ArticlePDF Available

Sustained Dysfunction of Antiviral CD8+ T Lymphocytes after Infection with Hepatitis C Virus

American Society for Microbiology
Journal of Virology
Authors:

Abstract and Figures

Hepatitis C virus (HCV) sets up persistent infection in the majority of those exposed. It is likely that, as with other persistent viral infections, the efficacy of T-lymphocyte responses influences long-term outcome. However, little is known about the functional capacity of HCV-specific T-lymphocyte responses induced after acute infection. We investigated this by using major histocompatibility complex class I-peptide tetrameric complexes (tetramers), which allow direct detection of specific CD8+T lymphocytes ex vivo, independently of function. Here we show that, early after infection, virus-specific CD8+ T lymphocytes detected with a panel of four such tetramers are abnormal in terms of their synthesis of antiviral cytokines and lytic activity. Furthermore, this phenotype is commonly maintained long term, since large sustained populations of HCV-specific CD8+ T lymphocytes were identified, which consistently had very poor antiviral cytokine responses as measured in vitro. Overall, HCV-specific CD8+T lymphocytes show reduced synthesis of tumor necrosis factor alpha (TNF-α) and gamma interferon (IFN-γ) after stimulation with either mitogens or peptides, compared to responses to Epstein-Barr virus and/or cytomegalovirus. This behavior of antiviral CD8+ T lymphocytes induced after HCV infection may contribute to viral persistence through failure to effectively suppress viral replication.
Content may be subject to copyright.
JOURNAL OF VIROLOGY,
0022-538X/01/$04.000 DOI: 10.1128/JVI.75.12.5550–5558.2001
June 2001, p. 5550–5558 Vol. 75, No. 12
Copyright © 2001, American Society for Microbiology. All Rights Reserved.
Sustained Dysfunction of Antiviral CD8
T Lymphocytes after
Infection with Hepatitis C Virus
NORBERT H. GRUENER,
1
FRANZISKA LECHNER,
2
MARIA-CHRISTINA JUNG,
3
HELMUT DIEPOLDER,
3
TILMAN GERLACH,
3
GEORG LAUER,
4
BRUCE WALKER,
4
JOHN SULLIVAN,
5
RODNEY PHILLIPS,
2
GERD R. PAPE,
1,3
AND PAUL KLENERMAN
2
*
Institute for Immunology, D-80336 Munich,
1
and Medical Department II, Klinikum Grosshadern, D-81366 Munich,
3
Germany; Nuffield Department of Clinical Medicine, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
2
;
Infectious Diseases Unit and AIDS Research Center, Massachussets General Hospital and Harvard Medical
School, Boston, Massachusetts 02129
4
; and Australian Red Cross Blood Service, Sydney 2000, Australia
5
Received 23 January 2001/Accepted 12 March 2001
Hepatitis C virus (HCV) sets up persistent infection in the majority of those exposed. It is likely that, as with
other persistent viral infections, the efficacy of T-lymphocyte responses influences long-term outcome. However,
little is known about the functional capacity of HCV-specific T-lymphocyte responses induced after acute in-
fection. We investigated this by using major histocompatibility complex class I-peptide tetrameric complexes
(tetramers), which allow direct detection of specific CD8
T lymphocytes ex vivo, independently of function.
Here we show that, early after infection, virus-specific CD8
T lymphocytes detected with a panel of four such
tetramers are abnormal in terms of their synthesis of antiviral cytokines and lytic activity. Furthermore, this
phenotype is commonly maintained long term, since large sustained populations of HCV-specific CD8
T lymphocytes were identified, which consistently had very poor antiviral cytokine responses as measured in
vitro. Overall, HCV-specific CD8
T lymphocytes show reduced synthesis of tumor necrosis factor alpha
(TNF-) and gamma interferon (IFN-) after stimulation with either mitogens or peptides, compared to
responses to Epstein-Barr virus and/or cytomegalovirus. This behavior of antiviral CD8
T lymphocytes
induced after HCV infection may contribute to viral persistence through failure to effectively suppress viral
replication.
Hepatitis C virus (HCV) infects approximately 170 million
people worldwide and is a significant cause of liver failure. The
virus is able to set up chronic infection in about 80% of those
infected, although a minority are able to clear the virus from
blood after acute exposure. Although dynamic studies of in-
fection are hampered by the lack of acute symptoms in most
patients, there is nevertheless accumulating evidence that mul-
tiple immune responses are involved (8, 11, 19, 21, 24).
A role for CD4
T-helper lymphocytes is supported by func-
tional and genetic linkage studies (11). (7, 25, 28, 30). Re-
cent studies of acute disease have also indicated that antiviral
antibody responses directed against envelope proteins (E2)
influence viral evolution and disease outcome (9). CD8
T
lymphocytes specific for HCV also arise early after infection
(19–21) and, by analogy with hepatitis B virus infection, theo-
retically could suppress viral replication by secretion of anti-
viral cytokines (13). However, these populations are not sus-
tained and generally become difficult to detect in blood,
especially when techniques that rely on detection of gamma
interferon (IFN-) are used (29), although they can be recov-
ered upon stimulation in vitro (4, 17, 27, 32). There is evidence
from murine models that failure of T-cell responses to control
early-stage infection can lead to shifts in immune-selective
forces and viral escape (6, 16). We therefore asked whether a
specific dysfunction of antiviral CD8
T lymphocytes may oc-
cur after infection with HCV and addressed this question by
analyzing T-lymphocyte responses directly ex vivo with pep-
tide-major histocompatibility complex (MHC) class I tetram-
ers (1, 3, 12) combined with assays for lymphocyte function.
MATERIALS AND METHODS
Patients and blood samples. Patients were recruited from clinics in Munich,
Germany; Oxford, United Kingdom; Sydney, Australia; and Boston, Mass. Pe-
ripheral blood mononuclear cells (PBMCs) were prepared and frozen as previ-
ously described(19–21). For assays comparing different time points from the
same individual, samples were thawed and stained simultaneously. Informed
consent for venipuncture was obtained in all cases. For subjects A and B (both
HLA-A*0201-positive female intravenous drug users who presented acutely and
are studied here in the most detail), seroconversion to anti-HCV antibody pos-
itivity was determined at the time of presentation. Liver histology was not
obtained for subjects A and B, because it was not indicated in acute disease.
Their clinical details are shown in Table 1. Of the other subjects, not studied here
during acute infection, subject C also acquired virus via intravenous drug use,
subject D probably acquired the virus via sexual contact, and subject E acquired
the virus via infected blood products (subjects 1 and 3 and 15 in reference 19).
The time of acquisition of virus was determined from clinical history and from
transfusion data. The full details of the cohorts from which these subjects were
selected, including age, tissue type, sex, PCR and treatment status, and, where ap-
propriate, genotype and liver histology, have been published previously (19–21).
Class I-peptide tetramers. Class I-peptide tetramers were prepared and vali-
dated exactly as previously described. The following peptides were obtained from
Research Genetics (Huntsville, Ala.): NS3 peptide 1073–1081 (abbreviated here
as NS3 1073; CINGVWCTV), NS5B peptide 2594–2602 (NS5 2594; ALYDV
VTKL), NS4B 1406–1415 (NS4 1406; KLVALGINAV), and NS4B 1807–1816
(NS4 1807; LLFNILGGWV). The tetramers for the control Epstein-Barr virus
(EBV) and cytomegalovirus (CMV) peptides restricted by HLA-A2 (EBV
BMLF-1 [GLCTLVAML] and CMV pp65 [NLVPMVATV]) were validated by
using peripheral blood samples from EBV- and CMV-seropositive, healthy in-
* Corresponding author. Mailing address: Nuffield Department of
Clinical Medicine, John Radcliffe Hospital, Oxford OX3 9DU, United
Kingdom. Phone: 01865 221335. Fax: 01865 220993. E-mail: klener
@molbiol.ox.ac.uk.
5550
dividuals as described previously (19, 21). In screening the HCV patients, where
appropriate, tetramers for two HCV epitopes restricted by HLA-B8 (HSKKKL
DEL and LIRLKPTL) and two restricted by HLA-B7 (GPRLGVRAT and
DPRRRSRNL) were also used, as previously described (19, 20). However, since
these did not yield large tetramer-positive populations amenable to functional
analysis, they are not detailed further.
