ArticlePDF Available

MHC Class I Cross-Presentation by Dendritic Cells Counteracts Viral Immune Evasion

Frontiers
Frontiers in Immunology
Authors:

Abstract and Figures

DCs very potently activate CD8⁺ T cells specific for viral peptides bound to MHC class I molecules. However, many viruses have evolved immune evasion mechanisms, which inactivate infected DCs and might reduce priming of T cells. Then MHC class I cross-presentation of exogenous viral Ag by non-infected DCs may become crucial to assure CD8⁺ T cell responses. Although many vital functions of infected DCs are inhibited in vitro by many different viruses, the contributions of cross-presentation to T cell immunity when confronted with viral immune inactivation in vivo has not been demonstrated up to now, and remains controversial. Here we show that priming of Herpes Simplex Virus (HSV)-, but not murine cytomegalovirus (mCMV)-specific CD8⁺ T cells was severely reduced in mice with a DC-specific cross-presentation deficiency. In contrast, while CD8⁺ T cell responses to mutant HSV, which lacks crucial inhibitory genes, also depended on CD8α⁺ DCs, they were independent of cross-presentation. Therefore HSV-specific CTL-responses entirely depend on the CD8α⁺ DC subset, which present via direct or cross-presentation mechanisms depending on the immune evasion equipment of virus. Our data establish the contribution of cross-presentation to counteract viral immune evasion mechanisms in some, but not all viruses.
mCMV responses do not depend on cross-presentation. BMDC were infected with mCMVwt, mCMVmut, or mock-infected. DCs were utilized to either stimulate 5 × 10⁴ CFSE-labeled TCR-transgenic gp33-41-specific CD8⁺ P14 T cells as described in Figure 1B (A). CFSE-profiles of gated CD8⁺ T cells are shown for the peptide concentration 0.1 ng/ml. Graphs show results of gated divided T cells of one experiment out of two with similar results. (B) Batf3-ko and C57BL/6-mice were infected i.v. with 4 × 10⁶ infectious particles of mCMVwt or mCMVmut. Mice were sacrificed 7 days later and spleens were analyzed for presence of mCMV-specific CD8⁺ T cells by ex vivo restimulation with the indicated mCMV-peptides and subsequent intracellular FACS-analysis for IFNγ-production. Dot plots are gated on lymphocytes first (not shown). (C) Data from (B) shown as bar graphs are mean ± SEM from n = 3 mice per group. Open bars represent C57BL/6-wild type (wt) mice and filled bars represent Batf3-ko mice. This experiment has been repeated twice with similar outcome. (D) CD11c-Rac- or C57BL/6-mice were infected i.v. with 4 × 10⁶ infectious particles of mCMVwt or mCMVmut. Mice were sacrificed 5 days later and spleens were analyzed for presence of mCMV-specific CD8⁺ T cells by ex vivo restimulation with the indicated mCMV-peptides and subsequent intracellular FACS-analysis for IFNγ-production. Dot plots are gated on lymphocytes first (not shown). (E) Data from (D) shown as bar graphs are mean ± SEM from n = 3 mice per group. Open bars represent C57BL/6-wt mice, black bars represent Batf3-ko mice, gray bars are non-immunized controls. This experiment has been repeated twice with similar outcome.
… 
This content is subject to copyright.
ORIGINAL RESEARCH ARTICLE
published: 26 November 2012
doi: 10.3389/fimmu.2012.00348
MHC class I cross-presentation by dendritic cells
counteracts viral immune evasion
Katrin Nopora1, Caroline A. Bernhard1, Christine Ried1, Alejandro A. Castello1, Kenneth M. Murphy2,
Peggy Marconi 3, Ulrich Koszinowski 4and Thomas Brocker1*
1Institute for Immunology, Ludwig-Maximilians-University Munich, Munich, Germany
2Department of Pathology and Immunology, Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO, USA
3Department of Life Science and Biotechnology, University of Ferrara, Ferrara, Italy
4Max von Pettenkofer-Institut, Ludwig-Maximilians-University Munich, Munich, Germany
Edited by:
Peter M.Van Endert, Université Paris
Descartes/INSERM, France
Reviewed by:
Sven Burgdorf, Rheinische
Friedrich-Wilhelms-Universität,
Germany
Marianne Boes, University Medical
Centre Utrecht, Netherlands
*Correspondence:
Thomas Brocker, Institute for
Immunology, Ludwig-Maximilians-
University Munich, Goethestraße 31,
80336 München, Germany.
e-mail: tbrocker@med.
uni-muenchen.de
Present address:
Alejandro A. Castello, Universidad
Nacional de Quilmes, Buenos Aires,
Argentina.
Katrin Nopora and
Caroline A. Bernhard contributed
equally to this paper and share the
first authorship.
DCs very potently activate CD8+T cells specific for viral peptides bound to MHC class
I molecules. However, many viruses have evolved immune evasion mechanisms, which
inactivate infected DCs and might reduce priming of T cells. Then MHC class I cross-
presentation of exogenous viral Ag by non-infected DCs may become crucial to assure
CD8+T cell responses. Although many vital functions of infected DCs are inhibited in vitro
by many different viruses, the contributions of cross-presentation toT cell immunity when
confronted with viral immune inactivation in vivo has not been demonstrated up to now,
and remains controversial. Here we show that priming of Herpes Simplex Virus (HSV)-
, but not murine cytomegalovirus (mCMV)-specific CD8+T cells was severely reduced
in mice with a DC-specific cross-presentation deficiency. In contrast, while CD8+T cell
responses to mutant HSV, which lacks crucial inhibitory genes, also depended on CD8α+
DCs, they were independent of cross-presentation.Therefore HSV-specific CTL-responses
entirely depend on the CD8α+DC subset, which present via direct or cross-presentation
mechanisms depending on the immune evasion equipment of virus. Our data establish
the contribution of cross-presentation to counteract viral immune evasion mechanisms in
some, but not all viruses.
Keywords: dendritic cells, cross-priming, immune evasion
INTRODUCTION
Many viruses utilize a diversity of mechanisms to evade the
immune system (Tortorella et al., 2000;Yewdell and Hill, 2002).
Especially herpesviruses are extremely potent immune evaders and
Herpes Simplex Virus (HSV) shuts down host cell transcription,
RNA splicing, and protein synthesis by expressing an arsenal of
viral proteins (Hardy and Sandri-Goldin, 1994;Hill et al., 1995;
Spencer et al., 1997;Song et al., 2001;Smiley, 2004). HSV-1 infects
DCs with high efficiency as part of its life cycle (Coffin et al.,
1998;Salio et al., 1999;Kruse et al., 2000;Mikloska et al., 2001).
Consequently, infected DCs are severely compromised and fail to
mature, do not upregulate expression of MHC and other surface
molecules, cannot produce cytokines nor migrate properly (Salio
et al., 1999;Kruse et al., 2000;Samady et al., 2003;Prechtel et al.,
2005). As a consequence, HSV-infected DCs are non-functional
and cannot efficiently prime naive T cells in vitro (Salio et al.,
1999;Kruse et al., 2000).
It has been speculated that non-infected fully functional
bystander DCs could cross-present exogenous viral Ag derived
from infected and dying cells to secure priming of virus-
specific CD8+T cells (Heath and Carbone, 2001;Jirmo et al.,
2009). By experimentally excluding infection of DCs, it was
demonstrated that cross-presentation is principally sufficient to
mediate anti-viral CD8+T cell responses (Sigal et al., 1999;Nor-
bury et al., 2001). The selective capacity of CD8α+DCs to take
up dead cells (Iyoda et al., 2002) and to cross-prime CD8 T cells
(den Haan et al., 2000) suggests, that this DC subset may be espe-
cially important in anti-viral immunity. Indeed, several studies
have found CD8α+DCs to present viral Ag to CD8 T cells, when
mice were infected with HSV-1, lymphocytic choriomeningitis
virus, vaccinia virus, influenza virus, or respiratory syncytial virus
(Allan et al., 2003;Smith et al., 2003;Belz et al., 2004a,b, 2005;
Bedoui et al., 2009;Jirmo et al., 2009;Lukens et al., 2009) and
ablation of CD8α+DCs in Batf-3-deficient mice abrogated T cell
responses to west nile virus completely (Hildner et al., 2008).
However, experiments with murine cytomegalovirus (mCMV)
and viral mutants lacking immune evasion genes have not revealed
substantial differences in CD8 T cell responses, leaving the ques-
tion on the role of cross-presentation in the situation of viral
immune evasion open (Gold et al., 2004;Munks et al., 2007). In
the present study we compare immune evasion-competent HSV
and mCMV with their respective mutant forms lacking inhibitory
genes. We demonstrate that CD8 T cell responses to all wt and
mutant viruses studied depend on CD8α+DCs, which perform
www.frontiersin.org November 2012 | Volume 3 | Article 348 | 1
Nopora et al. Cross-priming enhances generation of virus-specific CTL
both, direct and cross-presentation, depending on the grade of
inhibition of virus infected DCs.
