immunodeficiency virus and simian immunodeficiency virus (SIV) replication, we assessed the functional nature of SIV-specific CD8+ lymphocytes. After vaccination and early after infection, nearly all tetramer-staining CD8+ cells produced gamma interferon in response to their specific stimulus. However, by 4 months postinfection with pathogenic SIVmac239, signs of functional impairment in the CD8+ T-cell compartment were detected which might prevent these T cells from efficiently controlling the infection during the chronic phase.
Content may be subject to copyright.
JOURNAL OF VIROLOGY,
0022-538X/01/$04.000 DOI: 10.1128/JVI.75.5.2458–2461.2001
Mar. 2001, p. 2458–2461 Vol. 75, No. 5
Copyright © 2001, American Society for Microbiology. All Rights Reserved.
Functional Impairment of Simian Immunodeficiency Virus-
Specific CD8
T Cells during the Chronic Phase of Infection
THORSTEN U. VOGEL,
1
TODD M. ALLEN,
1
JOHN D. ALTMAN,
2
AND DAVID I. WATKINS
1,3
*
Wisconsin Regional Primate Research Center
1
and Department of Pathology and Laboratory Medicine,
3
University of
Wisconsin, Madison, Wisconsin 53715, and Emory Vaccine Center, Emory University School of Medicine,
Atlanta, Georgia 30322
2
Received 2 October 2000/Accepted 7 December 2000
In an attempt to determine why high frequencies of circulating virus-specific CD8
T cells are unable to
control human immunodeficiency virus and simian immunodeficiency virus (SIV) replication, we assessed the
functional nature of SIV-specific CD8
lymphocytes. After vaccination and early after infection, nearly all
tetramer-staining CD8
cells produced gamma interferon in response to their specific stimulus. However, by
4 months postinfection with pathogenic SIVmac239, signs of functional impairment in the CD8
T-cell
compartment were detected which might prevent these T cells from efficiently controlling the infection during
the chronic phase.
It is still unclear why the immune system is not able to clear
an infection with immunodeficiency viruses. These lentiviruses
appear to have devised multiple strategies to evade the im-
mune response (reviewed in reference 26), including major
histocompatibility complex class I downregulation (10) and
escape from cytotoxic T lymphocyte (CTL) responses (1, 7, 9,
13, 17, 27). However, the maintenance of high frequencies of
virus-specific cells against certain viral epitopes in human im-
munodeficiency virus (HIV)-infected humans (4, 23, 30) and
simian immunodeficiency virus (SIV)-infected monkeys (19,
20) indicates that these epitopes are still being recognized and
have not mutated. While the invention of tetramers (4) made
it possible to detect these high frequencies of virus-specific
cells in HIV and SIV infection, tetramer staining simply iden-
tifies antigen-specific lymphocytes but does not provide infor-
mation about the functional nature of these virus-specific cells.
Functionally impaired CD8
T-cell responses have been pre-
viously described by Zajac et al. in chronic lymphocytic cho-
riomeningitis virus (LCMV) infection (31) and by Lee et al. in
a tumor system (22), where CTL were unable to directly lyse
their specific target cells and produce cytokines in response to
mitogens. We therefore were interested in investigating
whether similar functional defects could account for the inabil-
ity of CD8
T cells to control HIV and SIV infections. To
address this question we combined tetramer-staining technol-
ogy (4) and the intracellular cytokine assay (18, 25) to deter-
mine whether SIV-specific CD8
T cells manifest any func-
tional defects.
The majority of tetramer-positive cells produce IFN-after
peptide-specific stimulation in vaccinated animals and early
after infection with pathogenic SIVmac239. Using an epitope-
based DNA prime-modified vaccinia virus Ankara boost vac-
cine, we induced Mamu-A*01-restricted, p11C,C-M (CM9)-
specific CTL (2) in three Mamu-A*01-positive rhesus
macaques (95058, 95045, and 96031), as previously described
(3). We followed the levels of Mamu-A*01/CM9-specific cells
in these animals before and after intrarectal infection with
SIVmac239 (molecular clone) by tetramer staining and inves-
tigated their ability to produce gamma interferon (IFN-) after
antigen-specific stimulation using the intracellular cytokine as-
say. Fresh or thawed peripheral blood mononuclear cells (PB-
MCs) were stimulated for 6.5 h with mitogen (50 ng of phorbol
myristate acetate/ml and 1 g of ionomycin/ml) or with 5 M
CM9 peptide in the presence of B-lymphoblastoid cell line
cells (B-LCL) as antigen-presenting cells. Brefeldin A (10 g/
ml) was present for the last5htoinhibit the secretion of any
produced cytokines. The cells were then surface stained with
anti-CD8 antibodies (conjugated to peridinin chlorophyll pro-
tein; Becton-Dickinson) alone or together with the Mamu-
A*01/CM9 tetramer (labeled with phycoerythrin) for 40 min at
room temperature. The cells were then washed with flow buffer
(2% fetal calf serum in phosphate-buffered saline), fixed with
paraformaldehyde (2% in phosphate-buffered saline) over-
night, permeabilized with 0.1% saponin, and stained intracel-
lularly with anti-IFN-antibodies (labeled with fluoroscein
isothiocyanate; Pharmingen), as described previously (3). As
the T-cell receptor is downregulated after antigen-specific
stimulation (29), the tetramer staining of CM9-specific cells
nearly completely disappeared following CM9-specific stimu-
lation. Therefore, we were not able to express the percentage
of tetramer-positive cells able to produce IFN-. However, we
observed that in immunized animals and early after infection,
the percentage of CD8
cells expressing IFN-after peptide-
specific stimulation correlated with the levels of tetramer stain-
ing in unstimulated samples (Fig. 1; see also reference 3). All
tetramer-positive lymphocytes produced IFN-at weeks 3, 0,
and 2 postchallenge (the ratio of IFN--producing cells to
tetramer-positive cells was approximately 1). We also observed
this correlation between the intracellular cytokine assay and
tetramer staining in other immunized animals and for another
Mamu-A*01-restricted epitope (data not shown). Between
weeks 6 to 8 postchallenge, some tetramer-positive cells from
* Corresponding author. Mailing address: Wisconsin Regional Pri-
mate Research Center, 1220 Capitol Ct., Madison, WI 53715. Phone:
(608) 265-3380. Fax: (608) 265-8084. E-mail: watkins@primate.wisc
.edu.