Tetramer staining and functional assays. The following conditions were used
for staining. From 0.5 to 1 million PBMCs were coincubated with tetramer for 20
min at 37°C, followed by washing and phenotypic staining or stimulation. The
following monoclonal antibodies (MAbs) were used: anti-CD8-PerCP, anti-
CD27–fluorescein isothiocyanate (FITC) conjugate, anti-CD45RO–allophyco-
cyanin (APC), anti-CD45RA–FITC (all Becton Dickinson Immunocytometry
Systems), anti-CC chemokine receptor 5 (CCR-5)–FITC, anti-CD38–PC, anti-
CD69–APC (all PharMingen), anti-human MHC class II–FITC (HLA-DR,
-DP, and -DQ; Dako), anti-Ki 67–FITC (Coulter Immunotech, Marseille,
France), anti-perforin–FITC (Pharmingen), and FITC-conjugated anti-T-cell re-
ceptor (TCR) (TCR V1, -2, -3, -5.1, -5.2, -8, -12, -13.1, -14, -16, -17, -20, and -22;
unlabeled anti-TCR V5.3, -9, and -23 [all Coulter Immunotech]). Unlabeled
anti-TCR Vantibodies were detected with FITC-conjugated antimouse immu-
noglobulin (Ig) [F(ab)
2
; Biosource, Camarillo, Calif.]. Phorbol myristate acetate
(PMA)-ionomycin stimulation was performed exactly as described in reference
21, but the tetramer staining was done first (20 min), and then a PMA-ionomycin
mixture was added without washing. Determination of peptide stimulation and
intracellular cytokine secretion was performed in the presence of 10 M cognate
peptide or control and 1 g of costimulatory anti-CD28 (Pharmingen, San
Diego, Calif.) and anti-CD49b antibodies (Becton Dickinson, San Jose, Calif.)
per ml (6 h). Peptide stimulation and CD69 staining were performed in the
absence of costimulation (4 h). Flow cytometric analysis was performed with a
Becton Dickinson FACSCalibur fluorescence-activated cell sorter (FACS), and
analysis was performed with CellQuest software.
CD4 proliferative assays. CD4 proliferative assays were performed with re-
combinant HCV proteins and tritium incorporation exactly as previously de-
scribed (11).
RESULTS
Analysis of CD8
T-lymphocyte responses ex vivo after
acute HCV infection. We first tracked T-lymphocyte responses
in a set of four HLA-A*0201-positive patients for whom a clear
date of onset of infection was available (Table 1). Two exam-
ples (subjects A and B) in which samples were obtained very
close to the time of acquisition of virus are illustrated in detail
in Fig. 1a and described in Table 1. We analyzed CD8
T-
lymphocyte responses against four HLA-A2-restricted HCV
peptides by using peptide-MHC class II tetramers, as previ-
ously described (Fig. 1a, lower panel) (19, 21). In subject A, a
significant response was observed against NS3 1073 (CINGVW
CTV [shown in Fig. 1a, middle panel]) and NS5 2594 (ALYD
VVTKL), but not NS4 1406 (KLVALGINAV) or NS4 1807
(LLFNILGGWV) (not shown). In subject B, only a response
to NS3 1073 was observed, but in this instance, we also had the
opportunity to compare this response with established memory
responses to HLA-A2-restricted epitopes from either EBV
(GLCTLVAML) or CMV (NLVPMVATV [0.74 and 0.25% of
CD8
lymphocytes, respectively]).
Dynamics and phenotype and of antiviral CD8
T-lympho-
cyte responses over time. Interestingly, in subject A, a shift in
dominance between the two detectable responses was noted
over time. This occurred during a period in which the subject’s
blood was already negative for viral RNA by PCR, and a very
similar pattern was observed under similar circumstances in a
previously described case (21), as well as in cases later after
infection (19, 21). The shift in numerical dominance was par-
alleled by a striking shift in expression of markers of activation
between the two populations (Fig. 1b). The response, which
peaked second (directed against NS5 2594), showed peak
CD38 and class II expression 2 weeks later than the first wave
of cells directed against NS3 1073. Also, both populations
showed increased expression of chemokine receptor CCR5 at
the time of their maximal activation, which might contribute to
recruitment into the inflamed liver. The level of expression of
the intracellular marker of proliferation Ki-67 was high for the
first wave of cells observed, at a time when their numbers in
blood were actually waning. This is consistent with either ex-
haustion of this response or redistribution into the liver.
Analysis of function of tetramer-positive cells ex vivo. We
next examined the function of these virus-specific CD8
T-
lymphocyte populations ex vivo by using an established assay
for intracellular detection of IFN-synthesis after stimulation
with PMA-ionomycin (21). These are illustrated in Fig. 1c to e
in detail for subject B, for whom an internal comparison was
available. In this individual, the level of IFN-synthesis after
stimulation was low in the HCV-specific tetramer-positive pop-
ulation and was significantly lower at both time points tested
than those in the control CMV- and EBV-specific populations
(Fig. 1c). The release of IFN-in response to peptide stimu-
lation (a more physiological stimulus, since triggering occurs
through the TCR) showed even more profound defects (Fig.
1d) compared to the internal control of the CMV-specific
response. This dysfunction was sustained over a period of 3
months. Consistent with this, a highly sensitive overnight ex
vivo IFN-ELISpot assay (Mabtech, Upsalla, Sweden) per-
formed with the same NS3 peptide was also completely nega-
tive throughout this period (data not shown) (18). Intermedi-
ate time points over this period showed a very similar profile of
unresponsiveness. For example, at week 7 in subject B, at the
peak of the CD4 responses (Fig. 1a, lower panels), the IFN-
ELISpot remained negative for responses against NS3 1073, as
was synthesis of tumor necrosis factor alpha (TNF-) after
PMA stimulation.
Analysis of CD69 upregulation in comparison to control
responses. To confirm that the defect in cytokine synthesis
represented a failure of triggering (rather than a switch in
secretion pattern to other cytokines), upregulation of CD69
after NS3 1073 peptide stimulation in vitro was also measured
at these time points, as previously analyzed (21). Again, weak
responses were observed, most obviously when compared with
the internal CMV control (Fig. 1e and f).
Examination of tetramer-positive responses of different
specificities and in different individuals. The specificities of
TABLE 1. Clinical details of subjects studied during or after acute infection
Subject Sex Age (yr) Time of analysis HLA class I Clinical outcome
A Female 31 During acute hepatitis A2, A26, B38, B50 Relapse
B Female 37 During acute hepatitis A1, A2, B8, B40 Sustained PCR negative in blood
C Male 42 After acute hepatitis A2, A26, B37 Sustained PCR negative in blood
D Female 32 After acute hepatitis A2, A3, B41, B44 Sustained PCR negative in blood
VOL. 75, 2001 CYTOKINE RELEASE BY HCV-SPECIFIC CD8
T LYMPHOCYTES 5551
FIG. 1. Dynamics of hepatitis and acute immune responses in two subjects. (a) Time course of disease and immune responses. (Upper panels)
Time course of alanine aminotransferase (ALT [international units per milliliter]) in serum over time. Subject A was PCR positive () for HCV
RNA at the first time point and subsequently became PCR negative () over time as indicated, with recrudescence at week 15. In subject B, virus
did not recrudesce, even during longer follow-up periods of 1 year. (Middle panels) Dynamics of tetramer-positive responses. Frozen PBMCs were
thawed and tested in parallel with tetramers for four HLA-A2-restricted peptides (NS3 1073, NS3 1406, NS4B 1807, and NS5B 2594). Thawed
PBMCs were stained exactly as previously described (19, 21). Only positive stains are shown. The proportions were calculated after gating on live
CD8
lymphocytes. (Lower panels) Fresh PBMCs were tested in standard proliferation assays by incorporation of [
3
H]thymidine after stimulation
with HCV antigens as previously described (11). (b) Phenotype of acute responses in subject A. Frozen PBMCs were thawed and stained in parallel
with the tetramers for NS3 1073 and NS5 2094, shown to be positive, together with the antibodies for MHC class II CD38, CCR-5, or (after
permeabilization) Ki-67 (see Materials and Methods). The proportions of tetramer-positive cells staining positive at each time point for each
marker are shown. (c) PMA-ionomycin-stimulated cytokine synthesis over time in subject B. Frozen PBMCs were thawed, tetramer stained for
20 min, and then stimulated with PMA-ionomycin as previously described (21). Staining with PerCP-labeled anti-CD8 was followed
by permeabilization as in panel b and intracellular staining with FITC–anti-IFN-(Becton Dickinson). After four-color flow cytometry, the
proportion of tetramer-positive cells staining positive for intracellular IFN-was calculated. (d) Peptide-stimulated synthesis of IFN-in subject B:
comparison of HCV and control response. Frozen PBMCs from subject B at weeks 2 and 14 were thawed, stained with tetramers for HCV NS3 1073
5552 GRUENER ET AL. J. VIROL.
or CMV, stimulated with the appropriate peptide and costimulatory molecules, permeabilized, and stained for CD8 and intracellular IFN-(21). After
flow cytometric analysis, the CD8
population is displayed. The proportion of tetramer-positive cells staining positive for IFN-is shown. Staining
of cells in the absence of peptide revealed stimulation of 2% in both cases. (e) Peptide-stimulated upregulation of CD69 in subject B: comparison
of HCV and control responses. PBMCs from the same time points as in panel d above were stimulated in the same manner with peptide (21) and
stained thereafter with PerCP–anti-CD8 and FITC–anti-CD69. The proportion of tetramer-positive cells expressing CD69 is illustrated. Expression
in ex vivo samples or in the absence of peptide was 2%. (f) Example of CD69 upregulation in tetramer-positive populations by peptide stimulation.