RESULTS
To study the extent to which CD8α+cross-presenting DCs can
compensate immune evasion, we utilized the KOS wild type
(wt) strain HSVwt, and its mutant (mut) derivative HSVmut
(UL41,ICP4,22,27), which lacks four viral genes responsible
for different viral “immune evasion”-strategies (Krisky et al.,
1998;Lauterbach et al., 2004). Vhs, the virion host shutoff protein
encoded by the gene UL41 causes destabilization and degrada-
tion of infected host cell mRNAs and is among other effects
responsible for down regulation of MHC I synthesis and expres-
sion (Hill et al., 1994;Tigges et al., 1996;Hinkley et al., 2000;
Koppers-Lalic et al., 2001). ICP4 reduces the stability of host cell
mRNA (Mogensen et al., 2004), ICP22 modifies the host RNA
polymerase II (Rice et al., 1995), while ICP27 inhibits mRNA bio-
genesis leading to shutoff of host protein synthesis (Hardwicke and
Sandri-Goldin, 1994;Hardy and Sandri-Goldin, 1994). Deletion
of these viral genes rescued maturation and immune functions of
infected human monocyte-derived DCs in vitro (Samady et al.,
2003). To confirm the effect of these viral genes on capacities of
murine DCs to prime CD8+T cells in vitro, we infected bone
marrow derived DCs (BMDCs) with wt HSVwt or HSVmut and
utilized them as APC for naïve CD8 T cells. The priming of naïve
HSV-glycoproteinB (gB)-specific CD8+T cells by HSVwt-infected
BMDCs was extremely inefficient and only at the highest DC: T
cell-ratio very few divided cells could be detected (Figure 1A). In
contrast, when BMDCs were infected with HSVmut, they were
highly efficient to prime T cells to divide (Figure 1A). To exclude
the possibility that these results were caused by different amounts
of viral gB-antigen being expressed by cells infected with the
different variants of HSV, we loaded infected DCs with titrated
amounts of an HSV-irrelevant LCMVgp33-41 peptide and tested
their capacity to prime specific CD8+P14 T cells (Figure 1B).
HSVmut-infected DCs were as efficient in priming naïve P14
T cells as non-infected mature DCs (Figure 1B). In contrast,
HSVwt-infected DCs needed 104times higher peptide concen-
trations to induce comparable CD8+T cell priming (Figure 1B).
These results show that deletion of these viral genes can rescue the
capacity of infected DCs to prime CD8+T cells in vitro.
Next we tested, if these different effects of wt- and mut-viruses
on DCs were of relevance for in vivo-priming of virus-specific
CTL and investigated the role of CD8α+DCs. Comparative stud-
ies between wt virus and mutant variants are generally difficult
to interpret due to different and partially uncharacterized viral
properties. Therefore we compared in the following studies only
CD8+T cell responses to same virus, either wt or mut, but in dif-
ferent mouse strains. To study the role of CD8α+DCs, we infected
Batf3/mice, which lack the transcription factor Batf3 leading to
DC (mock)
DC (HSVwt)
DC (HSVmut)
0
10
20
30
40
50
60
70
80
90
% divided cells
1/80 1/40 1/20
DC : T cell ratio
CFSE
+ DC (HSVwt) + DC (HSVmut)
gBT-1A
+ DC
CFSE
+ DC (HSVwt)
+ LCMVgp33-41
+ DC (HSVmut)
+ LCMVgp33-41
P14
+ DC
+ LCMVgp33-41
DC (mock)
DC (HSVwt)
DC (HSVmut)
% dividing cells
peptide (ng/ml)
0
10
20
30
40
50
60
70
80
90
100
010
-3 10
-1 10
110
3
B
cell numbercell number
FIGURE 1 | Deletion of HSV-1 immune evasion genes rescues
APC-functions of infected DCs in vitro.BMDC were infected with
HSVwt, HSVmut, or mock-infected and utilized to stimulate CFSE-labeled
TCR-transgenic HSVgB-specific CD8+gBT-I T cells (A). In addition DCs
were loaded with the LCMVgp33-41 peptide at the indicated
concentration and cultured with CFSE-labeled TCR-transgenic
gp33-41-specific CD8+P14 T cells (B). Appropriate conditions for DC:
Tcell ratio and peptide concentration were determined by titration [(A,B)
side insets]. Main graphs show results of CFSE-profiles of gated CD8+T
cells for one experiment out of four with similar results in which 2500
DCs were cultured with 5×104CFSE-labeledT cells for 4 days and
peptide concentration was 0.1 ng/ml.
Frontiers in Immunology | Antigen Presenting Cell Biology November 2012 | Volume 3 | Article 348 | 2
Nopora et al. Cross-priming enhances generation of virus-specific CTL
defective development of CD8α+and CD103+CD11bDCs, the
two major cross-presenting DC-subpopulations (Hildner et al.,
2008;Edelson et al., 2010). As HSVmut does not interfere effi-
ciently with immune functions of infected DCs (Figures 1A,B),
we speculated that CD8+T cell responses against this viral
mutant should be more independent of cross-presentation than
those induced by HSVwt. While C57BL7/6 mice mounted strong
HSVgB-specific CD8+T cell responses as measured with the
respective MHC-tetramers, we could not detect significant gB-
specific CD8+T cells in HSVwt-infected Batf3/-mice at all
(Figure 2, upper panel). Surprisingly, also HSVmut, despite of
being devoid of major inhibitory genes, could not elicit gB-specific
T cell responses in Batf3/-mice, while it did so very efficiently
in wt mice (Figure 2, upper panel). When the spleens of the same
animals were monitored for IFN-γ-producing CD8+T cells, only
very low frequencies of IFN- γ+CD8+T cells were detectable
in HSVwt-infected Batf3/-mice (Figure 2, lower panel). IFN-
γ-producing CD8+gB-specific CD8+T cells were not present in
HSVmut-infected mice at all (Figure 2, lower panel). This data
indicates that the DC-subpopulations lacking in Batf3/-mice
are central for inducing CD8+T cell responses upon infection
with HSVwt and HSVmut.
In order to discriminate between the contributions of direct vs.
cross-presentation we next infected CD11c-Rac mice with HSV
viruses. In DCs of these mice, the dominant negative N17Rac-
transgene causes inhibition of uptake of exogenous soluble, cel-
lular, or apoptotic antigen leading to strong reduction of cross-
presentation (Kerksiek et al., 2005). As a consequence, CD11c-Rac
mice show deficient peripheral tolerance to self proteins (Luck-
ashenak et al., 2008), absent CD8 T cell responses to extracellu-
lar bacteria, to cell associated proteins, to apoptotic, or soluble
protein antigens (Kerksiek et al., 2005;Neuenhahn et al., 2006;
Luckashenak et al., 2008). Upon infection with HSVwt-virus we
could detect approximately half the frequency and numbers of
HSVgB-specific CD8+T cells in spleens of cross-presentation-
defective CD11c-Rac mice, as compared to non-transgenic lit-
termates (Figure 3A). Both, detection of specific T cells by
MHCI-tetramers (Figure 3A, upper panel), as well as analysis
of IFN-γ-producing CD8+T cells upon restimulation with viral
HSVgB-peptide in vitro (Figure 3A, lower panel) revealed strongly
reduced frequencies and total numbers of HSV-specific CD8+
T cells in CD11c-Rac mice. These differences were highly sig-
nificant, also when data from several independent experiments
were pooled (Figure 3B). In marked contrast, infection with
HSVmut elicited comparable frequencies and total amounts of
HSVgB-specific IFN-γ-producing CD8+T cells in CD11c-Rac-
and control mice (Figure 3C). These data indicate, that lack of
inhibitory genes in HSVmut allows normal priming of HSVgB-
specific CD8+T cells independently of cross-priming in CD11c-
Rac mice. It has been speculated that CD8+DCs preferentially
capture dying cells (Iyoda et al., 2002) and efficiently cross-present
relatively high-dose tissue-associated antigens (Kurts et al., 1998)
as probably occur during viral infections. To test if dependency
on cross-presentation was observed also upon inoculation with
low doses of virus, we infected mice with graded amounts of
HSVwt or HSVmut (Figure 3D). Our data indicate that indepen-
dently of the amount of virus utilized for infection, the priming
of approximately half of all HSV-specific CD8+T cells depends
on cross-presentation, as the responses to infection with HSVwt
were reduced approximately 50% in CD11c-Rac mice (Figure 3D).
However, when viral immune evasion genes were deleted in HSV-
mut, the amount of primed HSVgB-specific CD8+T cells in
CD11c-Rac mice was indistinguishable from those observed in
wt mice (Figure 3D).