2458
all three animals (95058, 95045, and 96031) were unable to
produce IFN-(ratios below 1). However, by week 16 the
ratios came back up to values around or higher than 1. As this
drop was only temporary, it is not clear if this represents a
significant change in phenotype and could indicate a functional
impairment of tetramer-positive cells shortly after the primary
peak of viremia reached its high point in week 3 (Allen et al.,
unpublished observations). However, the two naive control
animals (95114 and 95115), which were infected with the same
virus at the same time as the immunized animals (Allen et al.,
unpublished), also evidenced a reduced ratio during the time
of primary viremia, which peaked at week 4 postchallenge.
This may indicate an impaired ability of some tetramer-posi-
tive cells to produce IFN-shortly after peak viral replication
was resolved in these animals. Nevertheless, the ratios re-
turned to values of approximately 1 in week 16 in these two
control animals (Fig. 1). These results indicate that the major-
ity of tetramer-positive cells produce IFN-in response to
stimulation with their cognate peptide up to 4 months after
infection with pathogenic SIV.
Discrepancy between tetramer staining and intracellular
cytokine production after 4 months of infection with patho-
genic SIVmac239. Unexpectedly, the number of tetramer-pos-
itive cells that produced IFN-in response to peptide-specific
stimulation dropped dramatically after 4 months of infection
(Fig. 1). The ratio of IFN--positive cells to tetramer-positive
cells dropped below 0.66 in all animals and remained relatively
stable at these lower values thereafter. There was no obvious
difference between previously immunized and naive control
animals. The possibility that the reduced cytokine production
is caused by a destruction of antigen-presenting cells was ex-
cluded, because B-LCL were added as antigen-presenting cells
in the intracellular cytokine assay. It is, therefore, likely that
the virus-specific cells during chronic SIV infection have de-
fects in cytokine production in response to stimulation by their
cognate peptide. Interestingly, we were able to demonstrate
that most of the tetramer-positive cells in our SIV-infected
macaques were still able to produce IFN-after mitogen stim-
ulation at week 24 (data not shown) and at week 36 postchal-
lenge (Fig. 2). This contrasts with the “silent” phenotype de-
scribed by Zajac et al. in chronic LCMV infection (31) and by
Lee et al. in a tumor system (22). Therefore, the defect of the
virus-specific cells in our monkeys is not as prominent, at least
at this early stage of infection with SIV, as the defect observed
by Zajac and colleagues in LCMV-infected mice (31). It is
interesting that Donahoe and colleagues found a good corre-
lation of intracellular cytokine production after peptide-spe-
cific stimulation, in this case tumor necrosis factor alpha, and
Mamu-A*01/CM9-tetramer staining in monkeys infected with
an attenuated, nonpathogenic SIV, SIVmac239delta nef (11).
It is possible that tetramer-positive cells in animals infected
with an apathogenic SIV might not demonstrate a reduced
ability of cytokine production as found in our animals, but this
needs to be investigated further.
Similarities to functional defects of LCMV- and HIV-spe-
cific immune responses. Zajac and colleagues also showed that
the silent phenotype of virus-specific CTL was especially prev-
alent when there was inadequate CD4 help. Infection with SIV
or HIV does impair CD4 help (5, 12, 21, 24), and it may
therefore not only result in a loss of virus-specific CD8 T-cell
response over time (8, 15) but also result in a functional im-
pairment of the virus-specific CTL response. Although the
absolute numbers of CD4-positive cells per microliter of blood
in our SIV-infected monkeys were continuously decreasing
over time (data not shown), there was no obvious correlation
between the drop in IFN-production in tetramer-positive
cells with CD4 counts. A recent study by Gea-Banacloche and
colleagues described an inability of 30 to 50% of tetramer-
FIG. 1. Functional nature of tetramer-positive, CD8
T cells. PBMCs were stimulated with the CM9 peptide for 6.5 h, with the last5hinthe
presence of BFA. The percentage of CD8
cells producing IFN-(IFNg), as detected in the cytokine assay, was divided by the percentage of
CD8-positive cells staining with the Mamu-A*01/CM9-tetramer before stimulation, to obtain the ratios plotted. Only values of 0.1% (above
background) of CD8
cells were considered positive for both the tetramer staining and the intracellular cytokine assay (see Fig. 2 legend). Ratios
below 1 indicate that some tetramer-positive cells are unable to produce IFN-in response to specific peptide stimulation. Weeks 3, 6, 24, 28,
40, 44, and 48 were tested using thawed PBMCs, and samples from week 6, 28, 44, and 48 were all tested in one assay.