Examples from the first time point of CD69 surface staining in tetramer-positive cells. The tetramer-positive CD8
population was gated upon and
CD69 expression was analyzed after peptide stimulation. No upregulation of CD69 on tetramer-negative cells was observed (data not shown).
VOL. 75, 2001 CYTOKINE RELEASE BY HCV-SPECIFIC CD8
T LYMPHOCYTES 5553
tetramer-positive responses in different subjects were repro-
duced through analyses of other responses in other patients
and other epitopes. Figure 2a illustrates cytokine staining pat-
terns in three separate individuals in whom three distinct epi-
topes (NS3 1073, NS3 1406, and NS5 2594) from HCV were
targeted. In these cases, little or no cytokine synthesis was
measured after maximal stimulation with PMA-ionomycin.
This was clear by measurement of IFN-and TNF-. These
FIG. 2. Functional analysis of HCV-specific responses against three separate epitopes in three separate donors. (a) Cytokine release from
CD8
lymphocytes of different specificities. Frozen samples from three separate HCV antibody-positive individuals were thawed and tested for
synthesis of antiviral cytokines after PMA-ionomycin stimulation exactly as in Fig. 1c. Both stains for TNF-(right panels [marked FL4-H]) and
IFN-(left panels [ifngFITC]) are illustrated. The numbers shown in each plot represent the percentage of tetramer-positive cells that stained
positive for the particular cytokine. PCR-ve, PCR negative. (b) Control unstimulated cells and a reference for gating on the CD8 high population.
Samples obtained from subject C (NS3 1073 specific) are illustrated. No synthesis of IFN-is seen. (c) Comparison of PMA-ionomycin and peptide
stimulation. A CMV-specific response from a control HCV-negative patient is shown. The samples were tested in parallel for TNF-synthesis
according to the peptide stimulation and PMA stimulation protocols, and the CD8 high population is shown. Approximately similar proportions
of CD8
cytokine-positive cells were obtained, as indicated in the right upper quadrant of each FACS plot. (d) Analysis of Vusage of tetramer-
positive cells in subjects A and B. PBMCs were anti-CD8 and tetramer stained as described above and costained with a panel of FITC-conjugated
V-specific MAbs (Immunotech, Marseille, France). Staining for V3 only is shown, after gating on live CD8
lymphocytes. A large population
of V3-positive, tetramer-positive lymphocytes is seen in subject B (30% of tetramer-positive cells), and a smaller population is seen in subject A
(5%). No dominant Vusage was seen in subject A, C, or D. Tetramer-negative populations in these subjects did not reveal a major oligoclonal
expansion when this restricted panel of antibodies was used (V1, -2, -3, -5.1, -5.2, -5.3, -8, -9, -12, -13.1, -14, -16, -17, -20, -22, and -23).
5554 GRUENER ET AL. J. VIROL.
individuals were all PCR negative at the time of study and had
been so for many months. The plots illustrated demonstrate
the extremes of the range of cytokine responses seen among
HCV-specific CD8
lymphocytes. In all cases, the proportion
of cytokine-positive cells among the tetramer-positive popula-
tion was lower than that in the tetramer-negative CD8
pop-
ulation. Exactly the reverse result is seen in the case of CMV-
and EBV-specific tetramer populations (described below; Fig.
3b). Again, no or very low levels of synthesis were also ob-
served after peptide stimulation (data not shown).
A series of control experiments were performed to confirm
that these results were not artifactual due to stimulation of T
cells by tetramers or downregulation effects due to stimulation.
The tetramer-positive controls left unstimulated for the dura-
tion of the assay are illustrated in Fig. 2b, which demonstrates
that staining with the tetramer alone does not lead to synthesis
of cytokines with this protocol. A typical control stain directly
comparing PMA and peptide-stimulated responses is also
shown (Fig. 2c), which demonstrates that the populations of
CD8
cells that make cytokine after peptide stimulation are
comparable with both stimuli. This experiment also shows that
the number of tetramer-stained cells remains high in phyco-
erythrin (PE) fluorescence under this assay system. Internal-
ization of tetramer occurs normally within a few minutes of
staining (31), and strong signals appear to be intact at the end
of the assay in both cytokine-positive and cytokine-negative
FIG. 3. Functional analysis of a long-term antiviral CD8
T-lymphocyte response. (a) Peptide-stimulated synthesis of cytokines in subject E:
comparison of HCV and control responses. Experiments were performed exactly as in Fig. 1c. The CD8
population is shown, and the proportions
within the tetramer-positive populations that stain positive for intracellular cytokine are indicated. The upper panels represent stimulation with
HCV peptide NS3 1073 (also indicated as “peptide 11” in the FACS plot title line), and the middle and lower panels represent HLA-A2-restricted
peptides from CMV and EBV, respectively. (b) PMA-ionomycin-stimulated TNF-synthesis in patient E. Experiments were performed exactly
as in Fig. 2a. The CD8
population is shown, and the proportions within the tetramer-positive and tetramer-negative populations positive for
intracellular cytokine are indicated.
VOL. 75, 2001 CYTOKINE RELEASE BY HCV-SPECIFIC CD8
T LYMPHOCYTES 5555
subsets with this protocol. This experiment also demonstrates
the lack of cytokine secretion by tetramer alone in the presence
of costimulatory antibodies.
Analysis of TCR Vusage in acutely expanded HCV-specific
CD8
T-lymphocyte populations. We addressed whether the
HCV-specific CD8
phenotype was the result of expansion of
a single aberrant clone by ex vivo analysis of Vusage with a
panel of V-specific MAbs in the large expansions in subjects
A to D. Only in subject B was an expansion of a single V-
bearing subset seen (Fig. 2d, right panel), comprising 30% of
the tetramer-positive population at the first time point. This
was not seen in the other patients (for example, subject A [Fig.
2d, left panel]) and was not sustained in subject B, in whom the
proportion dropped to about 10% by week 14 (data not shown).
Thus, the observed phenotype of weak or absent cytokine
secretion upon stimulation appears to extend across popula-
tions of CD8
T lymphocytes targeting distinct epitopes and
using distinct TCRs.
Analysis of lytic function and perforin staining ex vivo. We
also had the opportunity with a single patient (subject A) to
address whether the cells identified at the peak of activation
during acute hepatitis were directly cytotoxic ex vivo. In this
situation, at the first time point, 3% of CD8
T lymphocytes
were NS3 1073 specific and were highly activated (92% CD38
high and 64% MHC class II high), as previously observed in
other patients (19) (Fig. 1b). Nevertheless at this time point,
no specific lysis of peptide-pulsed targets was observed in a 5-h
or extended assay for chromium release, even though the same
targets were readily lysed by a specific clone (data not shown).
Consistent with this finding, the cells were low in perforin
(1.5% positive) and high in CD27 (90% positive), a phenotype
which has been associated with a low lytic capacity and what
has been described as an “immature” or “early differentiation”
phenotype (2). Of eight responses tested for perforin staining,
all were 10% positive (mean, 3%; range, 0 to 10%) (data not
shown).
Analysis of function of CD8
T lymphocytes ex vivo in pa-
tients later after infection. We and others have previously
reported that levels of virus-specific CD8
lymphocytes (as-
sessed by tetramer) in patients identified outside the setting of
acute disease are generally very low, which renders analyses of
phenotype and function technically difficult without in vitro
manipulation (14, 19, 21). To further address the question of
whether the dysfunction of these cells is sustained long term,
we screened blood from another 56 HLA-A2-positive subjects
from time points outside the first 24 months of infection. This
was performed with tetramers containing the four different
HLA-A2-restricted peptides exactly as used in subjects A to D
(19–21). Where appropriate, we also tested subjects by using
HLA-B8 and -B7 tetramers (see Materials and Methods). We
obtained samples from a further three such individuals (two
PCR negative and one PCR positive) with expansions of HCV-
specific HLA-A2 tetramer-positive cells representing 0.1%
of CD8
lymphocytes in whom intracellular cytokine staining
could be assessed after stimulation.
