ArticlePDF Available

Extralymphatic virus sanctuaries as a consequence of potent T-cell activation

Authors:

Abstract and Figures

T helper cells can support the functions of CD8(+) T cells against persistently infecting viruses such as murine lymphocytic choriomeningitis virus (LCMV), cytomegalovirus, hepatitis C virus and HIV. These viruses often resist complete elimination and remain detectable at sanctuary sites, such as the kidneys and other extralymphatic organs. The mechanisms underlying this persistence are not well understood. Here we show that mice with potent virus-specific T-cell responses have reduced levels and delayed formation of neutralizing antibodies, and these mice fail to clear LCMV from extralymphatic epithelia. Transfer of virus-specific B cells but not virus-specific T cells augmented virus clearance from persistent sites. Virus elimination from the kidneys was associated with the formation of IgG deposits in the interstitial space, presumably from kidney-infiltrating B cells. CD8(+) T cells in the kidneys of mice that did not clear virus from this site were activated but showed evidence of exhaustion. Thus, we conclude that in this model of infection, site-specific virus persistence develops as a consequence of potent immune activation coupled with reductions in virus-specific neutralizing antibodies. Our results suggest that sanctuary-site formation depends both on organ anatomy and on the induction of different adaptive immune effector mechanisms. Boosting T-cell responses alone may not reduce virus persistence.
Content may be subject to copyright.
Extralymphatic virus sanctuaries as a consequence of
potent T-cell activation
Mike Recher
1,10
, Karl S Lang
1,10
, Alexander Navarini
1,10
, Lukas Hunziker
1,2,10
, Philipp A Lang
1
,
Katja Fink
1
, Stefan Freigang
1
, Panco Georgiev
3
, Lars Hangartner
1
, Raphael Zellweger
1
, Andreas Bergthaler
1
,
Ahmed N Hegazy
1,4
, Bruno Eschli
1
, Alexandre Theocharides
5
, Lukas T Jeker
6
, Doron Merkler
1,7
,
Bernhard Odermatt
8
, Martin Hersberger
9
, Hans Hengartner
1
& Rolf M Zinkernagel
1
T helper cells can support the functions of CD8
+
T cells against persistently infecting viruses such as murine lymphocytic
choriomeningitis virus (LCMV), cytomegalovirus, hepatitis C virus and HIV. These viruses often resist complete elimination and
remain detectable at sanctuary sites, such as the kidneys and other extralymphatic organs. The mechanisms underlying this
persistence are not well understood. Here we show that mice with potent virus-specific T-cell responses have reduced levels and
delayed formation of neutralizing antibodies, and these mice fail to clear LCMV from extralymphatic epithelia. Transfer of virus-
specific B cells but not virus-specific T cells augmented virus clearance from persistent sites. Virus elimination from the kidneys
was associated with the formation of IgG deposits in the interstitial space, presumably from kidney-infiltrating B cells. CD8
+
T cells
in the kidneys of mice that did not clear virus from this site were activated but showed evidence of exhaustion. Thus, we conclude
that in this model of infection, site-specific virus persistence develops as a consequence of potent immune activation coupled with
reductions in virus-specific neutralizing antibodies. Our results suggest that sanctuary-site formation depends both on organ
anatomy and on the induction of different adaptive immune effector mechanisms. Boosting T-cell responses alone may not reduce
virus persistence.
Some viruses are prone to developing persistent infections despite the
presence of virus-specific immune responses
1
. Identified mechanisms
of persistence include mutational escape from T-cell responses, resis-
tance to interferons, expression of cytokine decoy receptors, inhibition
of the cellular antigen-presentation machinery, glycan shielding of
surface glycoproteins and integration into the host’s genome
2–5
. Virus
sanctuaries—persistence of virus in extralymphatic tissues—not only
complicate virus elimination, they also enable reactivation during
immune suppression (for example, after organ transplantation)
6
.
Virus persistence in certain tissues is partly restricted by the expression
of specific receptors on tissue target cells; however, classical lympho-
tropic and/or hepatotropic viruses tend to persist in extralymphatic
organs such as the kidney, brain, lungs, testes and salivary glands
7–12
.
Persistence in these organs cannot always be explained by selective
receptor expression; it also cannot be due simply to continuous virus
spillover from the blood, as peripheral virus isolates differ phylo-
genetically from blood virus isolates and extralymphatic virus has been
detected in individuals without viremia
13
.
Virus persistence and distribution are also shaped by innate resis-
tance mechanisms
14,15
and drug therapy
16
. T helper cells are known to
support CD8
+
T-cell responses that destroy cells infected with persist-
ing viruses such as HIV, hepatitis C virus (HCV), cytomegalovirus
(CMV) and LCMV
17–19
. However, the outcome of chronic HIV,
HCV or CMV infections cannot always be predicted from T-cell
responses
20–22
. Paradoxically, potent immune activation is a risk factor
for end-stage disease in HIV
23
and simian immunodeficiency (SIV)
infections
24
and has been associated with disease severity in cases of
human influenza virus and severe acute respiratory syndrome (SARS)-
coronavirus infection
25,26
. Experimental immunization with T helper
epitopes has even been found to worsen the outcome of SIV infection
in rhesus macaques
27
. We and others have recently described T helper
cell–driven suppression of neutralizing antibody formation after
LCMV infection
28,29
. These findings together indicate that potent
immune activation, which is usually beneficial, can sometimes be
disadvantageous. To help delineate the mechanisms that influence this
equilibrium, we have investigated the roles of specific T cells and
Received 13 August; accepted 25 September; published online 4 November 2007; doi:10.1038/nm1670
1
Institute for Experimental Immunology, University Hospital Zu
¨
rich, Schmelzbergstrasse 12, CH-8091 Zu
¨
rich, Switzerland.
2
Department for Internal Medicine,
University Hospital Basel, 4031 Basel, Switzerland.
3
Department of Visceral and Transplantation Surgery, University Hospital Zu
¨
rich, Ra
¨
mistrasse 100, 8091 Zu
¨
rich,
Switzerland.
4
German Rheumatology Research Center, Charite
´
Platz 1, D-10117 Berlin, Germany.
5
Experimental Hematology, Department of Research, Basel
University Hospital, 4031 Basel, Switzerland.
6
Pediatric Immunology, Center for Biomedicine, University of Basel and University Children’s Hospital of Basel,
Mattenstrasse 28, 4058 Basel, Switzerland and Transplantation Immunology and Nephrology, University Hospital Basel, 4031 Basel, Switzerland.
7
Department of
Neuropathology, Georg August University, Goettingen, Germany.
8
Department of Pathology, University Hospital, Schmelzbergstrasse 12, CH-8091 Zu
¨
rich, Switzerland.
9
Institute of Clinical Chemistry, University Hospital Zu
¨
rich, Ra
¨
mistrasse 100, CH-8091 Zu
¨
rich, Switzerland.
10
These authors contributed equally to this work.
Correspondence should be addressed to M.R. (rechermike@bluewin.ch).
1316 VOLUME 13
[
NUMBER 11
[
NOVEMBER 2007 NATURE MED ICI NE
ARTICLES
© 2007 Nature Publishing Group http://www.nature.com/naturemedicine
protective antibodies in enhancing or preventing the formation of
extralymphatic persisting virus sanctuaries.
RESULTS
Induction and function of CD8
+
T cells depends on T helper cells
C57BL/6 mice infected with low-dose (200 plaque-forming units
(PFU)) LCMV (WE strain), rapidly cleared the virus from all organs
tested within 10 d (Fig. 1a). Infection of extralymphatic organs was
absent or low, even at the peak of virus replication. To allow the virus
to spread widely in the mouse before the onset of the CD8
+
T cell–
derived immune response, we transiently depleted CD8
+
T cells by
administering monoclonal antibodies 1 or 2 d before infection (that is,
on day –2 or day –1) with LCMV-WE (standard protocol, used
throughout this manuscript). Ten days after infection, mice that had
been depleted of CD8
+
T cells showed virus replication in all organs
tested (Fig. 1a). After their initial depletion, CD8
+
T cells reemerged
in the blood around day 10 after infection and then expanded, peaking
at day 20 after infection as assessed by fluorescence-activated cell
sorting (FACS) analysis of peripheral blood (Fig. 1b). This expansion
of CD8
+
T cells was almost completely virus specific, as up to 80% of
these CD8
+
T cells were tetramer-specific for known immunodomi-
nant virus epitopes (GP33-41, GP276-284 and NP396-404) (Fig. 1c;
for representative FACS plot, see Supplementary Fig. 1a online). The
CD8
+
T cells were mostly of a CD62L
lo
phenotype, indicating
activation (Fig. 1d). Most CD8
+
T cells expressed granzyme B, and
a small fraction produced interferon-g (IFN-g) directly ex vivo with-
out further restimulation (Fig. 1e; for representative FACS plots see
Supplementary Fig. 1b). To determine the importance of T helper
cells in CD8
+
T-cell priming and proliferation in our model, we
infected Cd4
–/–
mice with 200 PFU LCMV-WE using the standard
protocol described above. Expansion of CD8
+
T cells in peripheral
blood, as detected by FACS, was suppressed in Cd4
–/–
mice (Fig. 1b).