VOL. 75, 2001 NOTES 2459
positive cells to produce IFN-in response to stimulation with
recombinant vaccinia virus-infected B-LCL, which expressed
HIV genes, in two HIV-infected patients (14), which is very
similar to the defect of SIV-specific CD8
T cells described
here. In addition, Goepfert and colleagues have described a
10-fold difference between the number of peptide-specific cells
as measured by tetramer staining or ELISPOT assay in HIV-
infected patients (16). Therefore, as we have demonstrated
here, SIV infection seems to cause a functional impairment of
the specific immune response that is very similar to that which
has been described for HIV infection (6, 14, 16, 28). The
SIV/rhesus monkey model is well suited for investigating the
mechanism behind the impairment of the virus-specific im-
mune response. Finally, our finding that not all antigen-specific
CD8
lymphocytes effectively produce IFN-during the
chronic phase of infection has implications for the measure-
ment of antigen-specific CD8
lymphocytes during chronic
SIV infection. Intracellular staining for IFN-might result in
the underestimation of antigen-specific CD8
lymphocytes in
the chronic phase of SIV infection if this is the sole assay
employed. Multiple assays, therefore, need to be employed
when assessing antiviral immune responses in chronically in-
fected macaques and humans.
This work was supported by grants AI42512, AI41913, and
RR00167. D.I.W. is a recipient of an Elizabeth Glaser Scientist award.
REFERENCES
1. Allen, T. M., D. H. O’Connor, J. Peicheng, J. L. Dzuris, B. R. Mothe´, T. U.
Vogel, E. Dunphy, M. E. Liebl, C. Emerson, N. Wilson, K. J. Kunstman, X.
Wang, D. B. Allison, A. L. Hughes, R. C. Desrosiers, J. D. Altman, S. M.
Wolinsky, A. Sette, and D. I. Watkins. 2000. Tat-specific cytotoxic T lym-
phocytes select for SIV escape variants during resolution of primary viremia.
Nature 407:386–390.
FIG. 2. IFN-production in response to mitogen or peptide-specific stimulation at week 36 postchallenge. Tetramer-positive cells are still able
to respond to mitogen stimulation, although they demonstrate reduced ability to respond to specific peptide stimulation at week 36 postchallenge.
PBMCs were either stimulated with autologous B-LCL in the presence of a control peptide (B-LCL), with PMA/ionomycin (PMA/Iono.), or with
autologous B-LCL in the presence of CM9 peptide (CM9BLCL). Between 100,000 and 150,000 events were acquired, gated on small
lymphocytes. Numbers within graphs indicate the percentage of CD8
cells in each quadrant. The background for the tetramer staining, using
samples from Mamu-A*01-positive, uninfected or Mamu-A*01-negative, infected animals, was below 0.08%. The gate for IFN-staining was set
using an isotype-stained control, so that less than 0.1% of CD8
cells fell into the positive gate.
2460 NOTES J. VIROL.
2. Allen, T. M., J. Sidney, M. F. del Guercio, R. L. Glickman, G. L. Lensmeyer,
D. A. Wiebe, R. DeMars, C. D. Pauza, R. P. Johnson, A. Sette, and D. I.
Watkins. 1998. Characterization of the peptide binding motif of a rhesus
MHC class I molecule (Mamu-A*01) that binds an immunodominant CTL
epitope from simian immunodeficiency virus. J. Immunol. 160:6062–6071.
3. Allen, T. M., T. U. Vogel, D. H. Fuller, B. R. Mothe, S. Steffen, J. E. Boyson,
T. Shipley, J. Fuller, T. Hanke, A. Sette, J. D. Altman, B. Moss, A. J.
McMichael, and D. I. Watkins. 2000. Induction of AIDS virus-specific CTL
activity in fresh, unstimulated peripheral blood lymphocytes from rhesus
macaques vaccinated with a DNA prime/modified vaccinia virus Ankara
boost regimen. J. Immunol. 164:4968–4978.
4. Altman, J. D., P. A. H. Moss, P. J. R. Goulder, D. H. Barouch, M. G.
McHeyzer-Williams, J. I. Bell, A. J. McMichael, and M. M. Davis. 1996.
Phenotypic analysis of antigen-specific T lymphocytes. Science 274:94–96.
5. Ameisen, J. C., and A. Capron. 1991. Cell dysfunction and depletion in
AIDS: the programmed cell death hypothesis. Immunol. Today 12:102–105.
6. Appay, V., D. F. Nixon, S. M. Donahoe, G. M. Gillespie, T. Dong, A. King,
G. S. Ogg, H. M. Spiegel, C. Conlon, C. A. Spina, D. V. Havlir, D. D.
Richman, A. Waters, P. Easterbrook, A. J. McMichael, and S. L. Rowland-
Jones. 2000. HIV-specific CD8() T cells produce antiviral cytokines but are
impaired in cytolytic function. J. Exp. Med. 192:63–76.
7. Borrow, P., H. Lewicki, X. Wei, M. S. Horwitz, N. Peffer, H. Meyers, J. A.
Nelson, J. E. Gairin, B. H. Hahn, M. B. Oldstone, and G. M. Shaw. 1997.
Antiviral pressure exerted by HIV-1-specific cytotoxic T lymphocytes (CTLs)
during primary infection demonstrated by rapid selection of CTL escape
virus. Nat. Med. 3:205–211.
8. Carmichael, A., X. Jin, P. Sissons, and L. Borysiewicz. 1993. Quantitative
analysis of the human immunodeficiency virus type 1 (HIV-1)-specific cyto-
toxic T lymphocyte (CTL) response at different stages of HIV-1 infection:
differential CTL responses to HIV-1 and Epstein-Barr virus in late disease.