Examples of such an analysis (subject E) are shown in Fig. 3a
and b. These show, respectively, a profound defect in synthe-
sis of both TNF-and IFN-after peptide stimulation
and a relative defect in staining for TNF-after maximal
(PMA-ionomycin) stimulation, compared to CMV and EBV
responses. Subject E had been PCR negative after IFN-treat-
ment 5 years previously, and thus the defect cannot be attrib-
uted to ongoing viremia. It was sustained over a period of 1
year, with the relatively large HCV-specific tetramer-positive
population (0.3% of CD8
lymphocytes) maintained in a qui-
escent state (low in CD38 and HLA class II; similar to the
EBV- and CMV-specific populations; data not shown).
Internal and group comparisons between HCV-specific and
control responses. The relative lack of cytokine secretory ca-
pacity in HCV-specific CD8
T lymphocytes was further em-
phasized by comparisons with EBV- and CMV-specific re-
sponses within and between individuals. In Fig. 4 (upper
panel), the synthesis of IFN-after PMA stimulation from
HCV-specific CD8
lymphocytes is compared with that from
FIG. 4. Overall comparisons of HCV function and control re-
sponses. (Upper panel) Internal comparison. Five HCV antibody-
positive subjects (all HCV PCR negative) for whom HCV and control
responses were available were tested simultaneously for IFN-staining
after PMA-ionomycin stimulation, and the proportion of tetramer-
positive cells was calculated as in Fig. 2a, upper panels. Each HCV
response was compared with the EBV and/or CMV response in the
same individual (a total of eight comparisons). (Lower panel) Group
comparison. Cytokine synthesis from HCV tetramer-positive popula-
tions in seven HCV antibody-positive subjects (five PCR negative)
were tested exactly as described above (left-hand group, marked HCV)
and compared with EBV and/or CMV responses within themselves
and in seven normal controls (right-hand group). CMV and EBV
responses from HCV antibody-positive subjects are shown by solid
circles, and those from control subjects are shown by open circles.
5556 GRUENER ET AL. J. VIROL.
EBV- and CMV-specific populations in all of those individuals
in whom both responses were present. This shows a marked
skewing of the responses in favor of cytokine release from the
non-HCV-specific CD8
populations (P0.002). In some
individuals (as in subject A), more than one HCV-specific
response was present, and both showed a similar phenotype by
comparison with CMV or EBV.
In Fig. 4 (lower panel), the IFN-responses after PMA
stimulation are compared between HCV tetramer-positive
populations and non-HCV (EBV or CMV) populations in
seven HCV antibody-positive (dark dots) and seven control
subjects (clear dots). The overall level of responsiveness is
again lower at the population level (mean, 13.5% versus 60%;
P0.0001). There was no significant difference between non-
HCV (EBV and/or CMV) responses in HCV antibody-positive
and antibody-negative subjects.
DISCUSSION
These data indicate that HCV-specific CD8
T lymphocytes
possess a distinct phenotype, which may be maintained well
after virus is cleared from blood. Large populations of HCV-
specific CD8
lymphocytes are induced early and are highly
activated, as judged by surface expression of appropriate
marker molecules (21) (Fig. 1b), but have various degrees of
impairment of antiviral functions, as assessed in vitro. These
include an impaired ability to respond to peptide and to mi-
togen and are most obvious at the level of secretion of antiviral
cytokines.
The essential differences in response dynamics or function
dictating clearance versus persistence of HCV were not re-
vealed in this study. Relative defects in cytokine secretion were
seen in those who had cleared virus in the long term, and levels
of intracellular perforin were universally low among HCV-
specific CD8
lymphocytes. It is possible that the essential
mechanisms that control HCV in the long term lie outside of
these conventional functions or indeed are displayed by some
other subset of immune mediators, as has been clearly dem-
onstrated in murine models (26). An alternative explanation is
that the circulating CD8
lymphocytes represent an unusual
subset that differs from the truly functional lymphocytes. While
this seems likely during chronic hepatitis infection, where com-
partmentalization in the liver has been observed (23), it seems
unlikely in situations in which virus has been cleared and no
hepatitis is detectable.
These populations differ from previously identified dysfunc-
tional CD8
lymphocytes in that the phenotype is sustained
(21) and may occur in the presence of adequate CD4
help
(33), and the surface phenotype of the lymphocytes is other-
wise normal (e.g., CD45RO high; data not shown) (22). The
stability of these populations, compared with the dysfunction
seen in lymphocytes undergoing exhaustion (10), indicates a
long-lasting influence on function induced by the virus in an
antigen-specific manner. It is possible that the inducing envi-
ronment or potentially some influence on antigen-presenting
cell function (15) is responsible. For example, in murine mod-
els, a similar sustained phenotype has been seen after vaccina-
tion in the presence of interleukin-10 (5). In this context, a
description of “stunted” as opposed to “stunned” may be more
appropriate. How this may then have an impact on the persis-
tence of HCV remains to be determined, especially in the
context of the balance with other mediators of immunity (6, 9).
Methods to reverse this defect or to induce new populations of
functional T lymphocytes should be explored as part of immu-
notherapeutic strategies and vaccine design.
ACKNOWLEDGMENTS
Norbert Gruener and Franziska Lechner contributed equally to this
paper.
Funding was obtained from the Wellcome Trust, the Wilhelm-Sand-
er-Stiftung and the European Union (5th Framework, HCVacc, grant
no. QLK2-CT-1999-00356), the Deutsche Forschungsgemein, the Aus-
tralian Red Cross, the National Institutes of Health, and the Doris
Duke Charitable Foundation.
We are grateful to Jane Collier and the staff of the hepatitis and
immunology clinics at the John Radcliffe Hospital for providing pa-
tient samples to screen; Mike Bunce for tissue typing; Philip Goulder,
Rod Dunbar, Richard Cornall, Victor Appay, and Andrew McMichael
for helpful discussions; and A. Morse for preparation of the manu-
script. We also thank Carmen Amsel, Barbara Becker, Jutta Do¨hr-
mann, and Marion Satzger for excellent technical assistance and per-
sonal encouragement.
REFERENCES
1. Altman, J., P. A. H. Moss, P. Goulder, D. Barouch, M. McHeyzer-Williams,
J. I. Bell, A. J. McMichael, and M. M. Davis. 1996. Direct visualization and
phenotypic analysis of virus-specific T lymphocytes in HIV-infected individ-
uals. Science 274:94–96.
2. 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 lytic function. J. Exp. Med. 192:63–75.
3. Callan, M., L. Tan, N. Annels, G. Ogg, J. Wilson, C. O’Callaghan, N. Steven,
A. McMichael, and A. Rickinson. 1998. Direct visualisation of antigen spe-
cific CD8T cells during the primary immune response to EBV in vivo. J.
Exp. Med. 187:1395–1402.
4. Cerny, A., J. McHutchinson, C. Pasquellini, M. Brown, M. Brothers, B.
Grabschied, P. Fowler, M. Houghton, and F. Chisari. 1995. CTL response to
HCV-derived peptides containing the HLA-A2.1 binding motif. J. Clin.
Investig. 95:521–530.
5. Chun, S., M. Daheshia, S. Lee, S. K. Eo, and B. T. Rouse. 1999. Distribution
fate and mechanism of immune modulation following mucosal immunisa-
tion. J. Immunol. 163:2393–2402.
6. Ciurea, A., P. Klenerman, L. Hunziker, E. Horvath, B. M. Senn, A. F.
Ochsenbein, H. Hengartner, and R. M. Zinkernagel. 2000. Viral persistence
in vivo through selection of neutralizing antibody-escape variants. Proc. Natl.
Acad. Sci. USA 97:2749–2754.
7. Cramp, M. E., S. Rossol, S. Chokshi, P. Carucci, R. Williams, and N. V.
Naoumov. 2000. Hepatitis C virus-specific T-cell reactivity during interferon
and ribavirin treatment in chronic hepatitis C. Gastroenterology 118:346–
355.
8. Diepolder, H. M., R. Zachoval, R. M. Hoffmann, E. A. Wierenga, T. San-
tantonio, M. C. Jung, D. Eichenlaub, and G. R. Pape. 1995. Possible mech-
anism involving T-lymphocyte response to non-structural protein 3 in viral
clearance in acute hepatitis C virus infection. Lancet 346:1006–1007.
9. Farci, P., A. Shimoda, A. Coiana, G. Diaz, G. Peddis, J. C. Melpolder, A.
Strazzera, D. Y. Chien, S. J. Munoz, A. Balestrieri, R. H. Purcell, and H. J.
Alter. 2000. The outcome of acute hepatitis C predicted by the evolution of
the viral quasispecies. Science 288:339–344.
10. Gallimore, A., A. Glitheroe, A. Godkin, A. Tissot, H. Hengartner, T. Elliott,
and R. Zinkernagel. 1998. Induction and exhaustion of LCMV-specific CTL
visualised using soluble tetrameric MHC-peptide complexes. J. Exp. Med.