We next tested if murine CMV (mCMVwt) and its triple
mutant mCMVmut, which is devoid of several MHC class I
0 1021031041050 10210 3104105
0 1021031041050 10210 3104105
0 102103104105
0
102
103
104
105
control control Batf3-ko control Batf3-ko
HSVwt HSVmut
gB Tetramer
CD44
0
1
2
3
4
5
6
gBTet+ CD8+ T cells (%)
control
Batf3-ko
HSVwt HSVmut
0
2.5
5.0
7.5
10
gBTet+ CD8+ T cells (x 10-5)
HSVwt HSVmut
0 1021031041050 10210 3104105
0 102103104105
0 102103104105
0 102103104105
0
102
103
104
105
IFN-γ
CD8
IFN-γ+ CD8+ T cells (%)
HSVwt HSVmut
0
1
2
3
4
5
6
7
**
-
--
no virus
FIGURE 2 | Batf3-ko mice cannot mount HSV-specific CD8 T cell
responses. Batf3-ko and C57BL/6-mice were infected i.v. with 4×106
infectious particles of HSV. Mice were sacrificed 5days later and spleens
were analyzed for presence of HSVgB-specific CD8+T cells with
H-2 Kb/gB498-505-tetramers (top panel) or by ex vivo restimulation with gB498-505
peptide and subsequent intracellular FACS-analysis for IFNγ-production (lower
panel). Dot plots are gated on CD8+T cells (upper panel) or lymphocytes
(lower panel) first (not shown). Data shown as bar graphs are mean±SEM
from n=3 mice per group. (**P=0.0018 as compared to unstimulated
control). Open bars represent C57BL/6-wild type mice and filled bars
represent Batf3-ko mice.This experiment has been repeated twice with
similar outcome.
www.frontiersin.org November 2012 | Volume 3 | Article 348 | 3
Nopora et al. Cross-priming enhances generation of virus-specific CTL
0
1
2
3
4
5
0
1
2
3
gBTet+ CD8+ T cells (%)
gBTet+ CD8+ T cells (x 10-5)
0
0.5
1.0
1.5
2.0
2.5
3.0
3.5
0
0.5
1.0
1.5
2.0
2.5
HSVwt
gBTet+ CD8+ T cells (x 10-5)
gBTet+ CD8+ T cells (%)
*** *
0
50
100
150
% gBTet+ of CD8+ T cells
(relative to B6)
control
gB Tetramer
control CD11c-Rac
CD44
IFN-γ
CD8
010 21031041050 10 21031041050 10210 3104105
0
102
103
104
105
0.0
2.5
5.0
7.5
10.0
control
CD11c-Rac
IFN-γ+ CD8+ T cells (%)
0.5
1.0
1.5
2.0
2.5
3.0
3.5
4.0
4.5
5.0
5.5
6.0
6.5
IFN-γ+ CD8+ T cells (%)
*
0 10
2
10
3
10
4
10
5
0
10
2
10
3
10
4
10
5
0 10
2
10
3
10
4
10
5
0 10
2
10
3
10
4
10
5
0 10
2
10
3
10
4
10
5
0 10
2
10
3
10
4
10
5
0 10
2
10
3
10
4
10
5
0
10
2
10
3
10
4
10
5
0 10 210 310 410 5
0
10 2
10 3
10 4
10 5
0 10 210 310 410 5
0 10 210 310 410 5
gB Tetramer
CD44
IFN-γ
CD8
HSVwt
control control CD11c-Rac
HSVmut
control
CD11c-Rac
0.0
0.5
1.0
1.5
2.0
2.5
3.0
3.5
4.0
4.5
5.0
5.5
6.0
6.5
control
CD11c-Rac
IFN-γ+ CD8+ T cells (%)
0.4 4 40
HSVwt titers (1x105)
0
*
*
*
0.0
2.5
5.0
7.5
10.0
control
CD11c-Rac
IFN-γ+ CD8+ T cells (%)
0.4 4 40
HSVmut titers (1x105)
0
AB
C
HSVwt
HSVwt
HSVmut
HSVmut
HSVmut-
D
-
-
-
--
control
CD11c-Rac
no virus
no virus
***
FIGURE 3 | Cross-presentation deficient CD11c-Rac mice mount
reduced CD8T cell responses to HSVwt, but normal responses to
immune evasion-deficient HSVmut. Mice were immunized i.v. with
4×106infectious particles (A,B,C) or graded amounts (D) of HSVwt or
HSVmut and 5 days later CD8+T cells from spleens were analyzed as
described in Figure 2. Dot plots are gated on CD8+T cells [(A,C) upper
panel] or all lymphocytes [(A,C) lower panel] (not shown). Data from one
out of four experiments with similar results (n=3 mice per group) are
shown as mean ±SEM in bar graphs (a, c; side insets), t-test analysis, a,
upper panel: ***P<0.0001; *P=0.0284; a, lower panel *P=0.0243. (B)
Pooled data from seven experiments (n=3 mice per group, total n=21)
are displayed as percent of control (C57BL/6).The mean of each control
group was set to 100% and the data from the CD11c-Rac-groups were
calculated relative to the respective control group (***P=0.0001). (D)
Mice were immunized with the indicated amounts of HSVwt or HSVmut.
Analyses were performed as described in (A). HSVwt 0.4×105,
*P=0.0221; 4 ×105, *P=0.0223; 40 ×105, *P=0.0243; One out of two
experiments with similar outcome is shown (n=3 mice per group).
Frontiers in Immunology | Antigen Presenting Cell Biology November 2012 | Volume 3 | Article 348 | 4
Nopora et al. Cross-priming enhances generation of virus-specific CTL
inhibitory genes would be similarly dependent on cross-presenting
CD8α+DCs. The mutant form of mCMVwt (C3X), mCMVmut
(m04m06m152; Wagner et al., 2002), lacks three known
“immunoevasins” that act on the MHC class I presentation path-
way. While m04 binds to MHC class I (Kleijnen et al., 1997)
and prevents activation of T cells (Kavanagh et al., 2001), m06
retargets MHC class I to lysosomal degradation (Reusch et al.,
1999) and m152 retains MHC I/peptide complexes in the ER-
cis-Golgi compartment (Ziegler et al., 1997) leading to an over-
all reduction of MHC class I surface expression. Accordingly,
when mCMVwt- or mCMVmut-infected gp33-41 peptide-pulsed
BMDC were cocultured with gp33-41 peptide-specific P14 T
cells, the mCMVmut-infected DCs induced more efficient T cell
responses as compared to DCs infected with mCMVwt. How-
ever, in comparison to the HSV study (Figures 1A,B), the over-
all inhibitory effect of mCMVwt virus was weaker as compared
to HSV (Figure 4A).
In analogy to the experiments with HSV described above, we
next infected wt and Batf3-ko mice with the two viruses and mea-
sured IFN-γproducing virus-specific CD8 T cells. CTL-responses
against both, mCMVwt and mCMVmut, entirely depended on
the presence of CD8α+DCs, as we could not detect mCMV-
specific CD8 T cells in Batf-3 ko mice immunized with either
virus (Figures 4B,C). To further analyze if mCMV-specific CD8 T
cells would be primed via direct or cross-presentation by CD8α+
DCs, we infected CD11c-Rac mice and compared the responses to
those elicited in non-transgenic littermates (Figure 4D). How-
ever, and in contrast to the results obtained with HSVwt and
HSVmut, mCMV-specific cytotoxic T cells were indistinguish-
able between wt and CD11c-Rac mice for mCMVwt as well as
mCMVmut (Figure 4E). Therefore we conclude that priming of
mCMV-specific T cells crucially depends on CD8α+DCs, but is
independent of their cross-priming capacities.
DISCUSSION
CD8α+DCs are important for immunity as they have the spe-
cific ability to produce high levels of IL-12, direct Th1 responses
and produce CD8+T cell responses due to their capacity to cross-
present exogenous Ag (Shortman and Heath, 2010). In our study
we investigated the interdependency of CD8α+DCs, cross- and
direct presentation with viral inhibition of immune functions.
Such functionally inhibitory immune subversion mechanisms,
which affect DCs, have been identified for HSV and mCMV
and were deleted in the respective mutant strains selected for
the present study. A key factor for HSV with respect to inhibi-
tion of DC-functions is vhs, which inhibits expression of MHC
class I, class II, cytokine and chemokine production (reviewed
in Smiley, 2004). Accordingly, vhs-null mutations show severely
impaired viral pathogenesis and replication in mouse models
in vivo (Strelow and Leib, 1996;Leib et al., 1999), suggesting
that vhs is a virulence factor of HSV causing defects of DC-
functions and host immunity (reviewed in Smiley, 2004). We find
that DCs infected with HSVmut can very well prime CD8+T
cells in vitro. This finding suggests that also in vivo HSVmut-
infected cells should be able to prime CD8 T cells directly and
do not depend on cross-presentation by non-infected bystander
DCs. Our interpretation is corroborated by the finding that in
CD11c-Rac mice, which have reduced cross-priming capacities,
CD8 T cell responses to HSVmut are normal, and we conclude
that they do not depend on cross-priming. As HSVmut-infected
Batf3/-mice do not mount HSV-specific CD8 T cell-responses
in vivo, we assume that the DC-subpopulations absent in these
mice are responsible for direct HSV-presentation. In contrast,
HSVwt-infected DCs are severely inhibited to prime CD8 T cells
in vitro, HSVwt-induced CD8 T cell responses are also absent in
Batf3/-mice and do depend on cross-presenting DCs, as they
are strongly reduced in CD11c-Rac mice. A potential caveat to this
explanation could be the fact, that CD11c-Rac mice do not only
have a defect in endocytosis and cross-presentation, but also have
slightly lower numbers of CD8α+DCs in their spleens (Kerksiek
et al., 2005). It is therefore theoretically possible, that the reduced
T cell responses rather reflect the reduced numbers of CD8α+
DCs than their capacities to cross-present. However, this explana-
tion is unlikely, as responses to HSVmut are identical in both, wt
and CD11c-Rac mice, indicating sufficient DC numbers for the
induction of optimal CTL-responses.
In contrast, HSVwt-infected DCs can still prime CD8+T
cells in vitro (Figure 1), albeit with much lowered efficacy.
Such residual” DC-activity could account for the lower T cell
response measured in HSVwt-infected CD11c-Rac mice, when
cross-presentation is suboptimal. Our results therefore suggest,
that (i) CD8α+DCs are responsible for both, direct and cross-
priming of HSV-specific CD8+T cells and (ii) that cross-
priming becomes more important when the functional inhibition
of infected DCs increases. Cross-priming can compensate viral
immune evasion very efficiently. As a consequence, CD8 T cell
responses to HSVwt and HSVmut become indistinguishable in
cross-presentation competent mice.