J. Exp. Med. 177:249–256.
9. Chen, Z. W., A. Craiu, L. Shen, M. J. Kuroda, U. C. Iroku, D. I. Watkins, G.
Voss, and N. L. Letvin. 2000. Simian immunodeficiency virus evades a dom-
inant epitope-specific cytotoxic T lymphocyte response through a mutation
resulting in the accelerated dissociation of viral peptide and MHC class I. J.
Immunol. 164:6474–6479.
10. Collins, K. L., B. K. Chen, S. A. Kalams, B. D. Walker, and D. Baltimore.
1998. HIV-1 Nef protein protects infected primary cells against killing by
cytotoxic T lymphocytes. Nature 391:397–401.
11. Donahoe, S. M., W. J. Moretto, R. V. Samuel, P. A. Marx, T. Hanke, R. I.
Connor, and D. F. Nixon. 2000. Direct measurement of CD8T cell re-
sponses in macaques infected with simian immunodeficiency virus. Virology
272:347–356.
12. Epstein, J. S., W. R. Frederick, A. H. Rook, L. Jackson, J. F. Manischewitz,
R. E. Mayner, H. Masur, J. C. Enterline, J. Y. Djeu, and G. V. Quinnan, Jr.
1985. Selective defects in cytomegalovirus- and mitogen-induced lymphocyte
proliferation and interferon release in patients with acquired immunodefi-
ciency syndrome. J. Infect. Dis. 152:727–733.
13. Evans, D. T., D. H. O’Connor, P. Jing, J. L. Dzuris, J. Sidney, J. da Silva,
T. M. Allen, H. Horton, J. E. Venham, R. A. Rudersdorf, T. Vogel, C. D.
Pauza, R. E. Bontrop, R. DeMars, A. Sette, A. L. Hughes, and D. I. Watkins.
1999. Virus-specific cytotoxic T-lymphocyte responses select for amino-acid
variation in simian immunodeficiency virus Env and Nef. Nat. Med. 5:1270–
1276.
14. Gea-Banacloche, J. C., S. A. Migueles, L. Martino, W. L. Shupert, A. C.
McNeil, M. S. Sabbaghian, L. Ehler, C. Prussin, R. Stevens, L. Lambert, J.
Altman, C. W. Hallahan, J. C. de Quiros, and M. Connors. 2000. Mainte-
nance of large numbers of virus-specific CD8T cells in HIV-infected
progressors and long-term nonprogressors. J. Immunol. 165:1082–1092.
15. Geretti, A. M., M. E. Dings, C. A. van Els, C. A. van Baalen, F. J. Wijnholds,
J. C. Borleffs, and A. D. Osterhaus. 1996. Human immunodeficiency virus
type 1 (HIV-1) and Epstein-Barr virus-specific cytotoxic T lymphocyte pre-
cursors exhibit different kinetics in HIV-1-infected persons. J. Infect. Dis.
174:34–45.
16. Goepfert, P. A., A. Bansal, B. H. Edwards, G. D. Ritter, Jr., I. Tellez, S. A.
McPherson, S. Sabbaj, and M. J. Mulligan. 2000. A significant number of
human immunodeficiency virus epitope-specific cytotoxic T lymphocytes de-
tected by tetramer binding do not produce gamma interferon. J. Virol.
74:10249–10255.
17. Goulder, P. J., R. E. Phillips, R. A. Colbert, S. McAdam, G. Ogg, M. A.
Nowak, P. Giangrande, G. Luzzi, B. Morgan, A. Edwards, A. J. McMichael,
and S. Rowland-Jones. 1997. Late escape from an immunodominant cyto-
toxic T-lymphocyte response associated with progression to AIDS. Nat. Med.
3:212–217.
18. Jung, T., U. Schauer, C. Heusser, C. Neumann, and C. Rieger. 1993. Detec-
tion of intracellular cytokines by flow cytometry. J. Immunol. Methods 159:
197–207.
19. Kuroda, M. J., J. E. Schmitz, D. H. Barouch, A. Craiu, T. M. Allen, A. Sette,
D. I. Watkins, M. A. Forman, and N. L. Letvin. 1998. Analysis of Gag-specific
cytotoxic T lymphocytes in simian immunodeficiency virus-infected rhesus
monkeys by cell staining with a tetrameric major histocompatibility complex
class I-peptide complex. J. Exp. Med. 187:1373–1381.
20. Kuroda, M. J., J. E. Schmitz, W. A. Charini, C. E. Nickerson, C. I. Lord,
M. A. Forman, and N. L. Letvin. 1999. Comparative analysis of cytotoxic T
lymphocytes in lymph nodes and peripheral blood of simian immunodefi-
ciency virus-infected rhesus monkeys. J. Virol. 73:1573–1579.
21. Lane, H. C., J. M. Depper, W. C. Greene, G. Whalen, T. A. Waldmann, and
A. S. Fauci. 1985. Qualitative analysis of immune function in patients with
the acquired immunodeficiency syndrome. Evidence for a selective defect in
soluble antigen recognition. N. Engl. J. Med. 313:79–84.
22. Lee, P. P., C. Yee, P. A. Savage, L. Fong, D. Brockstedt, J. S. Weber, D.
Johnson, S. Swetter, J. Thompson, P. D. Greenberg, M. Roederer, and M. M.
Davis. 1999. Characterization of circulating T cells specific for tumor-asso-
ciated antigens in melanoma patients. Nat. Med. 5:677–685.
23. Moss, P. A., S. L. Rowland-Jones, P. M. Frodsham, S. McAdam, P. Gian-
grande, A. J. McMichael, and J. I. Bell. 1995. Persistent high frequency of
human immunodeficiency virus-specific cytotoxic T cells in peripheral blood
of infected donors. Proc. Natl. Acad. Sci. USA 92:5773–5777.