187:1383–1393.
11. Gerlach, J., H. Diepolder, M.-C. Jung, N. Gruener, W. Schraut, R. Zachoval,
R. Hoffman, C. Schirren, T. Santantonio, and G. Pape. 1999. Recurrence of
HCV after loss of virus specific CD4T cell response in acute hepatitis C.
Gastroenterology 117:933–941.
12. Gillespie, G. M. A., M. R. Wills, V. Appay, C. O’Callaghan, M. Murphy, N.
Smith, P. Sissons, S. Rowland-Jones, J. I. Bell, and P. A. H. Moss. 2000.
Functional heterogeneity and high frequencies of cytomegalovirus-specific
CD8
T lymphocytes in healthy seropositive donors. J. Virol. 74:8140–8150.
13. Guidotti, L., R. Rochford, J. Chung, M. Shapiro, R. Purcell, and F. Chisari.
1999. Viral clearance without destruction of infected cells during acute HBV
infection. Science 284:825–829.
14. He, X.-S., B. Rehermann, F. Lopez-Labrador, J. Boisvert, R. Cheung, J.
Mumm, H. Wedermeyer, M. Berenguer, T. Wright, and M. Davis. 1999.
VOL. 75, 2001 CYTOKINE RELEASE BY HCV-SPECIFIC CD8
T LYMPHOCYTES 5557
Quantitative analysis of HCV-specific CD8T cells in peripheral blood and
liver using peptide-MHC tetramers. Proc. Natl. Acad. Sci. USA 96:5692–
5697.
15. Hiasa, Y., N. Horiike, S. M. Akbar, I. Saito, T. Miyamura, Y. Matsuura, and
M. Onji. 1998. Low stimulatory capacity of lymphoid dendritic cells express-
ing hepatitis C proteins. Biochem. Biophys. Res. Commun. 249:90–95.
16. Klenerman, P., F. Lechner, M. Kantzanou, A. Ciurea, H. Hengartner, and R.
Zinkernagel. 2000. Viral escape and the failure of cellular immune re-
sponses. Science 289:2003a.
17. Koziel, M. J., D. Dudley, N. Afdhal, A. Grakoui, C. M. Rice, Q. L. Choo, M.
Houghton, and B. D. Walker. 1995. HLA class I-restricted cytotoxic T lym-
phocytes specific for hepatitis C virus. Identification of multiple epitopes and
characterization of patterns of cytokine release. J. Clin. Investig. 96:2311–
2321.
18. Lalvani, A., R. Brookes, S. Hambleton, W. J. Britton, A. V. Hill, and A. J.
McMichael. 1997. Rapid effector function in CD8memory T cells. J. Exp.
Med. 186:859–865.
19. Lechner, F., N. H. Gruener, S. Urbani, J. Uggeri, T. Santantonio, A. R.
Kammer, A. Cerny, R. Phillips, C. Ferrari, G. R. Pape, and P. Klenerman.
2000. CD8T lymphocyte responses are induced during acute hepatitis C
virus infection but are not sustained. Eur. J. Immunol. 30:2479–2487.
20. Lechner, F., J. Sullivan, H. Spiegel, D. Nixon, B. Ferrari, A. Davis, B.
Borkowski, H. Pollack, E. Barnes, G. Dusheiko, and P. Klenerman. 2000.
Why do cytotoxic T lymphocytes fail to eliminate HCV? Lessons from
studies using MHC class I tetramers. Phil. Trans. R. Soc. Lond. B. 355:1085–
1092.
21. Lechner, F., D. K. Wong, P. R. Dunbar, R. Chapman, R. T. Chung, P.
Dohrenwend, G. Robbins, R. Phillips, P. Klenerman, and B. D. Walker.
2000. Analysis of successful immune responses in persons infected with
hepatitis C virus. J. Exp. Med. 191:1499–1512.
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. Nat. Med. 5:677–685.
23. Minutello, M., P. Pileri, D. Unutmaz, S. Censini, G. Kuo, M. Houghton, M.
Brunetto, F. Bonino, and S. Abrignani. 1993. Compartmentalization of T
lymphocytes to the site of disease: intrahepatic T cells specific for the NS4
protein of HCV in patients with chronic hepatitis C. J. Exp. Med. 178:17–25.
24. Missale, G., R. Bertoni, V. Lamonaca, A. Valli, M. Massari, C. Mori, M.
Rumi, M. Houghton, F. Fiaccadori, and C. Ferrari. 1996. Different clinical
behaviours of acute HCV infection are associated with different vigor of the
anti-viral T cell response. J. Clin. Investig. 98:706–714.
25. Naoumov, N. V. 1999. Hepatitis C virus-specific CD4() T cells: do they help
or damage? Gastroenterology 117:1012–1014.
26. Planz, O., S. Ehl, E. Furrer, E. Horvath, M. Brundler, H. Hengartner, and
R. Zinkernagel. 1997. A critical role for neutralising antibody-producing B
cells, CD4cells and interferons in persistent infections of mice with
LCMV: implications for adoptive immunotherapy of virus carriers. Proc.
Natl. Acad. Sci. USA 94:6874–6879.
27. Rehermann, B., K. Chang, J. McHutchinson, R. Kokka, M. Houghton, and
F. Chisari. 1996. Quantitative analysis of the peripheral blood CTL response
in patients with chronic HCV infection. J. Clin. Investig. 98:1432–1440.
28. Rosen, H. R., D. J. Hinrichs, D. R. Gretch, M. J. Koziel, S. Chou, M.
Houghton, J. Rabkin, C. L. Corless, and H. G. Bouwer. 1999. Association of
multispecific CD4() response to hepatitis C and severity of recurrence after
liver transplantation. Gastroenterology 117:926–932.
29. Takaki, A., M. Wiese, G. Maertens, E. Depla, U. Seifert, A. Liebetrau, J. L.
Miller, M. P. Manns, and B. Rehermann. 2000. Cellular immune responses
persist and humoral responses decrease two decades after HCV infection.
Nat. Med. 6:578–582.
30. Thursz, M., R. Yallop, R. Goldin, C. Trepo, and H. C. Thomas. 1999.
Influence of MHC class II genotype on outcome of infection with hepatitis
C virus. The HENCORE group. Hepatitis C European Network for Coop-
erative Research. Lancet 354:2119–2124.
31. Whelan, J. A., P. R. Dunbar, D. A. Price, M. A. Purbhoo, F. Lechner, G. S.
Ogg, G. Griffiths, R. E. Phillips, V. Cerundolo, and A. K. Sewell. 1999.
Specificity of CTL interactions with peptide-MHC class I tetrameric com-
plexes is temperature dependent. J. Immunol. 163:4342–4348.
32. Wong, D., D. Dudley, N. Afdhal, J. Dienstag, C. Rice, L. Wang, M. Houghton,
B. Walker, and M. Koziel. 1998. Liver derived CTL in HCV infection:
breadth and specificity of responses in a cohort of patients with chronic
infection. J. Immunol. 160:1479–1488.
33. Zajac, A., J. Blattman, K. Murali-Krishna, D. Sourdive, M. Suresh, J.
Altman, and R. Ahmed. 1998. Viral immune evasion due to persistence of
activated T cells without effector function. J. Exp. Med. 188:2205–2213.
5558 GRUENER ET AL. J. VIROL.
... Chronic HCV infection disrupts many innate and adaptive immune cells, including cytotoxic CD8 T cells (15)(16)(17)(18)(19)(20). In the acute stage of HCV infection, weak and transient responses of HCVspecific CD8 T cells predict chronicity (21,22). ...
Article
Full-text available
Background The impact of chronic hepatic infection on antigen non-specific immune cells in circulation remains poorly understood. We reported lasting global hyperfunction of peripheral CD8 T cells in HCV-infected individuals with cirrhosis. Whether gene expression patterns in bulk CD8 T cells are associated with the severity of liver fibrosis in HCV infection is not known. Methods RNA sequencing of blood CD8 T cells from treatment naïve, HCV-infected individuals with minimal (Metavir F0-1 ≤ 7.0 kPa) or advanced fibrosis or cirrhosis (F4 ≥ 12.5 kPa), before and after direct-acting antiviral therapy, was performed. CD8 T cell function was assessed by flow cytometry. Results In CD8 T cells from pre-DAA patients with advanced compared to minimal fibrosis, Gene Ontology analysis and Gene Set Enrichment Analysis identified differential gene expression related to cellular function and metabolism, including upregulated Hedgehog (Hh) signaling, IFN-α, -γ, TGF-β response genes, apoptosis, apical surface pathways, phospholipase signaling, phosphatidyl-choline/inositol activity, and second-messenger-mediated signaling. In contrast, genes in pathways associated with nuclear processes, RNA transport, cytoskeletal dynamics, cMyc/E2F regulation, oxidative phosphorylation, and mTOR signaling, were reduced. Hh signaling pathway was the top featured gene set upregulated in cirrhotics, wherein hallmark genes GLI1 and PTCH1 ranked highly. Inhibition of Smo-dependent Hh signaling ablated the expression of IFN-γ and perforin in stimulated CD8 T cells from chronic HCV-infected patients with advanced compared to minimal fibrosis. CD8 T cell gene expression profiles post-DAA remained clustered with pre-DAA profiles and disparately between advanced and minimal fibrosis, suggesting a persistent perturbation of gene expression long after viral clearance. Conclusions This analysis of bulk CD8 T cell gene expression in chronic HCV infection suggests considerable reprogramming of the CD8 T cell pool in the cirrhotic state. Increased Hh signaling in cirrhosis may contribute to generalized CD8 T cell hyperfunction observed in chronic HCV infection. Understanding the lasting nature of immune cell dysfunction may help mitigate remaining clinical challenges after HCV clearance and more generally, improve long term outcomes for individuals with severe liver disease.