This observation seems to parallel earlier findings with other
herpesviruses such as mCMV, where deletion of genes showed lit-
tle or no impact on T cell responses in wt mice in vivo (Gold
et al., 2004;Munks et al., 2007). In the past it has therefore been
difficult to establish a role for cross-presentation by using viruses
and their specific mutants. For example, removal of mCMV genes
affecting MHC class I expression had no effect on the CD8+T
cell responses in vivo (Munks et al., 2007). The interpretation
of these findings were that either directly infected DCs can still
prime CD8+T cells efficiently despite viral inhibition, or that
cross-priming is so potent that it dominates CD8+T cell priming
to mCMV (Snyder et al., 2010), making responses to mutant and
wt virus similar (Munks et al., 2007). It has also been discussed
that responses induced by mCMVwt and mutant strains in pre-
vious studies were similar, because in addition to inhibition of
MHC class I presentation, mCMV is known to also inhibit other
functions of infected DCs. For example shutting off expression of
costimulatory molecules and upregulating inhibitory ligands by
mCMV results in failure to prime CD8+T cells in vitro (Andrews
et al., 2001;Loewendorf et al., 2004;Benedict et al., 2008).However,
these inhibitory mechanisms of mCMV have not been entirely
identified yet and therefore the responsible viral genes were nei-
ther deleted in the respective experiments (Munks et al., 2007)
nor in our study. In a recent publication, Torti et al. (2011)
analyzed primary and memory CTL-responses to a single dele-
tion mCMV-mutant (mCMVm157) in Batf3/-mice. While,
www.frontiersin.org November 2012 | Volume 3 | Article 348 | 5
Nopora et al. Cross-priming enhances generation of virus-specific CTL
m139(T8L)
M45(H9I)
M57(S9V)
0
1
2
3
0
1
2
3
IFN-γ+ CD8+ T cells (%)
IFN-γ+ CD8+ T cells (%)
B6 Batf3-ko
mCMVwt
B6 Batf3-ko
mCMVmut
IFN-γ
CD8
control
mCMVwt mCMVmut
m139 M45 M57
B6
Batf3-ko
B6
Batf3-ko
mCMVmut
0
1
2
3
4
mCMVwt
m139 M45 M57 m141 M38 -
0
1
2
3
4
5
6
7
B6
Rac
control
IFN-γ+ CD8+ T cells (%)
IFN-γ+ CD8+ T cells (%)
IFN-γ
CD8
m139
B6 Rac
mCMVwt
B6 Rac
mCMVmut
control
B
C
D
E
m139 M45 M57
m139 M45 M57 m141 M38 -
+ DC (mCMVwt)
+ LCMVgp33-41
+ DC (mCMVmut)
+ LCMVgp33-41
P14
+ DC
+ LCMVgp33-41
% dividing cells
0
10
20
30
40
50
60
70
80
90
100
010
-3 10
-1 10
110
3
DC (mock)
DC (mCMVwt)
DC (mCMVmut)
A
CFSE
cell number
peptide (ng/ml)
FIGURE 4 | Continued
Frontiers in Immunology | Antigen Presenting Cell Biology November 2012 | Volume 3 | Article 348 | 6
Nopora et al. Cross-priming enhances generation of virus-specific CTL
FIGURE 4 | mCMV responses do not depend on cross-presentation.
BMDC were infected with mCMVwt, mCMVmut, or mock-infected. DCs were
utilized to either stimulate 5 ×104CFSE-labeledTCR-transgenic
gp33-41-specific CD8+P14 T cells as described in Figure 1B (A). CFSE-profiles
of gated CD8+T cells are shown for the peptide concentration 0.1 ng/ml.
Graphs show results of gated divided T cells of one experiment out of two
with similar results. (B) Batf3-ko and C57BL/6-mice were infected i.v. with
4×106infectious particles of mCMVwt or mCMVmut. Mice were sacrificed
7 days later and spleens were analyzed for presence of mCMV-specific CD8+
T cells by ex vivo restimulation with the indicated mCMV-peptides and
subsequent intracellular FACS-analysis for IFNγ-production. Dot plots are
gated on lymphocytes first (not shown). (C) Data from (B) shown as bar
graphs are mean ±SEM from n=3 mice per group. Open bars represent
C57BL/6-wild type (wt) mice and filled bars represent Batf3-ko mice. This
experiment has been repeated twice with similar outcome. (D) CD11c-Rac- or
C57BL/6-mice were infected i.v. with 4 ×106infectious particles of mCMVwt
or mCMVmut. Mice were sacrificed 5 days later and spleens were analyzed
for presence of mCMV-specific CD8+T cells by ex vivo restimulation with the
indicated mCMV-peptides and subsequent intracellular FACS-analysis for
IFNγ-production. Dot plots are gated on lymphocytes first (not shown). (E)
Data from (D) shown as bar graphs are mean ±SEM from n=3 mice per
group. Open bars represent C57BL/6-wt mice, black bars represent Batf3-ko
mice, gray bars are non-immunized controls.This experiment has been
repeated twice with similar outcome.
similar to our findings, primary CD8 T cell responses to several
epitopes were impaired, development of memory T cells rather
seemed to depend on direct priming, as they were normal in
Batf3/-mice. Although mCMVwt exerts inhibitory effects on
DCs in vitro in our experiments, these effects did apparently not
affect CD8 T cell immunity in vivo. Different experimental strate-
gies have suggested that cross-presentation plays a dominant role
in CD8 T cell priming during viral infection (Wilson et al., 2006;
Snyder et al., 2010),but intravital microscopy could document also
direct priming of naive CD8 T cells by DCs infected with vaccinia
virus (Hickman et al., 2008), which also strongly inhibits DC-
functions (Engelmayer et al., 1999). The fact that the responses in
control and CD11c-Rac mice were comparable for both,mCMVw t
as well as mCMVmut, suggests that cross-presentation does not
contribute substantially to mCMV T cell immunity. Eventually,
mCMV-mediated inhibition of DCs is not sufficient to make
cross-presentation necessary. In contrast, the lack of responses in
Batf3/-mice indicates absolute necessity for (direct) presenta-
tion by CD8α+DCs. Accordingly, our findings that Batf3/-mice,
which are deficient for CD8α+DCs, cannot mount CD8+T cell
responses to neither HSV, HSVmut, mCMV, mCMVmut, under-
line that this DC-subpopulation is responsible for both direct
priming upon infection and cross-presentation of viral material
from infected surrounding cells. However, our findings can even-
tually not be generalized for all viruses and routes of infection. For
example, the“natural” route of infection for HSV is via the skin or
mucosa, where other DC-subpopulations will be involved in direct
and cross-presentation. In addition, the deleterious effects of viral
immune evasion genes may be different in different DC subtypes.
Also the interplay of different DC-subpopulation in skin, mucosa
and the respective draining lymph nodes may be different from
the scenario in the spleen.
The roles direct vs. cross-priming play during viral infection
are certainly dependent on the type of virus, its cellular tropism
and the route of infection. While our results establish that cross-
presentation counteracts effects of viral inhibitory genes on DCs,
full understanding of the different contributions of direct vs. cross-
presentation will help to improve classical vaccination as well as
DC-targeting approaches.
MATERIALS AND METHODS
ANIMALS
CD11c-Rac- (Kerksiek et al., 2005), Batf3/- (Hildner et al.,
2008), P14- (Pircher et al., 1989), and gBT-1-mice (Mueller et al.,
2002) have been described before and were all maintained on the
C57BL/6 background. Mice were infected with virus preparation
diluted in 50 µl PBS and sacrificed with CO2at the indicated time
points. Mice were bred and housed at the animal facilities of the
Institute for Immunology (LMU, Munich, Germany) and treated
in accordance with established guidelines of the Regional Ethics
Committee of Bavaria. Animal protocols were approved by local
authorities.
VIRUSES AND INFECTIONS
HSVwt (KOS), HSVmut (UL41,ICP4,22,27), mCMVwt (C3X),
mCMVmut (m04m06m152) were prepared and utilized as
described previously (Wagner et al., 2002;Lauterbach et al., 2004).
Mice were infected intravenously (i.v.) with the amount of virus
indicated in the respective Figure legends.
CELL ISOLATION AND PURIFICATION
Single cell suspensions from spleens or lymph nodes were obtained
by mechanical disruption using a pestle followed by enzymatic
digestion in serum-free RPMI medium containing Liberase CI
(0.42 mg/ml) and DNase I (0.2 mg/ml, both from Roche, Basel,
Switzerland) for 20 min at 37˚C. Cells were passed through a
70 µm nylon mesh strainer. Cells were counted on a cell counter
(Beckman Coulter, Munich, Germany).
GENERATION OF BONE MARROW DERIVED DENDRITIC CELLS
Femurs and tibiae were flushed with IMDM and erythrocytes were
lysed by incubation in ACK buffer for 2 min at room temperature.
1×107bone marrow cells were plated in 10ml IMDM contain-
ing 10% heat inactivated FCS, 2 mM glutamine, 100 U/ml peni-
cillin, 100 µg/ml streptomycin sulfate, 50 µM 2-mercaptoethanol,
and 20 ng/ml GM-CSF (IMDM complete) in Petri-dishes. On
day three suspension cells and loosely adherent cells were dis-
lodged by gentle pipetting and adherent cells were subsequently
released by incubation in cold PBS containing 1 mM EDTA.