24. Murray, H. W., B. Y. Rubin, H. Masur, and R. B. Roberts. 1984. Impaired
production of lymphokines and immune (gamma) interferon in the acquired
immunodeficiency syndrome. N. Engl. J. Med. 310:883–889.
25. Picker, L. J., M. K. Singh, Z. Zdraveski, J. R. Treer, S. L. Waldrop, P. R.
Bergstresser, and V. C. Maino. 1995. Direct demonstration of cytokine
synthesis heterogeneity among human memory/effector T cells by flow cy-
tometry. Blood 86:1408–1419.
26. Ploegh, H. L. 1998. Viral strategies of immune evasion. Science 280:248–253.
27. Price, D. A., P. J. Goulder, P. Klenerman, A. K. Sewell, P. J. Easterbrook, M.
Troop, C. R. Bangham, and R. E. Phillips. 1997. Positive selection of HIV-1
cytotoxic T lymphocyte escape variants during primary infection. Proc. Natl.
Acad. Sci. USA 94:1890–1895.
28. Trimble, L. A., and J. Lieberman. 1998. Circulating CD8 T lymphocytes in
human immunodeficiency virus-infected individuals have impaired function
and downmodulate CD3 zeta, the signaling chain of the T-cell receptor
complex. Blood 91:585–594.
29. Valitutti, S., S. Muller, M. Dessing, and A. Lanzavecchia. 1996. Different
responses are elicited in cytotoxic T lymphocytes by different levels of T cell
receptor occupancy. J. Exp. Med. 183:1917–1921.
30. Wilson, J. D., G. S. Ogg, R. L. Allen, P. J. Goulder, A. Kelleher, A. K. Sewell,
C. A. O’Callaghan, S. L. Rowland-Jones, M. F. Callan, and A. J. McMichael.
1998. Oligoclonal expansions of CD8() T cells in chronic HIV infection are
antigen specific. J. Exp. Med. 188:785–790.
31. Zajac, A. J., J. N. Blattman, K. Murali-Krishna, D. J. Sourdive, M. Suresh,
J. D. Altman, and R. Ahmed. 1998. Viral immune evasion due to persistence
of activated T cells without effector function. J. Exp. Med. 188:2205–2213.
VOL. 75, 2001 NOTES 2461
... CD8 T cell exhaustion | ATAC-seq | epigenetic profiles I n contrast to the highly functional memory CD8 T cells that are generated following resolution of an acute viral infection, continuous antigenic stimulation results in various stages of T cell dysfunction (1). This functional exhaustion of CD8 T cells has been documented during chronic viral infections as well as cancer (2)(3)(4)(5)(6)(7)(8). A characteristic feature of exhausted CD8 T cells is expression of various inhibitory receptors, most notably PD-1 (programmed cell death 1) (9,10). ...
Article
Full-text available
We have recently defined a novel population of PD-1 (programmed cell death 1)+ TCF1 (T cell factor 1)+ virus-specific CD8 T cells that function as resource cells during chronic LCMV infection and provide the proliferative burst seen after PD-1 blockade. Such CD8 T cells have been found in other chronic infections and also in cancer in mice and humans. These CD8 T cells exhibit stem-like properties undergoing self-renewal and also differentiating into the terminally exhausted CD8 T cells. Here we compared the epigenetic signature of stem-like CD8 T cells with exhausted CD8 T cells. ATAC-seq analysis showed that stem-like CD8 T cells had a unique signature implicating activity of HMG (TCF) and RHD (NF-κB) transcription factor family members in contrast to higher accessibility to ETS and RUNX motifs in exhausted CD8 T cells. In addition, regulatory regions of the transcription factors Tcf7 and Id3 were more accessible in stem-like cells whereas Prdm1 and Id2 were more accessible in exhausted CD8 T cells. We also compared the epigenetic signatures of the 2 CD8 T cell subsets from chronically infected mice with effector and memory CD8 T cells generated after an acute LCMV infection. Both CD8 T cell subsets generated during chronic infection were strikingly different from CD8 T cell subsets from acute infection. Interestingly, the stem-like CD8 T cell subset from chronic infection, despite sharing key functional properties with memory CD8 T cells, had a very distinct epigenetic program. These results show that the chronic stem-like CD8 T cell program represents a specific adaptation of the T cell response to persistent antigenic stimulation.
... Although virus-specific CD8 + T cells are critical for controlling HIV and SIV infections, they fail to fully suppress viral replication (20). Several mechanisms are thought to contribute to this failure including: the emergence of CTL escape variants (21)(22)(23)(24)(25)(26)(27)(28), viral induced MHC class I down-modulation (29,30), viral latency (31), CTL exhaustion (32)(33)(34), and potential Treg inhibition of CTL (35)(36)(37)(38)(39). A particularly compelling factor, which we address in this study, is that levels of virus-specific CD8 + T cells are low within B cell follicles, thereby permitting ongoing viral replication (8,9,(40)(41)(42). ...