CD8
+
T cells were less activated, as downregulation of the selectin
CD62L was impaired (Fig. 1d). Granzyme B–expressing CD8
+
T cells
were also relatively reduced in the absence of T helper cells (Fig. 1e
and Supplementary Fig. 1b). By contrast, adoptive transfer of naive,
virus-specific T-cell receptor transgenic CD4
+
T helper cells (Smarta
cells
30
) before LCMV infection of C57BL/6 mice (C57BL/6+Smarta)
enhanced CD8
+
T-cell activation. CD62L downregulation and direct
ex vivo granzyme B expression were both greater than in control
C57BL/6 mice (Fig. 1d,e). Transfer of Smarta T helper cells augmented
formation of IFN-g by T helper cells (Supplementary Fig. 1c). At the
same time that CD8
+
T cells peaked in mouse blood samples, the mice
lost weight (Fig. 1f) and developed hepatitis, as indicated by elevated
serum bilirubin concentrations and augmented liver transaminase
activity in serum (Fig. 1g,h). Both weight loss and hepatitis correlated
markedly with the number of available T helper cells (Fig. 1fh).
Thus, CD8
+
T-cell expansion and expression of effector functions
(granzyme B and IFNg) correlated with available T-cell help.
Delayed virus clearance in mice with potent T-cell responses
As expected, Cd4
–/–
mice infected with LCMV showed high viral titers
in blood at all time points measured (Fig. 2a). However, virus
elimination from the blood was, paradoxically, delayed in mice with
high T–helper cell responses (C57BL/6+Smarta) compared with con-
trol C57BL/6 mice (Fig. 2a). The impaired virus clearance in mice
7
6
5
4
<3
80
60
40
20
0
Virus organ titer
(log
10
ml
–1
)
CD8
+
cells
(% of blood lymphocytes)
CD8
+
Tetramer
+
cells
(% of CD8
+
cells)
CD62L
lo
CD8
+
cells
(% of CD8
+
cells)
3
Spleen Liver Kidney Lung Brain
20
15
Anti-CD8
Time after infection (d)
10
5
0
120
110
100
90
80
70
01020
Time after infection (d)
30 40
0 102030
60
80
60
40
20
0
40
20
6
1,500
1,000
500
0
5
4
3
2
1
<0.1
GP276 GP33 NP 396
C57BL/6+Smarta
C57BL/6
Cd4
–/–
C57BL/6+Smarta
C57BL/6
Cd4
–/–
0
CD8
+
T-cell function
(% of CD8
+
cells)
Weight
(% of starting weight)
C57BL/6+Smarta
C57BL/6
Cd4
/–
Serum bilirubin
(mg dl
–1
)
Serum ALT
(units per L)
C57BL/6+Smarta
C57BL/6
Cd4
–/–
abcd
hgfe
Figure 1 T helper cell–dependent CD8
+
T-cell response against distributed virus. (a) C57BL/6 mice were depleted of CD8
+
T cells at days –2 and –1 ()
or left untreated (&) before infection with 200 PFU LCMV-WE. Organ viral titers were measured 10 d later (n ¼ 3). (b) C57BL/6 mice (), C57BL/6 mice
supplemented with 10
7
Smarta splenocytes on day –1 before infection (C57BL/6+Smarta, &)orCd4
–/–
mice () were all transiently CD8
+
T-cell depleted
with monoclonal antibodies administered on days –2 and –1 before infection, then infected with 200 PFU LCMV-WE on day 0 or left uninfected (E).
CD8
+
T cells in peripheral blood were monitored at the indicated time points by FACS analysis (n ¼ 5–10). (c) Twenty days after infection of C57BL/6
mice as described above, reemerging CD8
+
T cells were tested for LCMV specificity using tetramer staining for three immunodominant epitopes
(GP33-41, GP276-284 and NP396-404) and FACS analysis (n ¼ 4–9). (d,e) CD62L expression (d) or direct ex vivo granzyme B (black) and IFN-g (gray)
expression (e) of CD8
+
T cells were measured by FACS 15 d (d)or20d(e) after infection (n ¼ 5). (fh) C57BL/6 mice (), C57BL/6+Smarta mice (&)
or Cd4
–/–
mice () were infected with LCMV according to the standard protocol (n ¼ 4–5). Mouse body weight was measured at the indicated time points
(f). (g,h) 20 d after LCMV infection, serum bilirubin concentrations and serum amino liver transaminase (ALT) activity were measured (n ¼ 5).
ARTICLES
NATURE MEDIC INE VOLUME 13
[
NUMBER 11
[
NOVEMBER 2007 1317
© 2007 Nature Publishing Group http://www.nature.com/naturemedicine
with potent T-cell responses was mostly due to a failure to clear the
virus from the kidneys and lungs (Fig. 2bf). Compared to Rag1
–/–
mice, which are deficient in T cells and B cells, virus clearance was
1,000–10,000 times greater in the spleen and liver of C57BL/6+Smarta
mice, although no significant difference was measured in the kidneys
and lungs (Fig. 2bf). The difficulty in eliminating the virus from the
kidneys was still observed 50 d after infection of C57BL/6+Smarta
mice, whereas virus control in the lungs and brain was comparable to
that of control C57BL/6 mice at this time point (Supplementary
Fig. 2a,b online). Notably, no virus could be detected in the blood at
this time point (Fig. 2a). The observed site-specific virus elimination
in C57BL/6+Smarta mice was not due to site-specific T helper cell
responses, as T helper cell activation and function were comparable in
the liver, kidneys and lungs (Supplementary Fig. 2c).
In summary, potent T-cell responses were apparently effective in
lowering viral burden in some organs but did not clear virus from
other tissues.
Virus persistence correlates with delayed antibody formation
LCMV-infected C57BL/6 mice developed measurable neutralizing
antibodies by 30 d after infection (Fig. 3a). In keeping with our earlier
results
28
, the formation of neutralizing antibodies was, paradoxically,
delayed and reduced in mice supplemented with Smarta T helper cells,
to a similar extent as in mice lacking CD4
+
T cells (Fig. 3a). To test
whether the delayed antibody formation was responsible for the
observed site-specific virus persistence, we analyzed virus elimination
in mice deficient in antibody formation. Aicda
–/–
mice lack activation-
induced cytidine deaminase (AID) and therefore do not undergo
7
6
5
4
3
7
P = 0.001
P < 0.001
P = 0.07
P = 0.07 P > 0.1
P = 0.062
P = 0.005 P < 0.001
P = 0.012
6
5
4
3
<3
<3
<3
2
0102030
Time after infection (d)
C57BL/6
C57BL/6
Rag1
/–
Rag1
–/–
C57BL/6+Smar
ta
C57BL/6+Smar
ta
C57BL/6
Rag1
/–
C57BL/6+Smar
ta
C57BL/6
Rag1
/
C57BL/6+Smar
ta
C57BL/6
Rag1
–/–
C57BL/6+Smar
ta
40 50
<1.7
Blood viral titer
(log
10
PFU ml
–1
)
Liver viral titer
(log
10
PFU)
Brain viral titer
(log
10
PFU)
Spleen viral titer
(log
10
PFU)
7
6
5
4
3
7
6
5
4
3
<3
7
6
5
4
3
<3
7
6
5
4
3
Kidney viral titer
(log
10
PFU)
Lung viral titer
(log
10
PFU)
P < 0.001
P < 0.001
P < 0.001 P < 0.001P > 0.1 P > 0.1
abcd
fe
Figure 2 Organ-specific viral persistence enhanced by potent
cellular immune responses. (a) Viral blood titers of C57BL/6
mice (), C57BL/6 mice supplemented with Smarta T helper
cells (C57BL/6+Smarta, &)orCd4
–/–
mice infected with LCMV
according to the standard protocol (
) were analyzed at the
indicated time points (n ¼ 3–5). (bf) Twenty days after LCMV
infection using the standard protocol, viral titers in the spleen
(b), liver (c), brain (d), kidneys (e)andlungs(f) were measured
in individual Rag1
–/–
mice, C57BL/6 mice and C57BL/6+Smarta
mice as indicated (individual values are shown).
5
7
P = 0.015
P = 0.015
P < 0.001
P < 0.001
P = 0.001
P = 0.03
P = 0.02
P = 0.06
6
5
4
3
<3
0 10203040
Time after infection (d)
50 60 Spleen Liver Brain Kidney Lung
4
3
2
1
<1
Neutralizing titer
(–(log
2
) × 10)
Viral organ titer
(log
10
PFU)
Viral organ titer
(log
10
PFU)
LCMV-Arm organ titer
(log
10
PFU)
Spleen
NS
Liver Brain Kidney Lung Spleen Liver Kidney Lung
Spleen Liver Brain Kidney Lung
7
6
5
4
3
<3
7
6
5
4
3
<3
P = 0.030
P = 0.003
P > 0.1
P = 0.014
7
6
5
4
3
<3
Viral organ titer
(log
10
PFU)
ab c d
e
Figure 3 Site-specific viral persistence as a consequence of delayed antibody responses. (a) C57BL/6 mice
(), C57BL/6 mice supplemented with transgenic virus–specific CD4
+
T cells (C57BL/6+Smarta, &), Cd4
–/–
mice (), Aicda
–/–
mice (E) and VI10Yen mice (B) were depleted of CD8
+
T cells on days –2 and –1, then
infected with 200 PFU LCMV-WE on day 0. (a) At the indicated time points, neutralizing antibody formation
was assessed by neutralization assay (n ¼ 4–9). (be) Aicda
–/–
mice were transiently depleted of CD8
+
T
cells on days –2 and –1, then infected with 200 PFU LCMV-WE (b) or LCMV-Armstrong (LCMV-Arm) (d)on
day 0. LCMV-WE–infected Rag1
–/–
mice were analyzed as a control (c). Alternatively, Aicda
–/–
mice that had
not been transiently depleted of CD8
+
T-cells were infected with 2 10
6
PFU LCMV-WE (e). Viral organ
titers were measured by plaque assay 30 d (b)or50d(c,d,e) after infection (individual values are shown).