7.5 ×106cells were reseeded in 10 ml fresh IMDM complete per
10 cm dish.
INFECTION OF DC
For in vitro studies BMDCs were infected with virus as described
previously (Samady et al., 2003) and utilized 48 h post trans-
fection for T cell stimulation assays. DC (5 ×105) from day 7
BMDC-cultures were pelleted at 1,400 rpm for 5 min at room tem-
perature. The DC were then infected at a multiplicity of infection
www.frontiersin.org November 2012 | Volume 3 | Article 348 | 7
Nopora et al. Cross-priming enhances generation of virus-specific CTL
(MOI) of 1, unless otherwise stated, by resuspension in 200 ml
of IMDM medium containing 5 ×105PFU of virus for 1 h at
37˚C and 5% CO2. One milliliter of IMDM supplemented with
granulocyte-macrophage colony-stimulating factor (0.1 mg/ml)
was then added, and the DC were incubated at 37˚C and 5% CO2.
CFSE LABELING
P14 and gB-T1 T cells were purified from lymphocyte cell sus-
pensions by negative selection (CD8 T cell columns; R&D Sys-
tems, Minneapolis, MN, USA). For CFSE labeling 1–50 ×106
erythrocyte-free cells from lymph nodes and spleens were washed
twice with PBS and labeled with 5 µM CFSE (Molecular Probes,
Eugene, OR, USA) for 10 min at 37˚C in PBS. After stopping the
reaction (PBS, 2% FBS) and washing in PBS, cells were utilized for
the in vitro assays.
T CELL STIMULATION
5×104CFSE-labeled P14 and gB-T1 T cells were cultured for
4 days with 2500 DCs, which were either virus infected or loaded
with 1 µg/ml of the respective peptide.
ANTIBODIES AND FLOW CYTOMETRY
Lymphocytes were analyzed using anti-CD8a-APC, anti-CD44-
FITC, CD19-PerCP, IFNg-PE from Caltag (Burlingame, CA,
USA). H-2 Kb/gB498–505-tetramer-PE complexes were purchased
from ProImmune Limited. Staining of surface molecules
was performed with 1 ×106to 6 ×106cells in cold stain-
ing buffer for 30 min at 4˚C (15 min at room tempera-
ture in the dark when MHC multimers were used). Dead-
cell exclusion was attained by incubation with 1 µg/ml ethid-
ium monoazide bromide (EMA, Molecular Probes) prior to
surface staining or the addition of 0.8 mg/ml propidium iodide
(PI, Sigma). Intracellular staining for cytokines was performed
with the Cytofix/Cytoperm kit (PharMingen). Flow cytome-
try was performed with a FACSCalibur or FACSaria (Bec-
ton Dickinson), and data were analyzed with FlowJo software
(Tree Star).
STIMULATION OF CYTOKINE PRODUCTION BY EPITOPE-SPECIFIC T
CELLS
Splenocyte suspensions were prepared according to standard pro-
cedures and lymphocytes were stimulated with 1 µg/ml of the
indicated peptides in the presence of Brefeldin A (10 µg/ml;
Sigma, St. Louis, MO, USA). Cells were surface stained for 30min
at 4˚C before the fixation and permeabilization in 500 µl 2x
FacsLyse (BD Biosciences) containing 0.05% Tween 20 (Sigma-
Aldrich, St. Louis, MO, USA) for 10 min at room temper-
ature. The intracellular staining of cytokines was performed
for 30 min at room temperature. Analytic flow cytometry was
performed on a FACScanto II (Becton Dickinson). Analysis
was performed using FlowJo software (Tree Star, San Carlos,
CA, USA).
STATISTICAL ANALYSIS
All statistical analyses were performed using the two-tailed Stu-
dent’s t-test with unequal variance.
ACKNOWLEDGMENTS
We thank A. Bol and W. Mertl for assistance with animal
maintenance and experimentation. This work was supported by
the Deutsche Forschungsgemeinschaft (DFG) grant BR1889/5 to
Thomas Brocker.
REFERENCES
Allan, R. S., Smith, C. M., Belz, G. T.,
van Lint, A. L., Wakim, L. M.,Heath,
W., et al. (2003). Epidermal viral
immunity induced by CD8alpha+
dendritic cells but not by Langerhans
cells. Science 301, 1925–1928.
Andrews, D. M., Andoniou, C. E.,
Granucci, F., Ricciardi-Castagnoli,
P., and Degli-Esposti, M. A. (2001).
Infection of dendritic cells by
murine cytomegalovirus induces
functional paralysis. Nat. Immunol.
2, 1077–1084.
Bedoui, S., Whitney,P. G., Waithman, J.,
Eidsmo, L., Wakim, L., Caminschi,
I., et al. (2009). Cross-presentation
of viral and self antigens by skin-
derived CD103+dendritic cells.
Nat. Immunol. 10, 488–495.
Belz, G. T., Shortman, K., Bevan,
M. J., and Heath, W. R. (2005).
CD8alpha+dendritic cells selec-
tively present MHC class I-restricted
noncytolytic viral and intracellu-
lar bacterial antigens in vivo. J.
Immunol. 175, 196–200.
Belz, G. T., Smith, C. M., Eich-
ner, D., Shortman, K., Karupiah,
G., Carbone, F. R., et al. (2004a).
Cutting edge: conventional CD8
alpha+dendritic cells are generally
involved in priming CTL immu-
nity to viruses. J. Immunol. 172,
1996–2000.
Belz, G. T., Smith, C. M., Kleinert, L.,
Reading, P., Brooks, A., Shortman,
K., et al. (2004b). Distinct migrat-
ing and nonmigrating dendritic cell
populations are involved in MHC
class I-restricted antigen presenta-
tion after lung infection with virus.
Proc. Natl. Acad. Sci. U.S.A. 101,
8670–8675.
Benedict, C. A., Loewendorf, A., Gar-
cia, Z., Blazar, B. R., and Janssen, E.
M. (2008). Dendritic cell program-
ming by cytomegalovirus stunts
naive T cell responses via the PD-
L1/PD-1 pathway. J. Immunol. 180,
4836–4847.
Coffin, R. S., Thomas, S. K., Thomas, N.
S., Lilley,C. E.,Pizzey,A. R.,Griffiths,
C. H., et al. (1998). Pure populations
of transduced primary human cells
can be produced using GFP express-
ing herpes virus vectors and flow
cytometry. Gene Ther. 5, 718–722.
den Haan, J. M., Lehar, S. M., and
Bevan, M. J. (2000). CD8(+) but not
CD8() dendritic cells cross-prime
cytotoxic T cells in vivo. J. Exp. Med.
192, 1685–1696.
Edelson, B. T., Kc, W., Juang, R.,
Kohyama, M., Benoit, L. A.,
Klekotka, P. A., et al. (2010). Periph-
eral CD103+dendritic cells form
a unified subset developmentally
related to CD8alpha+conventional
dendritic cells. J. Exp. Med. 207,
823–836.
Engelmayer, J., Larsson, M., Subklewe,
M., Chahroudi, A., Cox, W. I., Stein-
man, R. M., et al. (1999). Vac-
cinia virus inhibits the maturation
of human dendritic cells: a novel
mechanism of immune evasion. J.
Immunol. 163, 6762–6768.
Gold, M. C., Munks, M. W., Wag-
ner, M., McMahon, C. W., Kelly,
A., Kavanagh, D. G., et al. (2004).
Murine cytomegalovirus interfer-
ence with antigen presentation has
little effect on the size or the effec-
tor memory phenotype of the CD8
T cell response. J. Immunol. 172,
6944–6953.
Hardwicke, M. A., and Sandri-Goldin,
R. M. (1994). The herpes simplex
virus regulatory protein ICP27 con-
tributes to the decrease in cellu-
lar mRNA levels during infection. J.
Virol. 68, 4797–4810.
Hardy, W. R., and Sandri-Goldin, R.
M. (1994). Herpes simplex virus
inhibits host cell splicing, and regu-
latory protein ICP27 is required for
this effect. J. Virol. 68, 7790–7799.
Heath, W. R., and Carbone, F. R. (2001).
Cross-presentation in viral immu-
nity and self-tolerance. Nat. Rev.
Immunol. 1, 126–134.
Hickman, H. D., Takeda, K., Skon, C.
N., Murray, F. R., Hensley, S. E.,
Loomis, J.,et al. (2008). Direct prim-
ing of antiviral CD8+T cells in
the peripheral interfollicular region
of lymph nodes. Nat. Immunol. 9,
155–165.
Hildner, K., Edelson, B. T., Purtha, W.
E., Diamond, M., Matsushita, H.,
Kohyama, M., et al. (2008). Batf3
deficiency reveals a critical role for
CD8alpha+dendritic cells in cyto-
toxic T cell immunity. Science 322,
1097–1100.
Frontiers in Immunology | Antigen Presenting Cell Biology November 2012 | Volume 3 | Article 348 | 8
Nopora et al. Cross-priming enhances generation of virus-specific CTL
Hill, A., Jugovic, P., York, I., Russ, G.,
Bennink, J., Yewdell, J., et al. (1995).
Herpes simplex virus turns off the
TAP toe vade host immunity. Nature
375, 411–415.