... direct-acting antiviral (DAA)-mediated HCV clearance, dietary intervention in NAFLD and alcohol abstinence in AUD), whether immune dysfunctions are reversible is unknown. Chronic HCV infection disrupts immune function, affecting many innate and adaptive immune cells including cytotoxic CD8 + T cells 4,5,6,7,8,9 . Despite the spectacular results of DAAs, it is not clear if sustained virologic response (SVR) parallels restored immune function. ...
Preprint
Full-text available
Immune dysfunction, both depression and hyperactivation, in liver disease contributes to significant morbidities and mortalities, depending on liver damage severity and etiology. The underlying causes of immune dysfunction in advanced liver disease, whether pathogen or host-mediated, remain unclear. We reported lasting generalized CD8 T cell hyperfunction in individuals with advanced liver fibrosis in chronic HCV infection. The separation of viral and fibrosis-driven effects or the association of this phenomenon with clinical outcomes of advanced liver fibrosis remains to be determined. Here, a hepatotoxic murine model of liver fibrosis was used to decouple liver fibrosis from viral infection. Carbon tetrachloride (CCl 4 )-treated mice presented progressive liver fibrosis within ≈12 weeks, resulting in severe diffuse fibrosis, focal necrosis and surrounding mixed inflammation; pathology similar to that of chronic HCV infection. Taking advantage of this model, we investigated if liver fibrosis caused systemic CD8 T cell hyperfunction and evaluated its impact on host immune response. At peak liver fibrosis, circulating CD8+ T cells presented increased expression of IFN-γ and granzyme B (GrB) in comparison to control animals. CD8 T cell hyperfunction arose by 8 weeks of CCl 4 treatment and was sustained with continued liver insult. As a result, fibrotic mice were unable to resist an ectopic tumour challenge and were less responsive to immunotherapy. Furthermore, CD8 T cell dysfunction was observed in other contexts of chronic liver insult such as high fat diet-induced liver steatosis, even in the absence of significant fibrosis. Collectively, this study shows the impact of chronic liver insult on systemic CD8 T cell function and its association with impaired immune response, such as tumour surveillance.
... Isolating exhausted cells directly from human tissue is also a commonly used approach. Exhausted T cells in humans were [16,[67][68][69][70][71][72][73][74][75][76][77]]. Around the same time, exhausted tumour infiltrating T cells (defined at the time by inhibitory receptor upregulation and reduced cytokine production when stimulated ex vivo) were isolated, and continue to be identified, from a variety of tumours (e.g. ...
Article
Full-text available
‘Exhaustion’ is a term used to describe a state of native and redirected T-cell hypo-responsiveness resulting from persistent antigen exposure during chronic viral infections or cancer. Although a well-established phenotype across mice and humans, exhaustion at the molecular level remains poorly defined and inconsistent across the literature. This is, in part, due to an overreliance on surface receptors to define these cells and explain exhaustive behaviours, an incomplete understanding of how exhaustion arises, and a lack of clarity over whether exhaustion is the same across contexts, e.g. chronic viral infections versus cancer. With the development of systems-based genetic approaches such as single-cell RNA-seq and CRISPR screens applied to in vivo data, we are moving closer to a consensus view of exhaustion, although understanding how it arises remains challenging given the difficulty in manipulating the in vivo setting. Accordingly, producing and studying exhausted T-cells ex vivo are burgeoning, allowing experiments to be conducted at scale up and with high throughput. Here, we first review what is currently known about T-cell exhaustion and how it’s being studied. We then discuss how improvements in their method of isolation/production and examining the impact of different microenvironmental signals and cell interactions have now become an active area of research. Finally, we discuss what the future holds for the analysis of this physiological condition and, given the diversity of ways in which exhausted cells are now being generated, propose the adoption of a unified approach to clearly defining exhaustion using a set of metabolic-, epigenetic-, transcriptional-, and activation-based phenotypic markers, that we call ‘M.E.T.A’.
... It is now well-accepted that most T cells in such circumstances adopt a unique differentiation trajectory-exhaustion. 316,317 Exhausted T (Tex) cells have been identified in many high grade chronic viral infections, such as HIV, HBV, HCV, and LCMV-Clone 13 strain, [318][319][320][321] and in almost every mouse and human cancer. 322,323 A wealth of recent studies at single-cell level have revealed that Tex cells constitute heterogenous populations with distinct transcriptional, epigenetic and functional signatures, playing critical roles in protecting against infections and tumors. ...
Article
Full-text available
T cells are crucial for immune functions to maintain health and prevent disease. T cell development occurs in a stepwise process in the thymus and mainly generates CD4+ and CD8+ T cell subsets. Upon antigen stimulation, naïve T cells differentiate into CD4+ helper and CD8+ cytotoxic effector and memory cells, mediating direct killing, diverse immune regulatory function, and long-term protection. In response to acute and chronic infections and tumors, T cells adopt distinct differentiation trajectories and develop into a range of heterogeneous populations with various phenotype, differentiation potential, and functionality under precise and elaborate regulations of transcriptional and epigenetic programs. Abnormal T-cell immunity can initiate and promote the pathogenesis of autoimmune diseases. In this review, we summarize the current understanding of T cell development, CD4+ and CD8+ T cell classification, and differentiation in physiological settings. We further elaborate the heterogeneity, differentiation, functionality, and regulation network of CD4+ and CD8+ T cells in infectious disease, chronic infection and tumor, and autoimmune disease, highlighting the exhausted CD8+ T cell differentiation trajectory, CD4+ T cell helper function, T cell contributions to immunotherapy and autoimmune pathogenesis. We also discuss the development and function of γδ T cells in tissue surveillance, infection, and tumor immunity. Finally, we summarized current T-cell-based immunotherapies in both cancer and autoimmune diseases, with an emphasis on their clinical applications. A better understanding of T cell immunity provides insight into developing novel prophylactic and therapeutic strategies in human diseases.
... CCL3 (MIP-1α), CCL4 (MIP-1β), CCL5 (RANTES), and CCL8 ligandsˈ receptors are CCR1 and CCR5. CCR5 positive T cells can be detected with high frequency during acute HCV infection as they are an important part of antiviral immunity (Gruener et al., 2001). Differential expression of CCR5 and CCR1 chemokine receptors has been detected in the peripheral blood and intrahepatic T cells of HCV-infected individuals. ...
Chapter
Full-text available
Abstract Objective: A literature review was done to understand the possible pathophysiology of COVID-19 progression in those comorbid patients, who are considered at high-risk of severe COVID-19 outcomes. Background: An outbreak of a novel Coronavirus (nCOV), termed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), occurred in Wuhan, Hubei Province, China, in December 2019. Ever since December 2019, the virus’s transmission has escalated dramatically and has been widespread across the globe, affecting humans of all ethnicities, age groups, race, and genders. Even though pulmonary involvements are predominant, the disease has a wide range of clinical manifestations, from a common cold to multi-organ failure and eventually death. It is also noted that older patients with preexisting comorbidities, particularly those who are in the age group of 65 years old and above, are infected severely and have increased hospital admission rates. In respect to the demography, the African American population, in vulnerable working and/or living conditions, are at the highest risk of being infected with COVID-19. Methods: An electronic literature search was done using PubMed, Google Scholar, Research Gate, ScienceDirect and Mendeley. Discussion: A wide range of comorbidities are explored in this paper. 1) Chronic obstructive pulmonary disease (COPD) & asthma, 2) Cardiovascular diseases (CVD) & hypertension (HTN), 3) Obesity, 4) Diabetes & Thyroid, 5) Acute liver disease and 6) Acute kidney disease. Furthermore, the impact of COVID-19 on the global population is also discussed. Conclusion: In the mentioned comorbidities, we see that adipose tissue plays a vital role and has a high tendency to predispose patients to become infected with COVID-19. We observed that adipose tissue can contribute to the development of pulmonary fibrosis, cytokine storms, liver injuries, renal coagulation and other coagulopathic events in patients. Any vaccination given to the public has a higher effective percentage than no vaccination, regardless of the variant. Vaccinations can prevent severe progression and hospitalization in comorbid and non-comorbid populations alike. Keywords: SARS-CoV-2, Coronavirus, Pathophysiology, Comorbidity, Risk factors.