ARTICLES
1318 VOLUME 13
[
NUMBER 11
[
NOVEMBER 2007 NATURE MED ICI NE
© 2007 Nature Publishing Group http://www.nature.com/naturemedicine
immunoglobulin class switch recombination and affinity maturation,
whereas other B-cell functions, including IgM responses, remain
effective
31
. LCMV-infected Aicda
–/–
mice did not produce neutralizing
antibodies, as shown by a neutralization assay (Fig. 3a), and also did
not produce LCMV glycoprotein–specific IgG responses, as tested by
ELISAs (Supplementary Fig. 4b,c online). Notably, there were only
slight differences in numbers of virus-specific
CD8
+
T cells, distribution of epitopes, pro-
duction of IFN-g, and CD8
+
T-cell cytotoxi-
city between Aicda
–/–
mice and C57BL/6 mice
(Supplementary Fig. 3ae online). Like
C57Bl/6 mice supplemented with Smarta
cells, the LCMV-infected Aicda
–/–
mice
showed site-specific virus persistence, as
indicated by significantly lower viral titers
in the spleen and liver 30 d after infection
when compared to the kidneys, lungs and
brain (Fig. 3b). In contrast, RAG-deficient
mice had evenly distributed high viral titers
in all organs tested (Fig. 3c). Site-specific
persistence was also found in Aicda
–/–
mice
that were infected with another LCMV strain,
LCMV-Armstrong (Fig. 3d), and in Aicda
–/–
mice infected with high doses of LCMV-WE
without initial transient CD8
+
T-cell
depletion (Fig. 3e).
Reduction of virus sanctuaries by
virus-specific B cells
To analyze whether supplemented B cells
were more effective than T cells at reducing
viral titers, we adoptively transferred either
LCMV-specific CD8
+
T cells or neutralizing
LCMV-specific B cells (see Methods) 30 d
after LCMV infection of Aicda
–/–
mice (for
blood virus kinetics, see Supplementary
Fig. 4a). Viral organ titers were measured
20 d later (see experimental schedule,
Fig. 4a). Fifty days after LCMV infection, Aicda
/
mice without cell
transfer showed the lowest viral titers in the liver and brain but had
high titers in the spleen, lungs and kidneys (Fig. 4bf).Theincreasein
spleen viral titers at day 50 compared to day 30 is probably due to
CD8
+
T-cell exhaustion, which has been described to occur more
rapidly in lymphoid tissue than in other organs
32
. Control C57BL/6
Anti-CD8 LCMV Analysis
Spleen virus titer
log
10
PFU
Liver viral titer
(log
10
PFU)
Kidney viral titer
(log
10
PFU)
Brain viral titer
(log
10
PFU)
Time (d)
50
7
P < 0.001
P < 0.01
P < 0.05
P < 0.05
6
5
4
3
Without
+T cells
+B cells
VI10Yen
C57BL/6
<3
300–2,–1
Tra ns fe r
(B cells versus T cells)
7
6
5
4
3
<3
Without
+T cells
+B cells
VI10Yen
C57BL/6
P < 0.001
P < 0.01
P > 0.1
P > 0.1
7
6
5
4
3
<3
Without
+T cells
+B cells
VI10Yen
C57BL/6
P < 0.01
P > 0.1
P > 0.1
P > 0.1
P < 0.001
P < 0.01
P < 0.05
P > 0.05
7
6
5
4
3
<3
Without
+T cells
+B cells
VI10Yen
C57BL/6
Lung viral titer
(log
10
PFU)
7
6
5
4
3
<3
Without
+T cells
+B cells
VI10Yen
C57BL/6
P < 0.001
P < 0.05
P > 0.05
P > 0.1
abc
ef
d
Figure 4 Reduction of extralymphatic virus sanctuaries associated with LCMV
glycoprotein-specific B-cell responses. Aicda
–/–
mice were transiently depleted of
CD8
+
T cells on days –2 and –1, then infected with 200 PFU LCMV-WE on day 0.
Thirty days after infection, 1 10
7
LCMV-specific neutralizing B cells (+B cells)
or LCMV-specific CD8
+
T cells (+T cells) were adoptively transferred as described
in the Methods. As controls, Aicda
–/–
mice (without), VI10Yen mice (VI10Yen) or
C57BL/6 mice (C57BL/6) were infected with LCMV according to the standard
protocol but did not receive further adoptive cell transfer 30 d after infection. Viral
organ titers were measured 50 d after LCMV infection. (a) Experimental setup.
(bf) Viral organ titers measured 50 d after LCMV infection in spleen (b), liver
(c), brain (d), kidney (e)andlung(f). Individual values are shown.
20
100
175
150
125
100
75
50
25
0
Liver
Kidney
Lung
75
50
25
PD-1
hi
CD69
hi
CD62L
lo
0
15
10
5
0
60
50
40
30
20
10
0
GP33 GP276 NP396
Tetramer
+
CD8
+
T cells
(% of total CD8
+
cells)
Specific lysis in spleen (%)
Specific lysis in liver (%)
Specific lysis in kidney (%)
Activated CD8
+
cells
(% of CD8
+
cells)
60
50
40
30
20
10
0
60
50
40
30
20
10
Threefold dilutions of effector cells
(starting at effector/target ratio of 100:1)
0
60
50
40
30
20
10
0
60
50
40
30
20
10
0
60
50
40
30
20
10
0
a
d
bc
Granzyme B expression
(MFI of CD8
+
cells)
Figure 5 T cells at sanctuary sites are activated but lack direct cytotoxicity. (a) Fifty days after infection
using the standard protocol, virus was isolated from Aicda
–/–
mouse kidneys. We then injected 200 PFU
of each of two isolates (&,
) from two mice into separate naive C57BL/6 mice. As a control, we used
200 PFU of wild-type LCMV-WE (). Ten days later, GP33-41, GP276-284 and NP396-404 tetramer-
positive CD8
+
T cells were measured by FACS analysis (n ¼ 5). (b,c) Aicda
–/–
mice were infected with
LCMV according to the standard protocol. Thirty days after infection, CD8
+
T cells isolated from the
liver (black), kidneys (dark gray) or lungs (light gray) were assessed for expression of the indicated
activation markers (b) and for granzyme B expression (c)byFACSanalysis(n ¼ 3–4). (d) Aicda
–/–
mice
were infected with LCMV according to the standard protocol. Thirty days after infection, CD8
+
Tcells
isolated from the indicated organs were analyzed for cytotoxicity in a chromium-release assay. Target
cells were labeled with GP33-41 peptide () or unlabeled (&). Each line represents the mean and
s.e.m. for one animal (chromium release measured in duplicates; n ¼ 5–7 experiments).
ARTICLES
NATURE MEDIC INE VOLUME 13
[
NUMBER 11
[
NOVEMBER 2007 1319
© 2007 Nature Publishing Group http://www.nature.com/naturemedicine
mice had almost completely cleared virus from all organs at this time
point (Fig. 4bf). LCMV-specific adoptive B-cell transfer into Aicda
/
mice was successful, as indicated by the rapid formation of LCMV
glycoprotein–specific IgG (Supplementary Fig. 4b), and was asso-
ciated with significantly reduced viral titers in the kidneys and a trend
toward lower titers in the lungs (Fig. 4e,f). By contrast, T-cell transfer
did not significantly reduce viral titers in the kidneys or lungs
(Fig. 4e,f). Transfer of LCMV glycoprotein–specific neutralizing anti-
bodies into Aicda
/
mice did not lower viral titers in the kidneys or
lungs and did not induce measurable glycoprotein-specific IgG
responses, implying that the antibodies were rapidly consumed by
circulating virus particles (data not shown). We also analyzed LCMV-
infected VI10Yen mice. In these mice, most naive B cells are specific
for vesicular stomatitis virus (VSV)
33
. These mice did not produce
LCMV-neutralizing antibodies for up to 50 d after infection (Fig. 3a);
however, in contrast to AID-deficient mice, they eventually formed
LCMV glycoprotein–specific IgG antibodies (Supplementary Fig. 4c).
CD8
+
T-cell function in infected VI10Yen mice was comparable
to that in C57BL/6 mice (data not shown). Compared to those
in AID-deficient mice, viral titers in the kidneys were significantly
lower in VI10Yen mice, mimicking the result of virus-specific B-cell
transfer (Fig. 4bf).
Together, these results show that the reduction of virus replication
in some organs, such as the kidneys and lungs, correlates with the
amount of LCMV glycoprotein–specific IgG antibodies but not with
T-cell responses. Notably, B-cell transfer into Aicda
–/–
mice did not
augment CD8
+
T-cell activation and function in extralymphatic
organs (Supplementary Fig. 4df).
ActivatedbutexhaustedCD8
+
T cells in viral sanctuaries
We next investigated the mechanisms that are involved in the
formation of site-specific viral sanctuaries. First, we analyzed whether
the persisting extralymphatic virus population consisted of
CD8
+
T-cell escape variants and thus resisted T-cell attack. Fifty
days after infection using the standard protocol, we isolated the
virus from the kidneys of two Aicda
–/–
mice. We then injected 200
PFU of each kidney isolate or 200 PFU of wild-type LCMV-WE
separately into naive C57BL/6 mice. Ten days later, we measured the
expansion of LCMV GP33-41–specific, GP276-284–specific and
NP396-404–specific CD8
+
T cells by tetramer staining, nding only
minimal differences (Fig. 5a). Lack of mutation was further confirmed
by sequencing of one representative kidney isolate, which revealed
LCMV-WE wild-type codon usage within and near the immuno-
dominant GP276-284 epitope (data not shown).
Next, we compared the livers, kidneys and lungs of AID-deficient
mice and found that CD8
+
T-cell activation and granzyme B expres-
sion were similar in these organs (Fig. 5b,c). Numbers of virus-specific
CD8
+
T cells and levels of IFN-g were also similar in the kidneys, lungs
and liver (Supplementary Fig. 3b,d), suggesting that differences in
CD8
+
T-cell activation and function could not explain the site-specific
virus persistence observed in Aicda
–/–
mice.