Hill, A. B., Barnett, B. C., McMichael,
A. J., and McGeoch, D. J. (1994).
HLA class I molecules are not trans-
ported to the cell surface in cells
infected with herpes simplex virus
types 1 and 2. J. Immunol. 152,
2736–2741.
Hinkley, S., Ambagala, A. P., Jones, C.
J., and Srikumaran, S. (2000). Avhs-
like activity of bovine herpesvirus-1.
Arch. Virol. 145, 2027–2046.
Iyoda, T., Shimoyama, S., Liu, K.,
Omatsu,Y.,Akiyama, Y.,Maeda, Y., et
al. (2002). The CD8+dendritic cell
subset selectively endocytoses dying
cells in culture and in vivo. J. Exp.
Med. 195, 1289–1302.
Jirmo, A. C., Nagel, C. H., Bohnen,
C., Sodeik, B., and Behrens, G. M.
(2009). Contribution of direct and
cross-presentation to CTL immu-
nity against herpes simplex virus 1.
J. Immunol. 182, 283–292.
Kavanagh, D. G., Gold, M. C., Wag-
ner, M., Koszinowski, U. H., and
Hill, A. B. (2001). The multiple
immune-evasion genes of murine
cytomegalovirus are not redundant:
m4 and m152 inhibit antigen pre-
sentation in a complementary and
cooperative fashion. J. Exp. Med.
194, 967–978.
Kerksiek, K. M., Niedergang, F.,
Chavrier, P., Busch, D. H., and
Brocker, T. (2005). Selective Rac1
inhibition in dendritic cells dimin-
ishes apoptotic cell uptake and
cross-presentation in vivo. Blood
105, 742–749.
Kleijnen, M. F., Huppa, J. B., Lucin,
P., Mukherjee, S., Farrell, H., Camp-
bell, A. E., et al. (1997). A mouse
cytomegalovirus glycoprotein, gp34,
forms a complex with folded class
I MHC molecules in the ER which
is not retained but is transported
to the cell surface. EMBO J. 16,
685–694.
Koppers-Lalic, D., Rijsewijk, F. A., Ver-
schuren, S. B., van Gaans-Van den
Brink, J. A., Neisig, A., Ressing, M.
E., et al. (2001). The UL41-encoded
virion host shutoff (vhs) protein
and vhs-independent mechanisms
are responsible for down-regulation
of MHC class I molecules by bovine
herpesvirus 1. J. Gen. Virol. 82,
2071–2081.
Krisky, D. M., Marconi, P. C., Olig-
ino, T. J., Rouse, R. J., Fink, D. J.,
Cohen, J. B., et al. (1998). Devel-
opment of herpes simplex virus
replication-defective multigene
vectors for combination gene
therapy applications. Gene Ther. 5,
1517–1530.
Kruse, M., Rosorius, O., Kratzer, F.,
Stelz, G., Kuhnt, C., Schuler, G.,
et al. (2000). Mature dendritic
cells infected with herpes simplex
virus type 1 exhibit inhibited T-
cell stimulatory capacity. J. Virol. 74,
7127–7136.
Kurts, C., Miller, J. F., Subramaniam,
R. M., Carbone, F. R., and Heath,
W.R. (1998). MHC class I-restric ted
cross-presentation is biased towards
high dose Ag and those released dur-
ing cellular destruction. J. Exp. Med.
188, 409–414.
Lauterbach, H., Kerksiek, K. M., Busch,
D. H., Berto, E., Bozac, A., Mavro-
mara, P., et al. (2004). Protection
from bacterial infection by a sin-
gle vaccination with replication-
deficient mutant herpes simplex
virus type 1. J. Virol. 78, 4020–4028.
Leib, D. A., Harrison, T. E., Laslo, K.
M., Machalek, M. A., Moorman, N.
J., and Virgin, H. W. (1999). Inter-
ferons regulate the phenotype of
wild-type and mutant herpes sim-
plex viruses in vivo. J. Exp. Med. 189,
663–672.
Loewendorf, A., Kruger, C., Borst,
E. M., Wagner, M., Just, U., and
Messerle, M. (2004). Identification
of a mouse cytomegalovirus gene
selectively targeting CD86 expres-
sion on antigen-presenting cells. J.
Virol. 78, 13062–13071.
Luckashenak,N., Schroeder, S., Endt, K.,
Schmidt,D.,Mahnke, K., Bachmann,
M. F., et al. (2008). Constitutive
cross presentation of tissue antigens
by dendritic cells controls CD8+T
cell tolerance in vivo. Immunity 28,
521–532.
Lukens, M. V., Kruijsen, D., Coenjaerts,
F. E., Kimpen, J. L., and van Bleek,
G. M. (2009). Respiratory syncytial
virus-induced activation and migra-
tion of respiratory dendritic cells
and subsequent antigen presenta-
tion in the lung-draining lymph
node. J. Virol. 83, 7235–7243.
Mikloska, Z., Bosnjak, L., and Cun-
ningham, A. L. (2001). Immature
monocyte-derived dendritic cells are
productively infected with herpes
simplex virus type 1. J. Virol. 75,
5958–5964.
Mogensen,T. H., Melchjorsen, J.,Malm-
gaard, L., Casola, A., and Paludan,
S. R. (2004). Suppression of proin-
flammatory cytokine expression by
herpes simplex virus type 1. J. Virol.
78, 5883–5890.
Mueller, S. N., Heath, W., McLain, J.
D., Carbone, F. R., and Jones, C. M.
(2002). Characterization of two TCR
transgenic mouse lines specific for
herpes simplex virus. Immunol. Cell
Biol. 80, 156–163.
Munks, M. W., Pinto, A. K., Doom,
C. M., and Hill, A. B. (2007). Viral
interference with antigen presenta-
tion does not alter acute or chronic
CD8 T cell immunodominance in
murine cytomegalovirus infection. J.
Immunol. 178, 7235–7241.
Neuenhahn, M., Kerksiek, K. M.,
Nauerth, M., Suhre, M. H., Schie-
mann, M., Gebhardt, F. E., et al.
(2006). CD8alpha+dendritic cells
are required for efficient entry of
Listeria monocytogenes into the
Spleen. Immunity 25, 619–630.
Norbury, C. C., Princiotta, M. F., Bacik,
I., Brutkiewicz, R. R., Wood, P.,
Elliott, T., et al. (2001). Multi-
ple antigen-specific processing path-
ways for activating naive CD8+
T cells in vivo. J. Immunol. 166,
4355–4362.
Pircher, H., Burki, K., Lang, R., Hen-
gartner, H., and Zinkernagel, R. M.
(1989). Tolerance induction in dou-
ble specific T-cell receptor trans-
genic mice varies with antigen.
Nature 342, 559–561.
Prechtel, A. T., Turza, N. M., Kobelt,
D. J., Eisemann, J. I., Coffin, R. S.,
McGrath, Y., et al. (2005). Infection
of mature dendritic cells with her-
pes simplex virus type 1 dramati-
cally reduces lymphoid chemokine-
mediated migration. J. Gen. Virol.86,
1645–1657.
Reusch, U., Muranyi, W., Lucin, P.,
Burgert, H. G., Hengel, H., and
Koszinowski, U. H. (1999). A
cytomegalovirus glycoprotein re-
routes MHC class I complexes to
lysosomes for degradation. EMBO J.
18, 1081–1091.
Rice, S. A., Long, M. C., Lam, V., Schaf-
fer, P. A., and Spencer, C. A. (1995).
Herpes simplex virus immediate-
early protein ICP22 is required for
viral modification of host RNA poly-
merase II and establishment of the
normal viral transcription program.
J. Virol. 69, 5550–5559.
Salio, M., Cella, M., Suter, M., and Lan-
zavecchia, A. (1999). Inhibition of
dendritic cell maturation by herpes
simplex virus. Eur. J. Immunol. 29,
3245–3253.
Samady, L., Costigliola, E., MacCor-
mac, L., McGrath, Y., Cleverley, S.,
Lilley, C. E., et al. (2003). Dele-
tion of the virion host shutoff pro-
tein (vhs) from herpes simplex virus
(HSV) relieves the viral block to
dendritic cell activation: potential of
vhs-HSV vectors for dendritic cell-
mediated immunotherapy. J. Virol.
77, 3768–3776.
Shortman, K., and Heath, W. R. (2010).
The CD8+dendritic cell subset.
Immunol. Rev. 234, 18–31.
Sigal, L. J., Crotty, S., Andino, R.,
and Rock, K. L. (1999). Cytotoxic
T-cell immunity to virus-infected
non-haematopoietic cells requires
presentation of exogenous antigen.
Nature 398, 77–80.
Smiley, J. R. (2004). Herpes simplex
virus virion host shutoff protein:
immune evasion mediated by a viral
RNase? J. Virol. 78, 1063–1068.
Smith, C. M., Belz, G. T., Wilson,
N. S., Villadangos, J. A., Short-
man, K., Carbone, F. R., et al.
(2003). Cutting edge: conventional
CD8 alpha+dendritic cells are pref-
erentially involved in CTL priming
after footpad infection with herpes
simplex virus-1. J. Immunol. 170,
4437–4440.
Snyder, C. M., Allan, J. E., Bonnett, E. L.,
Doom, C. M., and Hill,A. B. (2010).
Cross-presentation of a spread-
defective MCMV is sufficient to
prime the majority of virus-specific
CD8+T cells. PLoS ONE 5, e9681.
doi:10.1371/journal.pone.0009681
Song, B., Yeh, K. C., Liu, J., and Knipe,
D. M. (2001). Herpes simplex virus
gene products required for viral
inhibition of expression of G1-phase
functions. Virology 290, 320–328.