... For the first time, this process was detected in CD8+T cells of mice infected with lymphocytic choriomeningitis virus (LCMV) as, not only were these cells not able to remove infected cells but also they did not cause any immunopathological death (11). After the discovery of this phenomenon in mice, studies were carried out to identify this phenomenon in primates such as humans, which led to the discovery of this phenomenon in human infections with Simian immunodeficiency virus (SIV), Human Immunodeficiency Virus (HIV) (12), Hepatitis B virus (HBV) (13), Hepatitis C virus (HCV) (14), and human Tcell lymphotropic virus (HTLV) (15), and bacterial infection with Mycobacterium tuberculosis (16), and fungal infections such as Candida albicans and Metastatic cancers (17). Also, in a recent study, the presence of a group of T cells with the exhaustion phenotype was detected in patients with common variable immune deficiency (CVID) (18). ...
Article
Full-text available
During chronic infections, a distinct physiological condition named “exhaustion” arises that is associated with the dysfunctionality in immune cells and their eventual removal from the environment, which evidently help the progress of infection or tumors in human or animal bodies. This state of immune cells could be under the control of different elements such as the antigen load, help from inhibitory cells, lack of costimulatory signals and etc. Exhaustion that has been found in different immune system cells, is usually accompanied by impaired effector function and proliferation of immune cells, poor memory recall, upregulation of inhibitory molecules, compromised metabolism and altered transcription program, and is considered a reversible process, unlike other physiological states like anergy or senescence, organized through the blockage of several factors. Although the emergence of these cells in viral infections and cancer is an undesirable event, the importance of the presence of exhausted cells in autoimmune diseases and organ transplantation is highlighted as a positive change. In this review, we aimed to determine the occurrence of this process in different immune cells, the characteristics obtained by these cells, effective and primitive factors on exhaustion, metabolic and transcriptional cell changes, and the use of these cells in autoimmune diseases and organ transplantation.
... Recent data on immune responses in the chronic phase of infection suggest that a therapeutic vaccine of stimulating functional CD4+ and CD8+ T cell responses in the chronic carrier may benefit [52,53]. Targets for DNA vaccine developments are mainly envelope glycoprotein (E1 or E2), core antigen and nonstructural proteins (NS3, NS4, NS5) [54]. ...
Article
Full-text available
Hepatitis C virus (HCV) is a major cause of chronic hepatitis, liver cirrhosis and hepatocellular carcinoma worldwide. A combination therapy comprising pegylated interferon and ribavirin currently represents the most effective therapy for chronic HCV infection. The limitations of this current therapy mainly its efficacy and significant side effects have prompted the development of new drugs. Few categories of therapeutic agents appear promising for future therapy, e.g. novel interferons, ribavirin analogs, antisense oligonucleotides, short interfering RNAs, ribozymes, enzyme inhibitors, immunomodulatory agents, antifibrotic agents, therapeutic vaccines and antibodies. Few drugs belong to afore-mentioned categories have already reached the different clinical phases of development. The present article highlights the status of current available therapies and emerging drugs for the treatment of hepatitis C.
Preprint
Full-text available
The impact of chronic hepatic infection on antigen non-specific immune cells in circulation is not well understood and may influence long term health. We reported lasting global hyperfunction of circulating CD8 T cells in HCV-infected individuals with cirrhosis. Whether gene expression patterns in bulk CD8 T cells are associated with the severity of liver fibrosis in HCV infection is not known. RNA sequencing of blood CD8 T cells from treatment-naive, HCV-infected individuals with minimal (Metavir F0-1 ≤ 7.0 kPa) or advanced fibrosis or cirrhosis (F4 ≥ 12.5 kPa), before and after direct-acting antiviral therapy, was performed. Principal component analyses determined robust differences in over 350 genes expressed by CD8 T cells from HCV-infected individuals with minimal or advanced fibrosis and data suggests this remains relatively stable after viral clearance. Gene ontology analyses identified disaggregated gene expression related to cellular metabolism, including upregulated phospholipase, phosphatidyl-choline/inositol activity and second-messenger-mediated signaling, while genes in pathways associated with nuclear processes, RNA transport and cytoskeletal dynamics were reduced. Gene Set Enrichment Analysis identified decreased expression of genes regulated by the cMyc and E2f transcription factors in cirrhotics, compared to the minimal fibrosis group, as well as reduced expression of genes linked to oxidative phosphorylation, mTOR signaling, and more. Upregulated gene sets in cirrhotics included IFN-α, -γ, TGF-β response genes, apoptosis and apical surface pathways, among others. The hedgehog (Hh) signaling pathway was the top featured gene set upregulated in cirrhotics. Inhibition of Hh signaling with cyclopamine ablated CD8 T cell IFN-γ and perforin production, suggesting its involvement in hyperfunction. This is the first analysis of bulk CD8 T cell gene expression profiles in HCV infection in the context of liver fibrosis severity, and suggests cirrhosis significantly reprograms the CD8 T cell pool. The novel finding of increased Hh signaling in cirrhosis may contribute to generalized CD8 T cell hyperfunction observed in chronic HCV infection. Understanding the lasting nature of immune cell dysfunction may help mitigate remaining clinical challenges after HCV clearance and more generally, improve long term outcomes for individuals with severe liver disease.
Article
Individuals who spontaneously clear hepatitis C virus (HCV) infection have demonstrated evidence of partial protective immunity, whereas treatment‐induced clearance provides little or no protection against reinfection. We aimed to investigate whether treatment of acute HCV infection with direct‐acting antivirals (DAA) prevents establishment of, or reverses, T cell exhaustion, leading to a virus‐specific T cell immune profile more similar to that seen in spontaneous clearance. The magnitude and breadth of HCV‐specific T cell responses before and after DAA or interferon‐based therapy in acute or chronic HCV were compared to those of participants with spontaneous clearance of infection, using Enzyme‐linked Immunospot (ELISPOT). PBMCs were available for 55 patients comprising 4 groups: spontaneous clearance (n=17), acute interferon (n=14), acute DAA (n=13) and chronic DAA (n=11). After controlling for sex, the magnitude of post‐treatment HCV‐specific responses after acute DAA treatment was greater than after chronic DAA or acute IFN treatment and similar to those found in spontaneous clearers. However, spontaneous clearers responded to more HCV peptide pools indicating greater breadth of response. In conclusion, early treatment with DAAs may prevent or reverse some degree of immune exhaustion and result in stronger HCV‐specific responses post‐treatment. However, individuals with spontaneous clearance had broader HCV‐specific responses.
Article
Full-text available
DNA vaccination has been widely studied in several models of vaccination and in the treatment of inflammatory diseases, even though the mechanism involved is still unclear. This report demonstrates that mucosal administration of plasmid DNA leads to rapid and widespread distribution around the body. Dissemination likely occurred via the bloodstream because plasmid DNA was present in blood plasma. The plasmid DNA was also detectable in several tissues including draining lymph nodes, spleen, liver, bone marrow, and even the dermis of ear pinnae. Except for the site of administration, plasmid DNA was no longer detectable in tissues after 3 wk postadministration. RNA and protein expression was also found in the tissues and bloodstream. Animals previously primed by HSV infection and subsequently given IL-10 DNA via the nasal mucosa, showed diminished Ag-induced delayed type hypersensitivity reactions for up to 5 wk posttreatment. The mechanism of modulation involved diminished the Ag-specific proliferation and production of Th1 cytokines. The Ag-specific silencing effects persisted beyond the duration of detectable plasmid encoded protein and was maintained upon adoptive transfer of T cells into a plasmid-free environment. The silenced T cells were not a source of IL-10, and their anergic state was reversible by exposure to Ag in the presence of exogenous IL-2.