In contrast, CD8
+
T-cell cytotoxicity was lower in the kidneys
than in the liver and spleen (Fig. 5d). Direct cytotoxicity correlated
well with the measured viral titers in kidney versus liver and spleen at
the same time point (Fig. 3). Thus, although we could not demon-
strate differences in the intrinsic activation and function of CD8
+
T cells, the kidney CD8
+
T-cell population as a whole showed low
cytotoxicity, which is indicative of exhaustion and associated with
high viral kidney titers.
Virus persistence at epithelial sites
We performed immunohistological analysis of kidneys 50 d after
infection of mice. In Rag1
–/–
mice, which lack B and T cells, the
virus was mainly located in the kidney tubule epithelium (Fig. 6a, i).
Rag1
–/–
VL4
a
bc
i ii iii iv v vi
vii viii ix x xi xii
xiii xiv xv xvi xvii xviii
xix
xx xxi xxii xxiii xxiv
lgG
CD8
CD4
B-cell transfer
lgG
CD138
VL4
Antibody transfer No transfer
i
ii
iii
VL4
VL4
VL4
RAG
AID
Aicda
–/–
low dose
Aicda
–/–
high dose
C57BL/6
+Smarta
C57BL /6
VI10Yen
without anti-CD8
Figure 6 Viral persistence at epithelial sites.
(a) Fifty days after LCMV infection, Rag1
–/–
mice,
Aicda
–/–
mice with or without transient CD8
+
T-cell depletion, C57BL/6 mice, Smarta-
supplemented C57BL/6 mice (C57BL/6+Smarta)
or VI10Yen mice were infected with LCMV using
the standard protocol, immunohistology was used
to detect LCMV nucleoprotein (VL4, ivi), IgG
(viixii) CD8 (xiiixviii) and CD4 (xixxxiv)in
kidney sections (n ¼ 2–5). (b) Aicda
–/–
mice
were LCMV infected with LCMV according to the
standard protocol. After 30 d, neutralizing
antibodies (monoclonal KL25 antibodies or
polyclonal neutralizing serum) or LCMV-specific
B cells were adoptively transferred. Twenty days
later, expression of IgG, LCMV antigen (VL4)
and plasma cells (CD138) was analyzed by
immunohistology as indicated. (c, i)C57BL/
6+Smarta mice were infected with LCMV
according to the standard protocol. Fifty days
later, immunohistological staining of LCMV-
nucleoprotein (VL4 antibody) was performed
on sections of the distal urinary tract (one
representative section is shown). (c, ii,iii) Aicda
–/–
mice were transiently depleted of CD8
+
T cells
on days –2 and –1, then infected with 200 PFU
LCMV-WE on day 0. As a control, Rag1
–/–
mice
were infected with LCMV on day 0. Fifty days
after infection, immunohistological staining
of LCMV-nucleoprotein (VL4 antibody) was
performed on liver sections (ii) or lung sections
(iii). Scale bars, 200 mm.
ARTICLES
1320 VOLUME 13
[
NUMBER 11
[
NOVEMBER 2007 NATURE MED ICI NE
© 2007 Nature Publishing Group http://www.nature.com/naturemedicine
Similar patterns of staining around the kidney tubules were observed
in AID-deficient mice, either with or without transient CD8
+
T-cell
depletion, and in mice transfused with Smarta T cells (Fig. 6a, ii,iii).
Hence, despite a T-cell response, viral distribution in the kidneys
remained the same as in Rag1
–/–
mice. By contrast, C57BL/6 mice had
already efficiently eliminated the virus from their kidneys 50 d after
infection (Fig. 6a, v). Extensive infiltrations of CD8
+
T cells (Fig. 6a,
xiiixviii) and CD4
+
T cells (Fig. 6a, xixxxiv) were observed in the
kidneys of all mice tested (except T cell–deficient Rag1
–/–
mice)
independently of virus distribution, confirming that insufficient
T-cell recruitment was not the reason for the epithelial persistence
of virus antigen in Aicda
–/–
mice and mice supplemented with Smarta
T helper cells.
To further analyze virus elimination, we carried out immunohisto-
logical analysis of kidneys from VI10Yen mice, which had low but still
detectable viral titers 50 d after infection (Fig. 4). In these mice, the
virus was found mainly in the kidney interstitial space, rather than in
epithelial cells. (Fig. 6a, vi). IgG staining in these mice localized to the
same interstitial sites, supporting the idea that virus-antibody immune
complexes form there (Fig. 6a, xii). Interstitial IgG deposition was also
found in C57BL/6 mice, implying that interstitial IgG deposition
remained after virus elimination (Fig. 6a, xi). As expected, IgG
staining was not detectable in infected Rag1
–/–
mice and Aicda
–/–
mice (Fig. 6a, viiix). When LCMV-neutralizing IgG was injected into
infected Aicda
–/–
mice IgG deposits were found mostly in kidney
glomeruli, but usually not in the interstitial space surrounding the
LCMV-infected epithelia (Fig. 6b), and LCMV kidney titers remained
unchanged (data not shown). Thus, virus elimination from kidneys
correlated with the formation of interstitial IgG deposits, presumably
produced by kidney-infiltrating B cells. Plasma cells could easily be
found within kidneys in immunohistological analyses (Fig. 6b).
Kidney plasma cells were also observed in Aicda
–/–
mice and
C57BL/6 mice, whereas IgG deposition between kidney tubules was
absent in Aicda
–/–
mice (Fig. 6b).
Virus was also detected in the lower urinary tract, where the
epithelium is built of multiple cell layers. There, the virus was detected
mainly in the upper cell layer (Fig. 6c), consistent with virus
propagation in the urinary tract tubular system. Histological analysis
of lung sections revealed similar virus sanctuaries in the bronchiolar
epithelium in Rag1
–/–
mice and Aicda
–/–
mice, indicating that lung
epithelia might show resistance to T-cell attack similar to that seen in
kidney epithelia (Fig. 6c). By contrast, in Aicda
–/–
mice, which had low
liver viral titers, the virus was found mainly in the Kupffer cell
compartment of the liver, whereas virus was detected in liver hepa-
tocytes in Rag1
–/–
mice (Fig. 3b and Fig. 6c).
DISCUSSION
Our experimental data mimic the paradoxical clinical finding that
potent immune activation is sometimes a risk factor for viral persis-
tence and severe disease. CD8
+
T cells, supported by helper T cells,
efficiently cleared the virus from some organs but not other organs.
Virus was particularly persistent in the kidneys and lungs, but more
efficiently cleared from the liver and spleen. We speculate that site-
specific viral persistence develops as a consequence of overactivated and
exhausted T-cell responses combined with slow induction of virus-
specific neutralizing antibodies. Such a combination is typical for
several chronic human virus infections
34
. B cell–deficient mice with
primed T cells specific for g-herpesvirus do not eliminate the virus
from the lungs, but do eliminate it from all other organs, upon rein-
fection
35
. In addition, X-linked lymphoproliferative disease is similarly
characterized by an overreactive cellular immune response combined
with slow induction and low levels of virus-specific antibodies; this
combination might lead to fatal outcomes of Epstein-Barr virus (EBV)
infection in humans and of LCMV infection in mice
36
.Inourmodelof
infection by LCMV, the CD8
+
T-cell response against distributed viral
antigen was strengthened by the help of CD4
+
T cells, as shown pre-
viously
18
. Similar findings exist for SIV and other persisting viruses
17,19
.
We found that the presence of virus-specific IgG responses was
associated with virus clearance and with changes in virus distribution
from epithelial cells to the interstitial space. Other models of persistent
virus infection have similarly proposed that B-cell responses are
associated with virus elimination
37,38
. Histological analysis showed
that T cells efficiently infiltrated virus-infected epithelia, in a pattern
reminiscent of autoreactivity’ (for example, interstitial nephritis,
bronchiolitis or Sjo
¨
gren-like disease, which might convert into overt
autoimmune disease). Although we did not detect CTL-escape var-
iants at extralymphatic sites up to 50 d after infection, the observed
colocalization of CD8
+
T cells with the virus might eventually create
sites conducive to the formation of CTL-escape mutants
39
. Lympho-
mas associated with chronic viral infection, which often arise in
extralymphatic tissues, could be a consequence of ongoing extralym-
phatic immune responses. Together, our ndings might explain why
chronic virus infections, lymphomas and autoimmunity have been
described as forming a triple association
40
.
In conclusion, the successful and complete elimination of a dis-
tributed persistence-prone viral infection requires efficient interplay
between cellular and neutralizing humoral responses. Whereas T-cell
responses can effectively clear the virus from both lymphatic and more
‘solid extralymphatic organs such as the liver, IgG responses are
especially needed in organs with tubular epithelial microanatomy,
such as the kidneys and lungs. The relative contributions of these two
adaptive arms of the immune system also regulate immunopatho-
logical disease mediated by chronic inflammation. Thus, boosting of
T-cell responses alone might not reduce virus persistence
41
and might
eventually induce immunopathology or autoimmunity
36
.Ourresults
indicate that virus replication, tropism and sanctuary formation
depend on the specific virus and on organ anatomy and physiology,
but also on the induction, kinetics and efficacy of different adaptive
immune effector mechanisms.
METHODS
Mice. All mice were bred and maintained under specific pathogen–free
conditions, and experiments were performed in accordance with institutional
and Swiss national guidelines. Animal care protocols were approved by the
animal experiment committe of Zu
¨
rich, Switzerland.