Article
Full-text available
There is a need to develop improved methods to treat and potentially cure HIV infection. During chronic HIV infection, replication is concentrated within T follicular helper cells (Tfh) located within B cell follicles, where low levels of virus-specific CTL permit ongoing viral replication. We previously showed that elevated levels of simian immunodeficiency virus (SIV)-specific CTL in B cell follicles are linked to both decreased levels of viral replication in follicles and decreased plasma viral loads. These findings provide the rationale to develop a strategy for targeting follicular viral-producing (Tfh) cells using antiviral chimeric antigen receptor (CAR) T cells co-expressing the follicular homing chemokine receptor CXCR5. We hypothesize that antiviral CAR/CXCR5-expressing T cells, when infused into an SIV-infected animal or an HIV-infected individual, will home to B cell follicles, suppress viral replication, and lead to long-term durable remission of SIV and HIV. To begin to test this hypothesis, we engineered gammaretroviral transduction vectors for co-expression of a bispecific anti-SIV CAR and rhesus macaque CXCR5. Viral suppression by CAR/CXCR5-transduced T cells was measured in vitro, and CXCR5-mediated migration was evaluated using both an in vitro transwell migration assay, as well as a novel ex vivo tissue migration assay. The functionality of the CAR/CXCR5 T cells was demonstrated through their potent suppression of SIVmac239 and SIVE660 replication in in vitro and migration to the ligand CXCL13 in vitro, and concentration in B cell follicles in tissues ex vivo. These novel antiviral immunotherapy products have the potential to provide long-term durable remission (functional cure) of HIV and SIV infections.
... A hallmark of the exhausted phenotype of CD8 and CD4 T cells in chronic infection is the progressive loss of cellular functions, such as the ability to produce different cytokines as well as the ability to proliferate in response to antigen. Indeed, in chronic simian immunodeficiency virus (SIV) infection, as CD8 T cell exhaustion progresses, SIV-specific CD8 T cells have been shown first to lose the ability to produce IL-2 in response to antigen, then TNF-␣, and finally IFN-␥ (71)(72)(73)(74). Furthermore, the ability to produce several cytokines upon stimulation with antigen has been observed to be sensitive to the amount of PD-1 present on a given cell, with previous studies showing inhibition of IL-2 production requiring little expression of PD-1 while MIP-1␤ production is only inhibited in the presence of high levels of PD-1 expression (43). ...
Article
Full-text available
Inhibitory receptors have been extensively described for their importance in regulating immune responses in chronic infections and cancers. Blocking the function of inhibitory receptors such as PD-1, CTLA-4, 2B4, Tim-3, and LAG-3 has shown promise for augmenting CD8 T cell activity and boosting pathogen-specific immunity. However, the prevalence of inhibitory receptors on CD4 T cells and their relative influence on CD4 T cell functionality in chronic HIV infection remains poorly described. We therefore determined and compared inhibitory receptor expression patterns of 2B4, CTLA-4, LAG-3, PD-1, and Tim-3 on virus-specific CD4 and CD8 T cells in relation to their functional T cell profile. In chronic HIV infection, inhibitory receptor distribution differed markedly between cytokine-producing T cell subsets with, gamma interferon (IFN-γ)- and tumor necrosis factor alpha (TNF-α)-producing cells displaying the highest and lowest prevalence of inhibitory receptors, respectively. Blockade of inhibitory receptors differentially affected cytokine production by cells in response to staphylococcal enterotoxin B stimulation. CTLA-4 blockade increased IFN-γ and CD40L production, while PD-1 blockade strongly augmented IFN-γ, interleukin-2 (IL-2), and TNF-α production. In a Friend retrovirus infection model, CTLA-4 blockade in particular was able to improve control of viral replication. Together, these results show that inhibitory receptor distribution on HIV-specific CD4 T cells varies markedly with respect to the functional subset of CD4 T cells being analyzed. Furthermore, the differential effects of receptor blockade suggest novel methods of immune response modulation, which could be important in the context of HIV vaccination or therapeutic strategies. IMPORTANCE Inhibitory receptors are important for limiting damage by the immune system during acute infections. In chronic infections, however, their expression limits immune system responsiveness. Studies have shown that blocking inhibitory receptors augments CD8 T cell functionality in HIV infection, but their influence on CD4 T cells remains unclear. We assessed the expression of inhibitory receptors on HIV-specific CD4 T cells and their relationship with T cell functionality. We uncovered differences in inhibitory receptor expression depending on the CD4 T cell function. We also found differences in functionality of CD4 T cells following blocking of different inhibitory receptors, and we confirmed our results in a Friend virus retroviral model of infection in mice. Our results show that inhibitory receptor expression on CD4 T cells is linked to CD4 T cell functionality and could be sculpted by blockade of specific inhibitory receptors. These data reveal exciting possibilities for the development of novel treatments and immunotherapeutics.
... HIV-specific T-helper dysfunction has been observed early in the infection when CD4 + T-cell counts are still within the normal range, thereby affecting both cellular and humoral immunity (Shearer 1991;Miedema 1994;Musey 1999, Altfeld 2000Wilson 2000). CD8 + T-cell dysfunction involves impaired cytolytic activity (Appay 2000; Shankar 2000) and reduction in cytokine production (Goepfert 2000;Vogel 2001). ...
Article
Chronic HIV infection profoundly perturbs CD8 T cell responses, although the mechanisms for this are not completely elucidated. We hypothesised that persistent antigen burden affects the proliferation potential and differentiation state of the general CD8 T cell pool. Using CFSE staining and multicolour flow cytometry, we compared the proportions and proliferation responses of naïve (CD45RA+CD27+), memory (CD45RA-CD27+,M), effector-memory (CD45RA-CD27-, EM), and terminally-effector (CD45RA+CD27-,TE) CD8 T subsets in HIV+ patients and HIV- controls. Two patterns of CD8 T cell proliferation and differentiation were observed in HIV+ patients: subgroup A (N=13) with expanding M (81% vs. 16% on D0), and steady EM (12 vs. 9% on D0) CD8 subsets, comparable to HIV- controls, and subgroup B (N=11): with predominant differentiation and accumulation of EM CD8 T cells (48% vs. 22%). No significant differences between subgroups A and B regarding CD4 AC or HIV-specific response were established. However, group A presented with a significantly lower VL (4.6 vs. 2.0 log HIV RNA copies/ml), and a higher level of circulating Treg cells (8.4% vs. 4.9%, p<0.05), combined with a significantly higher proportion of naïve CD8 T cells and a lower proportion of TE. Thus, HIV-induced Treg might determine the differentiation state and functional fitness of the CD8 T cell pool in the settings of long-lasting chronic infection and should be considered during immune monitoring.