Article
Full-text available
It is believed that the hepatitis C virus (HCV)-specific CD8+ cytotoxic T lymphocytes (CTLs) play a role in the development of liver cell injury and in the clearance of the virus. To develop a direct binding assay for HCV-specific CTLs, we generated two peptide-MHC tetramers by using the recombinant HLA A2.1 molecule and A2-restricted T cell epitopes of the HCV NS3 protein. With these reagents we are able to detect specific CD8+ cells in the blood of 15 of 20 HLA-A2+, HCV-infected patients, at a frequency ranging from 0.01% to 1.2% of peripheral CD8+ T cells. Phenotypic analysis of these specific cells indicated that there is a significant variation in the expression of the CD45 isoforms and CD27 in different patients. A 6-hour incubation of one patient’s blood with NS3 peptides resulted in the activation of the epitope-specific CD8+ cells, as indicated by their expression of CD69 and IFN-γ. We also detected NS3-specific CD8+ T cells in the intrahepatic lymphocyte population isolated from liver biopsies of two HCV-infected patients. The frequency of these specific CD8+ cells in the liver was 1–2%, at least 30-fold higher than in the peripheral blood. All of the intrahepatic NS3-specific CD8+ T cells were CD69+, suggesting that they were activated CTLs. Direct quantitation and characterization of HCV-specific CTLs should extend our understanding of the immunopathogenesis and the mechanism of clearance or persistence of HCV.
Article
Full-text available
As acute hepatitis C virus (HCV) infection is clinically inapparent in most cases, the immunologic correlates of recovery are not well defined. The cellular immune response is thought to contribute to the elimination of HCV-infected cells1 and a strong HCV-specific T-helper-cell (Th) response is associated with recovery from acute hepatitis C (ref. 2). However, diagnosis of resolved hepatitis C is based at present on the detection of HCV-specific antibodies and the absence of detectable HCV RNA, and detailed comparison of the humoral and cellular immune response has been hampered by the fact that patient cohorts as well as HCV strains are usually heterogeneous and that clinical data from acute-phase and long-term follow-up after infection generally are not available. We studied a cohort of women accidentally exposed to the same HCV strain of known sequence3 and found that circulating HCV-specific antibodies were undetectable in many patients 18−20 years after recovery, whereas HCV-specific helper and cytotoxic T-cell responses with an interferon (IFN)--producing (Tc1) phenotype persisted. The data indicate these HCV-specific CD4 + and CD8+ T cells are biomarkers for a prior HCV exposure and recovery. Because of undetectable antibodies against HCV, the incidence of self-limited HCV infections and recovery may be underestimated in the general population.
Article
Full-text available
Cytotoxic T lymphocytes (CTL) are important to the control of viral replication and their presence may be important to disease outcome. An understanding of the spectrum of proteins recognized by hepatitis C virus (HCV)-specific CTL and the functional properties of these cells is an important step in understanding the disease process and the mechanisms of persistent infection, which occurs in the majority of HCV-infected individuals. In this report we identify HCV-specific CTL responses restricted by the HLA class I molecules A2, A3, A11, A23, B7, B8, and B53. The epitopes recognized by these intrahepatic CTL conform to published motifs for binding to HLA class I molecules, although in some cases we have identified CTL epitopes for which no published motif exists. The use of vectors expressing two different strains of HCV, HCV-1 and HCV-H, revealed both strain-specific and cross-reactive CTL. These HCV-specific CTL were shown to produce cytokines including IFN-gamma, TNF-alpha, GM-CSF, IL-8, and IL-10 in an antigen- and HLA class I-specific manner. These studies indicate that the CTL response to HCV is broadly directed and that as many as five different epitopes may be targeted in a single individual. The identification of minimal epitopes may facilitate peptide-specific immunization strategies. In addition, the release of proinflammatory cytokines by these cells may contribute to the pathogenesis of HCV-induced liver damage.
Article
Full-text available
The HLA class I-restricted cytotoxic T lymphocyte (CTL) response is a major defense mechanism in viral infections. It has been suggested that the CTL response may contribute to viral clearance and liver cell injury during hepatitis C virus (HCV) infection. To test this hypothesis requires an understanding of the characteristics of HCV-specific cytotoxic effector cells and identification of the target antigens to which they respond. To begin this process we stimulated peripheral blood mononuclear cells (PBMC) from a group of HLA-A2 positive patients with chronic hepatitis C with a panel of 130 HCV-derived peptides containing the HLA-A2 binding motif. Effector cells were tested for their capacity to lyse HLA-A2-matched target cells that were either sensitized with peptide or infected with a vaccinia virus construct containing HCV sequences. Using this approach we have identified nine immunogenic peptides in HCV, three of which are derived from the putative core protein, three from the nonstructural (NS) 3 domain, two from NS4 and one from NS5. Selected responses were shown to be HLA-A2 restricted, mediated by CD8+ T cells and to recognize endogenously synthesized viral antigen. Unexpectedly, peptide-specific CTL responses could also be induced in sero-negative individuals, suggesting in vitro activation of naive CTL precursors. The precursor frequency of peptide-specific CTL was 10 to 100-fold higher in infected patients compared to uninfected controls, and the responses were greatly diminished by removal of CD45 RO+ (memory) T cells. Further quantitative studies are clearly required to establish whether a correlation exists between the HCV-specific CTL response and the clinical course of this disease. Definition of the molecular targets of the human CTL response to HCV creates this opportunity, and may also contribute to the development of a T cell-based HCV vaccine.
Article
Full-text available
The adult liver is an organ without constitutive lymphoid components. Therefore, any intrahepatic T cell found in chronic hepatitis should have migrated to the liver after infection and inflammation. Because of the little information available on the differences between intrahepatic and peripheral T cells, we used recombinant proteins of the hepatitis C virus (HCV) to establish specific T cell lines and clones from liver biopsies of patients with chronic hepatitis C and compared them with those present in peripheral blood mononuclear cells (PBMC). We found that the protein nonstructural 4 (NS4) was able to stimulate CD4+ T cells isolated from liver biopsies, whereas with all the other HCV proteins we consistently failed to establish liver-derived T cell lines from 16 biopsies. We then compared NS4-specific T cell clones obtained on the same day from PBMC and liver of the same patient. We found that the 22 PBMC-derived T cell clones represent, at least, six distinct clonal populations that differ in major histocompatibility complex restriction and response to superantigens, whereas the 27 liver-derived T cell clones appear all identical, as further confirmed by cloning and sequencing of the T cell receptor (TCR) variable and hypervariable regions. Remarkably, none of the PBMC-derived clones has a TCR identical to the liver-derived clone, and even with polymerase chain reaction oligotyping we did not find the liver-derived clonotypic TCR transcript in the PBMC, indicating a preferential intrahepatic localization of these T cells. Functionally, the liver-derived T cells provided help for polyclonal immunoglobulin (Ig)A production by B cells in vitro that is 10-fold more effective than that provided by the PBMC-derived clones, whereas there is no difference in the help provided for IgM and IgG production. Altogether these results demonstrate that the protein NS4 is highly immunogenic for intrahepatic CD4+ T cells primed by HCV in vivo, and that there can be compartmentalization of some NS4-specific CD4+ T cells to the liver of patients with chronic hepatitis C.
Article
The outcome of infection with hepatitis C virus (HCV) varies substantially among individuals. Host genetic factors are likely to give rise to some of this variability. Polymorphisms in the MHC class II loci may influence the outcome of HCV infection; however, reports of MHC class II allele associations have been inconsistent. We aimed to confirm these associations in a cohort of European patients with different clinical outcomes. The distribution of MHC class II alleles was compared between patients with self-limiting infection (n=85) and matched patients with persistent infection (n=170); between patients with mild (n=321) and severe (n=321) histological injury; and between patients who responded to interferon (n=96) and those who did not (n=192). The results of these comparisons were confirmed with a second-stage study of self-limiting infection (n=52) versus persistent infection (n=152). Self-limiting HCV infection was associated with HLA-DRB1*1101 (odds ratio 2.14 [95% CI 1.11-4.12]; p=0.013) and HLA-DQB1*0301 (2.22 [1.24-3.96], p=0.004). Persistent HCV infection was associated with HLA-DRB1*0701 (2.04 [1.03-4.17], p=0.027), and HLA-DRB4*0101 (2.38 [1.29-4.35], p=0.002). These results were confirmed in the second-stage study. No significant associations (p<0.05 after Bonferroni correction) were found between MHC class II alleles and severe histological injury or response to interferon therapy. Specific MHC class II alleles influence susceptibility or resistance to persistent HCV infection.
Article
In acute hepatitis C virus (HCV) infection only 20-50% of patients spontaneously clear the virus. To characterise the immune reaction during that phase we studied the response of peripheral blood mononuclear cells (PBMC) to the recombinant HCV proteins core, non-structural protein 3 (NS3), NS4, and NS5 in 14 patients with acute hepatitis C. All eight patients with self-limited disease compared with two of six with evolving chronic infection showed an NS3- specific PBMC response (p = 0.015). Of 65 patients with established chronic hepatitis C, five showed a PBMC response to NS3. NS3-specific CD4 T-cell clones from patients with self-limited infection predominantly produced interferon-gamma and may thus support cytotoxic effector mechanisms important for viral clearance.