Cd4
–/–
, VI10Yen–B-cell transgenic, TgH(KL25)–B-cell transgenic, 318–T-cell
transgenic, Smarta1–T-cell transgenic, Rag1
–/–
and Aicda
–/–
mice all have a
C57BL/6 genetic background. All mice were purchased from the Institute for
Labortierkunde, Faculty of Veterinary Medicine, University Zu
¨
rich-Irchel.
Virus. LCMV strains WE and Armstrong (Arm) were originally gifts
(see Acknowledgments) and were propagated on L929 or BHK21 cells.
Mice were infected with 2 10
2
or 2 10
6
PFU of LCMV injected
intravenously as indicated.
Detection of viral and neutralizing antibody titers. LCMV viral titers were
detected by a plaque-forming assay on MC57 fibroblasts as described
42
.
Detection of neutralizing activity against LCMV in mouse sera has also been
described
42
. Briefly, neutralizing activity against LCMV-WE was measured in a
plaque-reduction assay. The neutralizing titer was defined as the log
2
dilution
that caused a half-maximal reduction in plaques, with the reduction being
measured in comparison to the number of plaques formed by similar amounts
of the virus incubated with control sera from uninfected mice or medium
alone. A titer of o1 indicates no detectable neutralization at an initial serum
predilution of 1:10.
ARTICLES
NATURE MEDIC INE VOLUME 13
[
NUMBER 11
[
NOVEMBER 2007 1321
© 2007 Nature Publishing Group http://www.nature.com/naturemedicine
In vivo depletion of CD8
+
T cells. Cell depletions were performed using rat
monoclonal antibodies specific for CD8 (YTS 169.4) as described
28
.The
hybridomas were initially obtained from H. Waldmann. We injected 200 ml
of purified CD8
+
T cell–depleting antibodies intraperitoneally on days –2
and –1 before LCMV infection. The efficiency of depletion was always
confirmed by FACS analysis of peripheral blood cells using fluorescein
isothiocyanate (FITC)-, phycoerythrin (PE)-, or allophycocyanin (APC)-
labeled CD8-specific antibodies (Pharmingen).
Markers of liver injury. We measured serum activity of alanine aminotrans-
ferase (ALT) as a marker of hepatocellular injury and total serum bilirubin
concentrations using the serum multiple analyser (Ektachem DTSCII, Johnson
and Johnson) or the Roche Modular Analytics Analyser with pyridoxal
phosphate (Roche).
Adoptive cell transfer. Aicda
–/–
mice were transiently depleted of CD8
+
T cells
and infected with LCMV by the standard protocol, as described in Results.
After 30 d, either 1 10
7
MACS-sorted naive CD8
+
T cells from LCMV-
specific T-cell receptor–transgenic mice (318 mice) or in vivo CD8
+
Tcell
depleted naive TgHKL25 splenocytes (from LCMV-specific B cell–knock-in
mice
33
) were adoptively transferred. Immunohistology and measurement of
organ viral titers and CD8
+
T-cell function were performed 20 d later.
LCMV-neutralizing monoclonal antibodies (KL25, 1.5 mg) or polyclonal
LCMV-neutralizing hyperimmune serum (200 ml) were transfused intrave-
nously as a control 30 d after LCMV infection of Aicda
–/–
mice.
LCMV-glycoprotein GP1-specific IgG measurements. The detection of
LCMV-glycoprotein GP-1–specific IgG by ELISA has been described
28
.
Fluorescence-activated cell sorting analysis of T-cell activation. Single-cell
suspensions of the different organs were prepared. Cells were then incubated
with fluorescent antibodies specific for PD-1 (PE-labeled, eBioscience), CD69
and CD62L (FITC-labeled, both BD Pharmingen), counterstained with fluores-
cent CD8-specific antibodies (APC-labeled, BD Pharmingen) and analyzed
using a FACSCalibur system (BD Pharmingen). For CD4
+
T-cell activation,
antibodies to CD69, CD62L and CXCR3 (BD Pharmingen) were used.
For intracellular cytokine staining and T-cell restimulation, splenocytes were
incubated for 5 h with or without LCMV peptide (1 mM) or PMA (50 ng ml
–1
)
and ionomycin (500 ng ml
–1
) for 6 h in RPMI medium supplemented with
10% FBS, b-mercaptoethanol and 5 mgml
–1
brefeldin A (Sigma-Aldrich). The
cells were then harvested, washed once in PBS with 4% FBS and 12.5 mM
EDTA, and stained with PE- or APC-conjugated CD8-specific antibodies or
PE-labeled antibodies specific for CD4 (BD Pharmingen). After washing, cells
were fixed with 4% paraformaldehyde in PBS for 10 min, then permeabilized
using PBS with 4% FBS, 12.5 mM EDTA and 0.1% saponin (Sigma-Aldrich).
For intracellular cytokine staining, cells were incubated with PE-conjugated
granzyme B–specific antibody (Caltag) or APC-labeled IFN-gspecific antibody
(BD Pharmingen) for 30 min at 4 1C. After washing twice with permeabiliza-
tion buffer, we resuspended the cells in PBS with 4% FBS and 12.5 mM EDTA
and analyzed them using a FACSCalibur system (BD Pharmingen).
In vivo cytotoxicity. In vivo cytotoxicity was assayed using C57BL/6 splenocytes
incubated for 1 h with or without LCMV-derived MHC class I GP276-284
peptide and labeled for 10 min with 5 mgml
–1
carboxyfluorescein diacetate
succinimidylester from Molecular Probes (CFSE
high
, peptide-labeled spleno-
cytes) or 0.5 mgml
–1
CFSE (CFSE
low
, unlabeled splenocytes). We injected
10
7
cells of each fraction intravenously into AID-deficient or control C57BL/6
mice 50 d after LCMV infection using the standard protocol described in
Results. The number of CFSE-positive cells remaining in blood 12 h later was
determined by FACS analysis. Specific cytotoxicity was calculated from the cell
counts of LCMV-infected (memory) and noninfected (naive) mice under each
treatment, as 1 – (CFSE
high
memory
/CFSE
low
memory
)/(CFSE
high
naive
/CFSE
low
naive
),
and expressed as a percentage.
51
Cr-release cytotoxicity assay. We performed
51
Cr release assays as
described
43
. Single-cell preparations of organs were prepared and lymphocytes
separated by gradient centrifugation (Lympholyte, Cedarlane). Threefold dilu-
tions of effector cells with a starting effector/target ratio of 100:1 were analyzed.
Immunohistology. Histological analysis was performed on snap-frozen tissue
sections stained with rat monoclonal antibodies against LCMV-nucleoprotein
(VL4), CD8 (53-6.7), IgG (mixture of rat anti-mouse IgG1, IgG2a/b and IgG3)
or CD138. Monoclonal antibodies were detected with goat anti-rat (Caltag)
and alkaline phosphatase-labeled donkey anti-goat (Jackson ImmunoResearch)
secondary antibodies, which we visualized by using naphthol 6-bromo-2-
hydroxy-3-naphtholic acid 2-methoxy anilide (AS-BI) phosphate and new
fuchsin as a substrate. The enzyme reaction yielded a red reaction product.
Kidney sections were stained in some experiments with goat anti-rat secondary
antibody (Caltag), followed by donkey anti-goat labeled with horseradish
peroxidase. Incubation with 3-amino-9-ethylcarbazole (AEC) in the presence
of H
2
O
2
yielded a brown reaction product. Sections were counterstained
with hemalum.
Statistical analysis. Data are presented as mean ± s.e.m. or as single values
(median value visualized as a horizontal line). Values in treatment groups were
analyzed for significant differences using a one-way analysis of variance
followed by a Tukey least-significant difference test. For statistical analysis,
organ viral titers below detection limits were set to 10
1.5
PFU.
Note: Supplementary information is available on the Nature Medicine website.
ACKNOWLEDGMENTS
We thank A.J. MacPherson and D. Pinschewer for discussions and K. Tschannen
for technical assistance. Aicda
–/–
mice were provided by T. Honjo (Kyoto
University). LCMV strains WE and Armstrong (Arm) were originally obtained
from F. Lehmann-Grube (Heinrich Pette Institute) and M. Buchmeier (Scripps
Institute), respectively. This work was supported by Swiss National Foundation
Grants to H.H. (3100A0-100779) and R.M.Z. (3100A0-100068). K.S.L. was
partially supported by the Deutsche Forschungsgemeinschaft LA 1419/1-1. A.B.
holds a PhD fellowship of the Boehringer Ingelheim Fonds. A.N.H. is a fellow of
Graduate School 1121 of the German Science Foundation.
AUTHOR CONTRIBUTIONS
M.R., K.S.L., A.N. and L.H. planned and performed most experiments and wrote
the manuscript; P.A.L. performed experiments during the revision process; S.F.
performed some experiments with AID-deficient mice; A.B. sequenced persisting
kidney virus; A.N.H. helped isolate lymphocytes for cytotoxicity experiments,
K.F., L.H. and R.Z. helped perform experiments with B cell–transgenic mice and
provided materials; B.E. helped perform LCMV glycoprotein–specific ELISAs and
provided materials; D.M. did statistical testing; B.O. performed and supervised
immunohistology; P.G. and M.H. performed and supervised measurement of
serum hepatitis markers; A.T. and L.T.J. contributed to discussions and writing of
the manuscript; and H.H. and R.M.Z. supervised and financed the project and
helped write the manuscript.
Published online at http://www .nature.com/naturemedicine
Reprints and permissions information is available online at http://npg.nature.com/
reprintsandpermissions
1. Hilleman, M.R. Strategies and mechanisms for host and pathogen survival in acute and
persistent viral infections. Proc. Natl. Acad. Sci. USA 101 Suppl. 2, 14560–14566
(2004).