Chapter
Animal retroviruses are classified within seven different genera, of which members of five genera are present in primates C-type virus, D-type virus, human T-cell leukemia virus (HTLV), simian T-cell lymphotropic virus (STLV), bovine leukemia virus (BLV), lentiviruses, and spuma or foamy viruses. This chapter reviews the salient biology of each of these five viral groups. Emphasis is placed on the D-type simian retroviruses (SRVs) and lentiviruses (SIVs) because of their etiologic association with simian AIDS (SAIDS). Humans also harbor retroviruses: two strains of human T-lymphotropic virus (HTLV-1 and HTLV-2) with simian counterparts (STLV-1 and STLV-2) and two major strains of lentivirus, human immunodeficiency virus (HIV-1 and HIV-2), which also have simian counterparts (SIVs). In the natural African simian hosts, SIV does not cause any disease, but experimental infection of captive macaques with certain SIV strains, especially from the sooty mangabey, produces a progressive and fatal immunodeficiency syndrome similar to AIDS in humans, making this primate lentivirus model very useful for research into AIDS pathogenesis, vaccines, and antiviral therapy.
Article
Purpose of review: Despite eliciting an early antiviral T cell response, HIV-specific T cells are unable to prevent disease progression, partly because of their loss of effector functions, known as T cell exhaustion. Restoring this T cell functionality represents a critical step for regaining immunological control of HIV-1 replication, and may be fundamental for the development of a functional cure for HIV. In this context, the use of animal models is invaluable for evaluating the efficacy and mechanisms of novel therapeutics aimed at reinvigorating T cell functions. Recent findings: Although nonhuman primates continue to be a mainstay for studying HIV pathogenesis and therapies, recent advances in humanized mouse models have improved their ability to recapitulate the features of cell exhaustion during HIV infection. Targeting coinhibitory receptors in HIV-infected and simian immunodeficiency virus (SIV)-infected animals has resulted in viral load reductions, presumably by reinvigorating the effector functions of T cells. Additionally, studies combining programmed death-1 (PD-1) blockade with suppressive antiretroviral therapy provide further support to the use of coinhibitory receptor blockades in restoring T cell function by delaying viral load rebound upon antiretroviral therapy interruption. Future in-vivo studies should build on recent in-vitro data, supporting the simultaneous targeting of multiple regulators of cell exhaustion. Summary: In this review, we describe the most recent advances in the use of animal models for the study of cell exhaustion following HIV/SIV infection. These findings suggest that the use of animal models is increasingly critical in translating immunotherapeutics into clinical practice.
Article
Infection of rhesus macaques with simian immunodeficiency virus (SIV) is an excellent model system for studying viral adaptation to immune responses. In this review, we discuss how the SIV-infected macaque has provided unequivocal evidence for cytotoxic T-lymphocyte (CTL) selection of viral escape variants. This improved understanding of CTL escape may influence human immunodeficiency virus (HIV) vaccine design as well as our understanding of HIV pathogenesis.
Article
Full-text available
The virus-specific CD8+ T cell responses of 21 HIV-infected patients were studied including a unique cohort of long-term nonprogressors with low levels of plasma viral RNA and strong proliferative responses to HIV Ags. HIV-specific CD8+ T cell responses were studied by a combination of standard cytotoxic T cell (CTL) assays, MHC tetramers, and TCR repertoire analysis. The frequencies of CD8+ T cells specific to the majority of HIV gene products were measured by flow cytometric detection of intracellular IFN-γ in response to HIV-vaccinia recombinant-infected autologous B cells. Very high frequencies (0.8–18.0%) of circulating CD8+ T cells were found to be HIV specific. High frequencies of HIV-specific CD8+ T cells were not limited to long-tern nonprogressors with restriction of plasma virus. No correlation was found between the frequency of HIV-specific CD8+ T cells and levels of plasma viremia. In each case, the vast majority of cells (up to 17.2%) responded to gag-pol. Repertoire analysis showed these large numbers of Ag-specific cells were scattered throughout the repertoire and in the majority of cases not contained within large monoclonal expansions. These data demonstrate that high numbers of HIV-specific CD8+ T cells exist even in patients with high-level viremia and progressive disease. Further, they suggest that other qualitative parameters of the CD8+ T cell response may differentiate some patients with very low levels of plasma virus and nonprogressive disease.
Article
Full-text available
The vertebrate body is an ideal breeding ground for viruses and provides the conditions that promote their growth, survival, and transmission. The immune system evolved and deals with this challenge. Mutually assured destruction is not a viable evolutionary strategy; thus, the study of host-virus interactions provides not only a glimpse of life at immunity's edge, but it has also illuminated essential functions of the immune system, in particular, the area of major histocompatibility complex–restricted antigen presentation.