Spencer,C. A., Dahmus, M. E., and Rice,
S. A. (1997). Repression of host RNA
polymerase II transcription by her-
pes simplex virus type 1. J. Virol. 71,
2031–2040.
Strelow, L. I., and Leib, D. A. (1996).
Analysis of conserved domains of
UL41 of herpes simplex virus type
1 in virion host shutoff and patho-
genesis. J. Virol. 70, 5665–5667.
Tigges, M. A., Leng, S., Johnson, D. C.,
and Burke, R. L. (1996). Human
herpes simplex virus (HSV)-specific
CD8+CTL clones recognize HSV-
2-infected fibroblasts after treatment
with IFN-gamma or when virion
host shutoff functions are disabled.
J. Immunol. 156, 3901–3910.
Torti, N., Walton, S. M., Murphy, K.
M., and Oxenius, A. (2011). Batf3
transcription factor-dependent DC
subsets in murine CMV infection:
differential impact on T-cell prim-
ing and memory inflation. Eur. J.
Immunol. 41, 2612–2618.
Tortorella, D., Gewurz, B. E., Furman,
M. H., Schust, D. J., and Ploegh,
H. L. (2000). Viral subversion of
the immune system. Annu. Rev.
Immunol. 18, 861–926.
Wagner, M., Gutermann, A., Podlech, J.,
Reddehase, M. J., and Koszinowski,
U. H. (2002). Major histocompati-
bility complex class I allele-specific
www.frontiersin.org November 2012 | Volume 3 | Article 348 | 9
Nopora et al. Cross-priming enhances generation of virus-specific CTL
cooperative and competitive inter-
actions between immune evasion
proteins of cytomegalovirus. J. Exp.
Med. 196, 805–816.
Wilson, N. S., Behrens, G. M., Lundie,
R. J., Smith, C. M., Waithman, J.,
Young, L., et al. (2006). Systemicacti-
vation of dendritic cells by Toll-like
receptor ligands or malaria infec-
tion impairs cross-presentation and
antiviral immunity. Nat.Immunol. 7,
165–172.
Yewdell, J. W., and Hill, A. B. (2002).
Viral interference with antigen
presentation. Nat. Immunol. 3,
1019–1025.
Ziegler,H., Thale, R., Lucin, P.,Muranyi,
W., Flohr, T., Hengel, H., et al.
(1997). A mouse cytomegalovirus
glycoprotein retains MHC class
I complexes in the ERGIC/cis-
Golgi compartments. Immunity 6,
57–66.
Conflict of Interest Statement: The
authors declare that the research was
conducted in the absence of any
commercial or financial relationships
that could be construed as a potential
conflict of interest.
Received: 24 October 2012; paper pend-
ing published: 31 October 2012; accepted:
05 November 2012; published online: 26
November 2012.
Citation: Nopora K, Bernhard CA, Ried
C, Castello AA, Murphy KM, Mar-
coni P, Koszinowski U and Brocker T
(2012) MHC class I cross-presentation by
dendritic cells counteracts viral immune
evasion. Front. Immun. 3:348. doi:
10.3389/fimmu.2012.00348
This article was submitted to Frontiers
in Antigen Presenting Cell Biology, a
specialty of Frontiers in Immunology.
Copyright © 2012 Nopora, Bernhard,
Ried, Castello, Murphy, Marconi, Koszi-
nowski and Brocker . This is an open-
access article distributed under the terms
of the Creative Commons Attribution
License, which permits use, distribution
and reproduction in other forums, pro-
vided the original authors and source
are credited and subject to any copy-
right notices concerning any third-party
graphics etc.
Frontiers in Immunology | Antigen Presenting Cell Biology November 2012 | Volume 3 | Article 348 | 10
... Since vRAPs have been shown to be functional in pAPCs after hCMV and mCMV infection (34,35,(53)(54)(55), it has been hypothesized that cross-presentation is the major pathway of antigen presentation leading to effective CD8 T-cell priming by counteracting viral immune evasion (56)(57)(58). Consistent with this, MHC-I-negative fibroblasts infected with a spread-deficient mCMV, which prevents direct presentation in the first round and precludes further rounds of infection, have been shown to activate mCMV-specific CD8 T cells after immunization of WT mice (59). ...
... On the other hand, there is evidence that priming by crosspresentation does not exclude a role for direct presentation. While Batf3 -/mice completely lack CD8 + DCs, which could contribute also to direct priming, infection of CD11c-Rac mice, which are selectively defective in the uptake of exogenous antigens by DCs, showed an mCMV-specific priming comparable to that in WT mice (57). In analogy, mCMV-specific priming was not impaired in mice treated with the TLR9 agonist CpG to prevent cross-presentation (61). ...
... It was our original aim to identify the canonical pathway of viral antigen presentation to CD8 T cells, that is, to decide between direct antigen presentation and cross-presentation. Previous studies in mouse models of CMV infection have shown that mice can mount a virus-specific CD8 T-cell response as a "plan B" by either pathway when the respective other pathway is closed genetically or experimentally (57,(59)(60)(61)111). Specifically, as also shown here, Unc93b1 3d/3d mice genetically deficient in cross-presentation can perfectly develop a CD8 T-cell response by direct antigen presentation. ...
Article
Full-text available
CD8 T cells are important antiviral effectors in the adaptive immune response to cytomegaloviruses (CMV). Naïve CD8 T cells can be primed by professional antigen-presenting cells (pAPCs) alternatively by “direct antigen presentation” or “antigen cross-presentation”. In the case of direct antigen presentation, viral proteins are expressed in infected pAPCs and enter the classical MHC class-I (MHC-I) pathway of antigen processing and presentation of antigenic peptides. In the alternative pathway of antigen cross-presentation, viral antigenic material derived from infected cells of principally any cell type is taken up by uninfected pAPCs and eventually also fed into the MHC class-I pathway. A fundamental difference, which can be used to distinguish between these two mechanisms, is the fact that viral immune evasion proteins that interfere with the cell surface trafficking of peptide-loaded MHC-I (pMHC-I) complexes are absent in cross-presenting uninfected pAPCs. Murine cytomegalovirus (mCMV) models designed to disrupt either of the two presentation pathways revealed that both are possible in principle and can substitute each other. Overall, however, the majority of evidence has led to current opinion favoring cross-presentation as the canonical pathway. To study priming in the normal host genetically competent in both antigen presentation pathways, we took the novel approach of enhancing or inhibiting direct antigen presentation by using recombinant viruses lacking or overexpressing a key mCMV immune evasion protein. Against any prediction, the strongest CD8 T-cell response was elicited under the condition of intermediate direct antigen presentation, as it exists for wild-type virus, whereas the extremes of enhanced or inhibited direct antigen presentation resulted in an identical and weaker response. Our findings are explained by direct antigen presentation combined with a negative feedback regulation exerted by the newly primed antiviral effector CD8 T cells. This insight sheds a completely new light on the acquisition of viral immune evasion genes during virus-host co-evolution.
... This view seems to be corroborated by the molecular explanation that the virus interferes with direct antigen presentation by expressing immune evasion proteins, which inhibit the transport of recently-loaded pMHC-I complexes to the cell surface and thereby prevent recognition by virus-specific CD8 T cells [(39), reviewed in (40)]. In line with this, it has been shown that antigen cross-presentation by uninfected DCs can counteract viral immune evasion (41). Furthermore, high virus production at an early stage after HCT, when CD8 T-cell reconstitution is at its beginning and not yet sufficient to prevent viral spread, should provide large amounts of viral antigens to supply the crosspresentation pathway and thereby aid cross-priming. ...
Article
Full-text available
Cytomegalovirus (CMV) infection is the most critical infectious complication in recipients of hematopoietic cell transplantation (HCT) in the period between a therapeutic hematoablative treatment and the hematopoietic reconstitution of the immune system. Clinical investigation as well as the mouse model of experimental HCT have consistently shown that timely reconstitution of antiviral CD8 T cells is critical for preventing CMV disease in HCT recipients. Reconstitution of cells of the T-cell lineage generates naïve CD8 T cells with random specificities among which CMV-specific cells need to be primed by presentation of viral antigen for antigen-specific clonal expansion and generation of protective antiviral effector CD8 T cells. For CD8 T-cell priming two pathways are discussed: “direct antigen presentation” by infected professional antigen-presenting cells (pAPCs) and “antigen cross-presentation” by uninfected pAPCs that take up antigenic material derived from infected tissue cells. Current view in CMV immunology favors the cross-priming hypothesis with the argument that viral immune evasion proteins, known to interfere with the MHC class-I pathway of direct antigen presentation by infected cells, would inhibit the CD8 T-cell response. While the mode of antigen presentation in the mouse model of CMV infection has been studied in the immunocompetent host under genetic or experimental conditions excluding either pathway of antigen presentation, we are not aware of any study addressing the medically relevant question of how newly generated naïve CD8 T cells become primed in the phase of lympho-hematopoietic reconstitution after HCT. Here we used the well- established mouse model of experimental HCT and infection with murine CMV (mCMV) and pursued the recently described approach of up- or down-modulating direct antigen presentation by using recombinant viruses lacking or overexpressing the central immune evasion protein m152 of mCMV, respectively. Our data reveal that the magnitude of the CD8 T-cell response directly reflects the level of direct antigen presentation.