2. Peterlin, B.M. & Trono, D. Hide, shield and strike back: how HIV-infected cells avoid
immune eradication. Nat. Rev. Immunol. 3, 97–107 (2003).
3. Alcami, A. Viral mimicry of cytokines, chemokines and their receptors. Nat. Rev.
Immunol. 3, 36–50 (2003).
4. Wei, X. et al. Antibody neutralization and escape by HIV-1. Nature 422, 307–312
(2003).
5. Rehermann, B. & Nascimbeni, M. Immunology of hepatitis B virus and hepatitis C virus
infection. Nat. Rev. Immunol. 5, 215–229 (2005).
6. Fischer, S.A. et al. Transmission of lymphocytic choriomeningitis virus by organ
transplantation. N. Engl. J. Med. 354, 2235–2249 (2006).
7. Bruggeman, L.A. et al. Renal epithelium is a previously unrecognized site of HIV-1
infection. J. Am. Soc. Nephrol. 11, 2079–2087 (2000).
8. Arrieta, J.J. et al. In situ detection of hepatitis C virus RNA in salivary glands. Am. J.
Pathol. 158, 259–264 (2001).
9. Becker, J.L. et al. Epstein-Barr virus infection of renal proximal tubule cells:
possible role in chronic interstitial nephritis. J. Clin. Invest. 104, 1673–1681
(1999).
10. Stewart, J.P., Usherwood, E.J., Ross, A., Dyson, H. & Nash, T. Lung epithelial cells are
a major site of murine gammaherpesvirus persistence. J. Exp. Med. 187, 1941–1951
(1998).
11. Ahmed, R., Jamieson, B.D. & Porter, D.D. Immune therapy of a persistent and
disseminated viral infection. J. Virol. 61, 3920–3929 (1987).
ARTICLES
1322 VOLUME 13
[
NUMBER 11
[
NOVEMBER 2007 NATURE MED ICI NE
© 2007 Nature Publishing Group http://www.nature.com/naturemedicine
12. Zhang, H. et al. Human immunodeficiency virus type 1 in the semen of men
receiving highly active antiretroviral therapy. N. Engl. J. Med. 339, 1803–1809
(1998).
13. Marras, D. et al. Replication and compartmentalization of HIV-1 in kidney epithelium
of patients with HIV-associated nephropathy. Nat. Med. 8, 522–526 (2002).
14. Flodstrom, M. et al. Target cell defense prevents the development of diabetes after viral
infection. Nat. Immunol. 3, 373–382 (2002).
15. Lang, K.S. et al. Toll-like receptor engagement converts T-cell autoreactivity into overt
autoimmune disease. Nat. Med. 11, 138–145 (2005).
16. Wolf, D.G. et al. Emergence of late cytomegalovirus central nervous system disease in
hematopoietic stem cell transplant recipients. Blood 101, 463–465 (2003).
17. Lichterfeld, M. et al. Loss of HIV-1-specific CD8+ T cell proliferation after acute HIV-1
infection and restoration by vaccine-induced HIV-1-specific CD4+ T cells. J. Exp. Med.
200, 701–712 (2004).
18. Matloubian, M., Concepcion, R.J. & Ahmed, R. CD4+ T cells are required to sustain
CD8+ cytotoxic T-cell responses during chronic viral infection. J. Virol. 68,8056
8063 (1994).
19. Grakoui, A. et al. HCV persistence and immune evasion in the absence of memory T cell
help. Science 302, 659–662 (2003).
20. Kiepiela, P. et al. CD8(+) T-cell responses to different HIV proteins have discordant
associations with viral load. Nat. Med. 13, 46–53 (2007).
21. Thomson, M. et al. The clearance of hepatitis C virus infection in chimpanzees may not
necessarily correlate with the appearance of acquired immunity. J. Virol. 77, 862–870
(2003).
22. Bronke, C., Westerlaken, G.H., Miedema, F., Tesselaar, K. & van Baarle, D. Progression
to CMV end-organ disease in HIV-1-infected individuals despite abundance of
highly differentiated CMV-specific CD8+ T-cells. Immunol. Lett. 97, 215–224
(2005).
23. Sousa, A.E., Carneiro, J., Meier-Schellersheim, M., Grossman, Z. & Victorino, R.M.
CD4 T cell depletion is linked directly to immune activation in the pathogenesis of
HIV-1 and HIV-2 but only indirectly to the viral load. J. Immunol. 169, 3400–3406
(2002).
24. Silvestri, G. et al. Nonpathogenic SIV infection of sooty mangabeys is characterized by
limited bystander immunopathology despite chronic high-level viremia. Immunity 18,
441–452 (2003).
25. Kobasa, D. et al. Aberrant innate immune response in lethal infection of macaques
with the 1918 influenza virus. Nature 445, 319–323 (2007).
26. Lo, A.W., Tang, N.L. & To, K.F. How the SARS coronavirus causes disease: host or
organism? J. Pathol. 208, 142–151 (2006).
27. Staprans, S.I. et al. Enhanced SIV replication and accelerated progression to AIDS in
macaques primed to mount a CD4 T cell response to the SIV envelope protein. Proc.
Natl. Acad. Sci. USA 101, 13026–13031 (2004).
28. Recher, M. et al. Deliberate removal of T cell help improves virus-neutralizing antibody
production. Nat. Immunol. 5, 934–942 (2004).
29. Matter, M., Odermatt, B., Yagita, H., Nuoffer, J.M. & Ochsenbein, A.F. Elimination of
chronic viral infection by blocking CD27 signaling. J. Exp. Med. 203, 2145–2155
(2006).
30. Oxenius, A., Bachmann, M.F., Zinkernagel, R.M. & Hengartner, H. Virus-specific MHC-
class II-restricted TCR-transgenic mice: effects on humoral and cellular immune
responses after viral infection. Eur. J. Immunol. 28, 390–400 (1998).
31. Harada, Y., Muramatsu, M., Shibata, T., Honjo, T. & Kuroda, K. Unmutated immu-
noglobulin M can protect mice from death by influenza virus infection. J. Exp. Med.
197, 1779–1785 (2003).
32. Wang, X.Z. et al. Virus-specific CD8 T cells in peripheral tissues are more resistant to
apoptosis than those in lymphoid organs. Immunity 18, 631–642 (2003).
33. Hangartner, L. et al. Antiviral immune responses in gene-targeted mice expressing the
immunoglobulin heavy chain of virus-neutralizing antibodies. Proc. Natl. Acad. Sci.
USA 100, 12883–12888 (2003).
34. Legrand, E. et al. Course of specific T lymphocyte cytotoxicity, plasma and cellular viral
loads, and neutralizing antibody titers in 17 recently seroconverted HIV type 1-infected
patients. AIDS Res. Hum. Retroviruses 13, 1383–1394 (1997).
35. Andreansky, S. et al. The limits of protection by ‘memory’ T cells in Ig/ mice
persistently infected with a gamma-herpesvirus. Proc. Natl. Acad. Sci. USA 101,
2017–2022 (2004).
36. Crotty, S., McCausland, M.M., Aubert, R.D., Wherry, E.J. & Ahmed, R. Hypogamma-
globulinemia and exacerbated CD8 T cell mediated immunopathology in SAP-deficient
mice with chronic LCMV infection mimics human XLP disease. Blood 108, 3085–
3093 (2006).
37. Planz, O. et al. A critical role for neutralizing-antibody-producing B cells, CD4(+) T
cells, and interferons in persistent and acute infections of mice with lymphocytic
choriomeningitis virus: implications for adoptive immunotherapy of virus carriers. Proc.
Natl. Acad. Sci. USA 94, 6874–6879 (1997).
38. Lauterbach, H., Truong, P. & McGavern, D.B. Clearance of an immunosuppressive virus
from the CNS coincides with immune reanimation and diversification. Virol. J. 4,53
(2007).
39. Allen, T.M. et al. Tat-specific cytotoxic T lymphocytes select for SIV escape variants
during resolution of primary viraemia. Nature 407, 386–390 (2000).
40. Ramos-Casals, M. et al. Triple association between hepatitis C virus infection,
systemic autoimmune diseases, and B cell lymphoma. J. Rheumatol. 31, 495–499
(2004).
41. Oxenius, A. et al. Stimulation of HIV-specific cellular immunity by structured treatment
interruption fails to enhance viral control in chronic HIV infection. Proc. Natl. Acad.
Sci. USA 99, 13747–13752 (2002).
42. Battegay, M. et al. Quantification of lymphocytic choriomeningitis virus with an
immunological focus assay in 24- or 96-well plates. J. Virol. Methods 33, 191–198
(1991).
43. Junt, T. et al. Antiviral immune responses in the absence of organized lymphoid T cell
zones in plt/plt mice. J. Immunol. 168, 6032–6040 (2002).
ARTICLES
NATURE MEDIC INE VOLUME 13
[
NUMBER 11
[
NOVEMBER 2007 1323
© 2007 Nature Publishing Group http://www.nature.com/naturemedicine
... EBV-infected plasma cells in the CNS have been found synapsed with cytotoxic CD8 + T cells, suggesting a local inflammatory interaction initiated by EBV-positive B cells in the CNS 149 . There is evidence that EBV establishes an extralymphatic viral sanctuary in the CNS 150 , especially in vulnerable individuals during infectious mononucleosis, in which approximately 50% of memory B cells can be EBV positive 151 . However, several studies failed to find evidence of EBV-positive B cells in the CSF of patients with MS or MS lesions in the CNS [152][153][154][155][156][157] . ...