Article
Although human immunodeficiency virus (HIV)-infected subjects without acquired immunodeficiency syndrome have a high frequency of HIV-specific CD8 T lymphocytes, freshly isolated lymphocytes frequently lack detectable HIV-specific cytotoxicity. However, this effector function becomes readily apparent after overnight culture. To investigate reasons for T-cell dysfunction, we analyzed T-cell expression of the cytolytic protease granzyme A and of CD3ζ, the signaling component of the T-cell receptor complex. An increased proportion of CD4 and CD8 T cells from HIV-infected donors contain granzyme A, consistent with the known increased frequency of activated T cells. In 28 HIV-infected donors with mild to advanced immunodeficiency, a substantial fraction of circulating T cells downmodulated CD3ζ (fraction of T cells expressing CD3ζ, 0.74 ± 0.16 v 1.01 ± 0.07 in healthy donors; P < .0000005). CD3ζ expression is downregulated more severely in CD8 than CD4 T cells, decreases early in infection, and correlates with declining CD4 counts and disease stage. CD3ζ expression increases over 6 to 16 hours of culture in an interleukin-2–dependent manner, coincident with restoration of viral-specific cytotoxicity. Impaired T-cell receptor signaling may help explain why HIV-specific cytotoxic T lymphocytes fail to control HIV replication.
Article
The simian immunodeficiency virus (SIV) macaque model system has been used extensively to study AIDS pathogenesis and to test candidate vaccines for their ability to protect against homologous or heterologous challenge with pathogenic SIV or SHIV. Recent studies suggest that stimulation of HIV-1-specific CTL responses is important for effective vaccination against HIV-1. While quantitative measurements of SIV-specific cytotoxic T lymphocyte (CTL) responses have been facilitated by the use of tetrameric peptide complexes, this technique is currently limited to the study of Mamu-A*01-positive rhesus macaques. Furthermore, very few SIV-specific CTL epitopes have been identified, and there is limited identification of other MHC alleles in macaques. In this study, cytokine flow cytometry (CFC) was used to quantify SIV-specific CD8+ antigen-reactive T cells in macaques infected with SIV. We found a strong correlation (r = 0.96, P < 0.001) between CD8+ antigen-reactive T cells stained with the Mamu-A*01 p11C, C-M tetramer and production of intracellular TNF-α in the CFC assay. Furthermore, the CFC assay was used to identify a novel SIV-specific CTL epitope in Envelope (SIV Env, a.a. 486–494, sequence AEVAELYRL). The use of the CFC assay facilitates the study of antigen-reactive T cell responses in SIV infection and vaccination.
Article
Normal immature thymocytes respond to activation by undergoing programmed cell death (apoptosis), a physiological deletion mechanism involved in the selection of the T-cell repertoire. In this article, Jean Claude Ameisen and André Capron suggest that inappropriate induction of a form of programmed T-cell death could account for both qualitative and quantitative helper T-cell defects of HIV-infected patients. A model of AIDS pathogenesis is presented that may explain several features of HIV infection, including evolution of the disease and the development of defects in nonimmunological organs.
Article
To examine the defect in cellular immunity in patients with acquired immunodeficiency syndrome (AIDS), we studied in vitro lymphocyte proliferation and interferon (IFN) release in response to cytomegalovirus (CMV) antigen and Concanavalin A mitogen in 40 homosexual men with AIDS, 10 homosexual men with chronic lymphadenopathy syndrome, 7 healthy homosexual men, and 18 healthy heterosexual subjects of either sex. CMV serology by an enzyme-linked immunosorbent assay and viral cultures for CMV were performed. Lymphocytes of patients with AIDS showed impaired CMV-specific release of IFN but normal mitogen-induced IFN release. The defect was not attributable to CMV-infection per see Cell proliferation in response to both CMV antigen and mitogen was impaired in patients with AIDS who had opportunistic infections. The defect could not be attributed to CMV viremia. We concluded that impaired releaseof IFN in response to a viral antigen is characteristic of lymphocytes in patients with AIDS and that this defect is distinct from a defect in mitogenic responsiveness, which coexists predominantly in patients with opportunistic infections.
Article
We studied purified subpopulations of lymphocytes from patients with the acquired immunodeficiency syndrome (AIDS) in order to determine whether intrinsic defects in lymphocyte function, aside from those due to alterations in lymphocyte numbers, were present. Mitogen-stimulated DNA synthesis, production of gamma interferon, production of interleukin-2, and expression of interleukin-2 receptors, although variably decreased in unseparated cell populations, were normal in populations of purified T-cell subsets. In contrast, DNA synthesis in response to the soluble protein antigen tetanus toxoid was decreased in both unseparated and purified T-cell subpopulations. Cell-mixing experiments demonstrated that the hyporesponsiveness of the unfractionated lymphocytes from patients with AIDS was not due to active suppression. We conclude that the lymphocytes of patients with AIDS, although capable of undergoing a normal degree of blast transformation and lymphokine production after mitogenic stimulation, have an intrinsic defect in their ability to recognize and respond to soluble antigen.
Article
To examine the cellular immune defect that predisposes patients with the acquired immunodeficiency syndrome (AIDS) to opportunistic infections, we tested T lymphocytes from 16 patients for the capacity to secrete macrophage-activating products (lymphokines) including gamma interferon. Mononuclear cells from 10 of 11 patients did not generate an effective lymphokine in response to mitogen, and 11 of 16 produced subnormal levels of gamma interferon (less than 300 U per milliliter). In addition, upon stimulation with specific microbial antigen, cells from none of 14 patients generated active lymphokines, and cells from 13 to 14 completely failed to secrete gamma interferon. However, the antimicrobial function of monocytes from the patients was intact, and once stimulated with normal lymphokines or gamma interferon alone, macrophages derived from patients' monocytes responded with enhanced and effective intracellular antimicrobial activity. These results suggest that impaired lymphokine production may predispose patients with AIDS to opportunistic infections, and they provide a rationale for using gamma interferon as immunotherapy.