Article
Epstein–Barr virus (EBV) is a ubiquitous human lymphotropic herpesvirus with a well-established causal role in several cancers. Recent studies have provided compelling epidemiological and mechanistic evidence for a causal role of EBV in multiple sclerosis (MS). MS is the most prevalent chronic inflammatory and neurodegenerative disease of the central nervous system and is thought to be triggered in genetically predisposed individuals by an infectious agent, with EBV as the lead candidate. How a ubiquitous virus that typically leads to benign latent infections can promote cancer and autoimmune disease in at-risk populations is not fully understood. Here we review the evidence that EBV is a causal agent for MS and how various risk factors may affect EBV infection and immune control. We focus on EBV contributing to MS through reprogramming of latently infected B lymphocytes and the chronic presentation of viral antigens as a potential source of autoreactivity through molecular mimicry. We consider how knowledge of EBV-associated cancers may be instructive for understanding the role of EBV in MS and discuss the potential for therapies that target EBV to treat MS. Epstein–Barr virus infects most of the human population and, depending on other risk factors, contributes to the development of multiple sclerosis. In this Review, Soldan and Lieberman discuss supporting evidence and potential mechanisms that link Epstein–Barr virus to multiple sclerosis.
... arenaviruses such as LCMV will not kill the host cell by direct cytopathic effects, thus the virus has immense chances to maintain its tumor cell replication until the immune response is provoked within the tumour tissue, besides that it is also known that arenavirus replication is hardly limited by an anti-viral type I interferon response [21]. Another advantage in using LCMV is that it usually fails to induce rapid neutralising antibodies to limit tumoral replication early on [22]. The control of arenavirus is mostly dependent on CD8 + T cells, and as long as virus-specific CD8 + T cells do not infiltrate in the site of the tumour, arenavirus will keep replicating for several days or longer even if the tumour cells tend to respond to the virus-induced type I interferon. ...
Article
Full-text available
Viruses have been widely used to treat cancer for many years and they achieved tremendous success in clinical trials with outstanding results, which has led to the foundation of companies that develop recombinant viruses for a better tumor treatment. Even though there has been a great progress in the field of viral tumor immunotherapy, until now only one virus, the oncolytic virus talimogene laherparepvec (TVEC), a genetically modified herpes simplex virus type 1 (T-VEC), has been approved by the FDA for cancer treatment. Although oncolytic viruses showed progress in certain cancer types and patient populations but they have yet shown limited efficacy when it comes to solid tumors. Only recently it was demonstrated that the immune stimulatory aspect of oncolytic viruses can strongly contribute to their anti-tumoral activity. One specific example in this context are arenaviruses, which have been shown to be non-cytopathic in nature lead to the massive immune activation within the tumor resulting in strong anti-tumoral activity. This strong immune activation might be also linked to their noncytopathic features, as their immune stimulatory potential is not self-limiting as is the case for oncolytic viruses due to their fast eradication by anti-viral immune effects. Because of this strong immune activation, arenaviruses appear superior to oncolytic viruses when it comes to potent and long-lasting anti-tumor effects in a broad variety of tumor types. Currently one of the most promising therapeutics which has turned to be very much beneficial for the treatment of different cancer types is represented by antibodies targeting checkpoint inhibitors such as PD-1/PD-L-1. In this review, we will summarize anti-tumoral effects of arenaviruses, and will discuss their potential to be combined with checkpoint inhibitors for a more efficient tumor treatment, which further emphasizes that arenavirus therapy as a viroimmunotherapy can be an efficient tool for the better clearance of tumors.
... To achieve a strong and locally restricted (re-)activation of the immune system the LCMV is used as viral backbone. LCMV is almost non-cytopathic and can persist for several days to months in cell culture or mice [78]. This is one feature, which characterizes LCMV as a strong immune activator [79]. ...
Article
Full-text available
Virotherapy research involves the development, exploration, and application of oncolytic viruses that combine direct killing of cancer cells by viral infection, replication, and spread (oncolysis) with indirect killing by induction of anti-tumor immune responses. Oncolytic viruses can also be engineered to genetically deliver therapeutic proteins for direct or indirect cancer cell killing. In this review—as part of the special edition on “State-of-the-Art Viral Vector Gene Therapy in Germany”—the German community of virotherapists provides an overview of their recent research activities that cover endeavors from screening and engineering viruses as oncolytic cancer therapeutics to their clinical translation in investigator-initiated and sponsored multi-center trials. Preclinical research explores multiple viral platforms, including new isolates, serotypes, or fitness mutants, and pursues unique approaches to engineer them towards increased safety, shielded or targeted delivery, selective or enhanced replication, improved immune activation, delivery of therapeutic proteins or RNA, and redirecting antiviral immunity for cancer cell killing. Moreover, several oncolytic virus-based combination therapies are under investigation. Clinical trials in Germany explore the safety and potency of virotherapeutics based on parvo-, vaccinia, herpes, measles, reo-, adeno-, vesicular stomatitis, and coxsackie viruses, including viruses encoding therapeutic proteins or combinations with immune checkpoint inhibitors. These research advances represent exciting vantage points for future endeavors of the German virotherapy community collectively aimed at the implementation of effective virotherapeutics in clinical oncology.
Article
HIV encodes a milieu of biological functions in a small number of genes, and is pathologically optimized to perturb the immune system of humans and persist, resulting in dissemination. This review will introduce recent conceptual insights in the field of HIV basic research derived from technological advances.
Preprint
Full-text available
HIV and simian immunodeficiency virus (SIV) infections are known for impaired neutralizing antibody (NAb) responses. While sequential virus-host B cell interaction appears to be basally required for NAb induction, driver molecular signatures predisposing to NAb induction still remain largely unknown. Here we describe SIV-specific NAb induction following a virus-host interplay reciprocal to a congenital human antibody-dysregulating disease. Screening of seventy neutralization-resistant SIVmac239-infected macaques found nine NAb-inducing animals, with seven selecting for a specific CD8+ T-cell escape mutation in viral nef before NAb induction. This mutation reduced aberrant Nef interaction-mediated drive of B-cell maturation-limiting phosphoinositide 3-kinase (PI3K)/mammalian target of rapamycin complex 2 (mTORC2). In vivo imaging cytometry depicted preferential Nef perturbation of cognate Envelope-specific B cells, corroborating cognate B-cell maturation post-mutant selection up to NAb induction. Results exemplify a NAb induction pattern extrinsically reciprocal to human PI3K gain-of-function antibody dysregulation, shaped through two-front, sequential virus interaction with both wings of adaptive immunity.
Article
Full-text available
Interferon-beta (IFN-β) for Multiple Sclerosis (MS) is turning 30. The COVID-19 pandemic rejuvenated the interest in interferon biology in health and disease, opening translational opportunities beyond neuroinflammation. The antiviral properties of this molecule are in accord with the hypothesis of a viral etiology of MS, for which a credible culprit has been identified in the Epstein-Barr Virus. Likely, IFNs are crucial in the acute phase of SARS-CoV-2 infection, as demonstrated by inherited and acquired impairments of the interferon response that predispose to a severe COVID-19 course. Accordingly, IFN-β exerted protection against SARS-CoV-2 in people with MS (pwMS). In this viewpoint, we summarize the evidence on IFN-β mechanisms of action in MS with a focus on its antiviral properties, especially against EBV. We synopsize the role of IFNs in COVID-19 and the opportunities and challenges of IFN-β usage for this condition. Finally, we leverage the lessons learned in the pandemic to suggest a role of IFN-β in long-COVID-19 and in special MS subpopulations.
Article
Full-text available
Viral-based cancer therapies have tremendous potential, especially in the context of treating poorly infiltrated cold tumors. However, in tumors with intact anti-viral interferon (IFN) pathways, while some oncolytic viruses induce strong innate and adaptive immune responses, they are neutralized before exerting their therapeutic effect. Arenaviruses, particularly the lymphocytic choriomeningitis virus (LCMV) is a noncytopathic virus with preferential cancer tropism and evolutionary mechanisms to escape the immune system for longer and to block early clearance. These escape mechanisms include inhibition of the MAVS dependent IFN pathway and spike protein antigen masking. Regarding its potential for cancer treatment, LCMV is therefore able to elicit long-term responses within the tumor microenvironment (TME), boost anti-tumor immune responses and polarize poorly infiltrating tumors towards a hot phenotype. Other arenaviruses including the attenuated Junin virus vaccine also have anti-tumor effects. Furthermore, the LCMV and Pichinde arenaviruses are currently being used to create vector-based vaccines with attenuated but replicating virus. This review focuses on highlighting the potential of arenaviruses as anti-cancer therapies. This includes providing a molecular understanding of its tropism as well as highlighting past and present preclinical and clinical applications of noncytophatic arenavirus therapies and their potential in bridging the gap in the treatment of cancers weakly responsive or unresponsive to oncolytic viruses. In summary, arenaviruses represent promising new therapies to broaden the arsenal of anti-tumor therapies for generating an immunogenic tumor microenvironment.
Article
Full-text available
T cell-based therapies have been widely explored for the treatment of cancer and chronic infection, but B cell-based therapies have remained largely unexplored. To study the effect of B cell therapy, we adoptively transferred virus-specific B cells into mice that were chronically infected with lymphocytic choriomeningitis virus (LCMV). Adoptive transfer of virus-specific B cells resulted in increase in antibody titers and reduction of viral loads. Importantly, the efficacy of B cell therapy was partly dependent on antibody effector functions, and was improved by co-transferring virus-specific CD4 T cells. These findings provide a proof-of-concept that adoptive B cell therapy can be effective for the treatment of chronic infections, but provision of virus-specific CD4 T cells may be critical for optimal virus neutralization.
Article
Full-text available
Two gene-targeted immunoglobulin heavy chain transgenic mouse strains, TgH(KL25) and TgH(VI10), expressing neutralizing specificities for lymphocytic choriomeningitis virus and vesicular stomatitis virus, respectively, have been generated. Three days after lymphocytic choriomeningitis virus infection, TgH(KL25) mice showed a thymus-independent neutralizing IgM response followed by thymus-dependent (TD) IgG. In contrast, WT mice mounted only a TD IgG response around day 80. These observations indicated that not only structural properties of the virus but also immunological parameters such as the frequency of B cells were indicative for the induction of thymus-independent versus TD Ig responses. Naïve vesicular stomatitis virusspecific Ig heavy chain transgenic mice displayed greatly elevated natural antibody titers. However, despite these high naïve titers, de novo activation of naïve CD4+ T and B cells was not blocked. Therefore, B cells giving rise to natural antibodies do not participate in virus-induced antibody responses.
Article
Full-text available
Neutralizing antibodies (Nab) are a principal component of an effective human immune response to many pathogens, yet their role in HIV-1 infection is unclear. To gain a better understanding of this role, we examined plasma from patients with acute HIV infection. Here we report the detection of autologous Nab as early as 52 days after detection of HIV-specific antibodies. The viral inhibitory activity of Nab resulted in complete replacement of neutralization-sensitive virus by successive populations of resistant virus. Escape virus contained mutations in the env gene that were unexpectedly sparse, did not map generally to known neutralization epitopes, and involved primarily changes in N-linked glycosylation. This pattern of escape, and the exceptional density of HIV-1 envelope glycosylation generally, led us to postulate an evolving 'glycan shield' mechanism of neutralization escape whereby selected changes in glycan packing prevent Nab binding but not receptor binding. Direct support for this model was obtained by mutational substitution showing that Nab-selected alterations in glycosylation conferred escape from both autologous antibody and epitope-specific monoclonal antibodies. The evolving glycan shield thus represents a new mechanism contributing to HIV-1 persistence in the face of an evolving antibody repertoire.
Article
Full-text available
Chronic interstitial nephritis frequently accompanies renal diseases of different etiologies. Far less common is the entity of primary interstitial nephritis wherein the glomerular and vascular structures of the kidney are not the primary focus of the disease process. Using in situ hybridization and the polymerase chain reaction, we detected DNA from the Epstein-Barr Virus (EBV) exclusively in renal tissue of patients with the idiopathic variety of chronic interstitial nephritis. The EBV genome, but not that of cytomegalovirus or adenovirus, was detected primarily in renal proximal tubule cells. Furthermore, the CD21 antigen, which serves as the receptor for EBV in B lymphocytes, was detected by immunocytochemistry primarily on proximal tubule cells and was markedly upregulated in the EBV-infected tissue. Western blot analysis of primary cultures of normal proximal tubule cells identified a 140-kDa protein, confirming the expression of the CD21 antigen. Colocalization experiments using proximal and distal tubule markers confirmed that EBV DNA and the CD21 antigen are found primarily in proximal tubule cells. EBV infection of renal proximal tubular cells may participate in evoking a cellular immune response that results in a damaged renal interstitium.
Article
A transgenic mouse expressing MHC class II-restricted TCR with specificity for a lymphocytic choriomeningitis virus (LCMV) glycoprotein-derived T helper cell epitope was developed to study the role of LCMV-specific CD4+ T cells in virus infection in vivo. The majority of CD4+ T cells in TCR transgenic mice expressed the transgenic receptor, and LCMV glycoprotein-specific TCR transgenic CD4+ T cells efficiently mediated help for the production of LCMV glycoprotein-specific isotype-switched antibodies. In contrast, LCMV glycoprotein-specific TCR transgenic mice exhibited a drastically reduced ability to provide help for the generation of antibody responses specific for the virus-internal nucleoprotein, indicating that intramolecular/intrastructural help is limited to antigens that are accessible to B cells on the viral surface. Antiviral cellular immunity was studied with noncytopathic LCMV and recombinant cytopathic vaccinia virus expressing the LCMV glycoprotein. TCR transgenic mice failed to efficiently control LCMV infection, demonstrating that functional LCMV-specific CD4+ T cells – even if activated and present at extremely high frequencies – cannot directly mediate protective immunity against LCMV. Despite the fact that LCMV-primed CD4+ T cells from TCR transgenic mice as well as from control mice showed low MHC class II-restricted cytotoxic activity in vivo, this did not correlate with protection against LCMV replication in vivo. In contrast, CD4+ T cells from TCR-transgenic mice mediated efficient protection against infection with recombinant vaccinia virus. These results further support the need for different immune effector functions for protective immunity against different viral infections.
Article
Titres of lymphocytic choriomeningitis virus (LCMV) were determined on adherent fibroblast cell lines in 24, respectively 96 well plates. After absorption of virus by cells and 48 h incubation under a methylcellulose overlay, cell monolayers were fixed with 4% formaldehyde in phosphate buffered saline, permeabilized by incubating in 0.5% Triton X-100 in balanced salt solution and then stained with a monoclonal rat anti-LCMV and a peroxidase labeled second stage antibody. The sensitivity of the assay is within a factor of 2-4 of conventional plaquing methods. The method is quicker (2-3 days), as compared to conventional methods (4-6 days) and less expensive with respect to both workhours and materials involved. The method also detects poorly- or non-plaquin LCMV isolates, and therefore drastically reduces the needs for titration of LCMV in mice.
Article
Titers of lymphocytic choriomeningitis virus (LCMV) were determined on adherent fibroblast cell lines in 24- or 96-well plates. After absorption of virus by cells and 48 h incubation under a methylcellulose overlay, cell monolayers were fixed with 4% formaldehyde in phosphate-buffered saline, permeabilized by incubation in 0.5% Triton X-100 in balanced salt solution and then stained with a monoclonal rat anti-LCMV and a peroxidase-labeled second stage antibody. The sensitivity of the assay is within a factor of 2-4 of conventional plaquing methods. The method also detects poorly or non-plaquing LCMV isolates, and therefore drastically reduces the need for titration of LCMV in mice. The method is quicker (2-3 days), as compared to conventional methods (4-6 days) and less expensive in terms of work and materials.
Article
The mechanism of viral clearance was studied by using the mouse model of chronic infection with lymphocytic choriomeningitis virus. Distinct patterns of viral clearance and histopathology were observed in different organs after adoptive immune therapy of persistently infected (carrier) mice. Clearance from the liver occurred within 30 days and was accompanied by extensive mononuclear cell infiltrates and necrosis of hepatocytes. Infectious virus and viral antigen were eliminated concurrently. This pattern of viral clearance was also seen in most other tissues (i.e., lung, spleen, lymph nodes, pancreas, etc.). In contrast, a different pattern of clearance was observed in the brain. Infectious virus was eliminated within 30 days, but viral antigen persisted in the central nervous systems of treated carrier mice for up to 90 days. The urinary system was the most resistant to immune therapy. Elimination of infectious virus and viral antigen from the kidney took greater than 200 days and even then was not complete; trace levels of infectious virus were still present in the kidneys of some treated carrier mice. After immune therapy, viral antigen in the kidney was located within renal tubules that costained for intracellular mouse immunoglobulin G. This unusual staining pattern, coupled with the observation of large numbers of plasma cells within the kidney, suggests that virus-immunoglobulin G complexes found in the tubules may represent in situ immune complex formation as opposed to deposition of circulating immune complexes. In conclusion, these results suggest that the site (organ) of viral persistence is an important consideration in developing treatment strategies for controlling chronic viral infections.
Article
In this study, we have examined the relative contributions of CD4+ and CD8+ T cells in controlling an acute or chronic lymphocytic choriomeningitis virus (LCMV) infection. To study acute infection, we used the LCMV Armstrong strain, which is cleared by adult mice in 8 to 10 days, and to analyze chronic infection, we used a panel of lymphocyte-tropic and macrophage-tropic variants of LCMV that persist in adult mice for several months. We show that CD4+ T cells are not necessary for resolving an acute LCMV infection. CD4+ T-cell-depleted mice were capable of generating an LCMV-specific CD8+ cytotoxic T-lymphocyte (CTL) response and eliminated virus with kinetics similar to those for control mice. The CD8+ CTL response was critical for resolving this infection, since beta 2-microglobulin knockout (CD8-deficient) mice were unable to control the LCMV Armstrong infection and became persistently infected. In striking contrast to the acute infection, even a transient depletion of CD4+ T cells profoundly affected the outcome of infection with the macrophage- and lymphocyte-tropic LCMV variants. Adult mice given a single injection of anti-CD4 monoclonal antibody (GK1.5) at the time of virus challenge became lifelong carriers with high levels of virus in most tissues. Unmanipulated adult mice infected with the different LCMV variants contained virus for prolonged periods (> 3 months) but eventually eliminated infection from most tissues, and all of these mice had LCMV-specific CD8+ CTL responses. Although the level of CTL activity was quite low, it was consistently present in all of the chronically infected mice that eventually resolved the infection. These results clearly show that even in the presence of an overwhelming viral infection of the immune system, CD8+ CTL can remain active for long periods and eventually resolve and/or keep the virus infection in check. In contrast, LCMV-specific CTL responses were completely lost in chronically infected CD4-depleted mice. Taken together, these results show that CD4+ T cells are dispensable for short-term acute infection in which CD8+ CTL activity does not need to be sustained for more than 2 weeks. However, under conditions of chronic infection, in which CD8+ CTLs take several months or longer to clear the infection, CD4+ T-cell function is critical. Thus, CD4+ T cells play an important role in sustaining virus-specific CD8+ CTL during chronic LCMV infection. These findings have implications for chronic viral infections in general and may provide a possible explanation for the loss of human immunodeficiency virus-specific CD8+ CTL activity that is seen during the late stages of AIDS, when CD4+ T cells become limiting.