... Although CD8 + T cells can be cross-primed by DCs on many occasions, like viral infections, [24] CD4 + T cells are crucial to support long-lasting, effective cytotoxic and memory CD8 + T cell responses. [25] To explore the flexibility of the platform, murine tghCD40 BMDC or human moDCs were pulsed with a different set of pTag-Ag, UU60 and UU44. ...
Article
Full-text available
The agonistic potentials of therapeutic anti‐CD40 antibodies have been profiled in relation to antibody isotype and epitope specificity. Still, clinical impact relies on a well‐balanced clinical efficacy versus target‐mediated toxicity. As CD40‐mediated immune activation must rely on a combination of stimulation of antigen‐presenting cells (APCs) alongside antigen presentation, for efficient T cell priming, alternative approaches to improve the therapeutic outcome of CD40‐targeting strategies should focus on providing optimal antigen presentation together with CD40 stimulation. Herein, a bispecific antibody targeting CD40 as a means to deliver cargo (i.e., synthetic peptides) into APCs through a non‐covalent, high‐affinity interaction between the antibody and the cargo peptide, further referred to as the Adaptable Drug Affinity Conjugate (ADAC) technology, has been developed. The ADAC platform demonstrated a target‐specific CD4⁺ and CD8⁺ T cell expansion in vitro and significantly improved peptide‐specific CD8⁺ T cell proliferation in vivo. In addition, the strategy dramatically improved the in vitro and in vivo half‐life of the synthetic peptides. Future applications of ADAC involve pandemic preparedness to viral genetic drift as well as neoepitope vaccination strategies where the bispecific antibody is an off‐the‐shelf product, and the peptide antigen is synthesized based on next‐generation sequencing data mining.
... Coupled with CD4 + T-cell licensing, it is XCR1 + DCs that activate the subsequent CD8 + T-cell adaptive response. Indeed, many studies highlight the importance of cross-presenting DCs in peripheral HSV infections in both humans and mice [205][206][207][208][209][210][211][212][213][214]. Another important subset of DCs with a remarkable antiviral function is the pDCs. ...
Article
Full-text available
The ability of dendritic cells (DCs) to sense viral pathogens and orchestrate a proper immune response makes them one of the key players in antiviral immunity. Different DC subsets have complementing functions during viral infections, some specialize in antigen presentation and cross-presentation and others in the production of cytokines with antiviral activity, such as type I interferons. In this review, we summarize the latest updates concerning the role of DCs in viral infections, with particular focus on the complex interplay between DC subsets and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Despite being initiated by a vast array of immune receptors, DC-mediated antiviral responses often converge towards the same endpoint, that is the production of proinflammatory cytokines and the activation of an adaptive immune response. Nonetheless, the inherent migratory properties of DCs make them a double-edged sword and often viral recognition by DCs results in further viral dissemination. Here we illustrate these various aspects of the antiviral functions of DCs and also provide a brief overview of novel antiviral vaccination strategies based on DCs targeting.
... Batf3 −/− mice were the first in vivo models of cDC1 ablation, and they were useful in confirming the in vivo role of cDC1s in providing cross-presentation of various antigens of pathogens and tumor to CD8 T cells [5,[77][78][79][80][81][82][83][84][85][86][87][88][89][90]. In the original study, mice with Batf3 germline deficiency were examined [5] to ensure that different interpretations were possible. ...
Article
Full-text available
The clinical success of immune checkpoint therapy (ICT) has produced explosive growth in tumor immunology research because ICT was discovered through basic studies of immune regulation. Much of the current translational efforts are aimed at enhancing ICT by identifying therapeutic targets that synergize with CTLA4 or PD1/PD-L1 blockade and are solidly developed on the basis of currently accepted principles. Expanding these principles through continuous basic research may help broaden translational efforts. With this mindset, we focused this review on three threads of basic research directly relating to mechanisms underlying ICT. Specifically, this review covers three aspects of dendritic cell (DC) biology connected with antitumor immune responses but are not specifically oriented toward therapeutic use. First, we review recent advances in the development of the cDC1 subset of DCs, identifying important features distinguishing these cells from other types of DCs. Second, we review the antigen-processing pathway called cross-presentation, which was discovered in the mid-1970s and remains an enigma. This pathway serves an essential in vivo function unique to cDC1s and may be both a physiologic bottleneck and therapeutic target. Finally, we review the longstanding field of helper cells and the related area of DC licensing, in which CD4 T cells influence the strength or quality of CD8 T cell responses. Each topic is connected with ICT in some manner but is also a fundamental aspect of cell-mediated immunity directed toward intracellular pathogens.
... On the other hand, the host counteracts these strategies and mounts an adaptive antiviral immune response, mirrored by the generation of antigen-specific CD4 + and CD8 + T lymphocytes as well as the production of antibodies [3,[225][226][227]. Regarding HSV and HCMV infections, DC-dependent cross-presentation of viral antigens represents, among others, a crucial way to induce cytotoxic T lymphocyte (CTL) responses [228][229][230][231]. Moreover, there is strong evidence that bystander migratory submucosal and lymph node-resident DCs sequentially (cross-) present HSV-derived antigens in the lymph node. ...
Chapter
Full-text available
In the last decades, a multitude of distinct herpesvirus-mediated immune evasion mechanisms targeting dendritic cell (DC) biology were uncovered. Within this chapter, we summarize the current knowledge how herpesviruses, especially the α-herpesviruses HSV-1, HSV-2, varicella-zoster virus (VZV), and the β-herpesvirus HCMV, shape and exploit the function of myeloid DCs in order to hamper the induction of potent antiviral immune responses. In particular, the main topics covering herpesvirus-mediated immune evasion will involve: (i) the modulation of immature DC (iDC) phenotype, (ii) modulation of iDC apoptosis, (iii) the inhibition of DC maturation, (iv) degradation of the immune-modulatory molecule CD83 in mature DCs (mDCs), (v) interference with the negative regulator of β2 integrin activity, cytohesin-1 interaction partner (CYTIP), (vi) resulting in modulation of adhesion and migration of mDCs, (vii) autophagic degradation of lamins to support productive HSV-1 replication in iDCs, (viii) the release of uninfectious L-particles with immune-modulatory potential from HSV-1-infected mDCs, and (ix) the implications of DC subversion regarding T lymphocyte activation.
Preprint
Full-text available
CD8 T cells are the predominant effector cells of adaptive immunity in preventing cytomegalovirus (CMV) multiple-organ disease caused by cytopathogenic tissue infection. The mechanism by which CMV-specific, naïve CD8 T cells become primed and clonally expand is of fundamental importance for our understanding of CMV immune control. For CD8 T-cell priming, two pathways have been identified: direct antigen presentation by infected professional antigen-presenting cells (pAPCs) and antigen cross-presentation by uninfected pAPCs that take up antigenic material derived from infected tissue cells. Studies in mouse models using murine CMV (mCMV) and precluding either pathway genetically or experimentally have shown that, in principle, both pathways can congruently generate the mouse MHC/H-2 class-I-determined epitope-specificity spectrum of the CD8 T-cell response. Own recent studies, however, have shown that direct antigen presentation is the canonical pathway when both are accessible. This raised the question of why antigen cross-presentation is ineffective even under conditions of high virus replication thought to provide high amounts of antigenic material for feeding cross-presenting pAPCs. As delivery of antigenic material for cross-presentation is associated with programmed cell death, and as CMVs encode inhibitors of different cell death pathways, we pursued the idea that these inhibitors restrict antigen delivery and thus CD8 T-cell priming by cross-presentation. To test this hypothesis, we compared the CD8 T-cell responses to recombinant mCMVs lacking expression of the apoptosis-inhibiting protein M36 or the necroptosis-inhibiting protein M45 with responses to wild-type mCMV and revertant viruses expressing the respective cell death inhibitors. The data reveal that increased programmed cell death caused by deletion of either M36 or M45 improves CD8 T-cell priming in mice capable of antigen cross-presentation but not in a mutant mouse strain unable to cross-present. These findings strongly support the conclusion that CMV cell death inhibitors restrict the priming of CD8 T cells by antigen cross-presentation. Author Summary In patients as well as in experimental mouse models, CD8 T cells represent the most potent antiviral effector cells in preventing CMV disease in immunocompromised recipients of hematopoietic cell transplantation. Despite the clinical relevance of mounting a protective response, the mode of CMV antigen presentation to naïve CD8 T cells remained unclear. In principle, naïve CD8 T cells can be sensitized through “direct antigen presentation” on the surface of infected professional antigen-presenting cells (pAPCs) or through “antigen cross-presentation” by uninfected pAPCs that take up antigenic material derived from infected cells following cell death. As CMVs are cytopathogenic, eventually killing their host cells, and as they encode immune evasion proteins interfering with the MHC/HLA class-I pathway of direct antigen presentation in infected cells, it was reasonable to propose a dominant role for antigen cross-presentation. Mouse models precluding either pathway, however, revealed that both can raise an equivalent CD8 T-cell response, and recent work has identified direct antigen presentation as the canonical pathway taken when both are accessible. Here we show that virus-encoded inhibitors of two programmed cell death modalities, apoptosis and necroptosis, prevent an antigen release sufficient for a notable cross-presentation. This answers a long-debated open question in CMV immunology.
Chapter
Dendritic cells perform critical functions in bridging innate and adaptive immunity. Their ability to sense adjuvant signals in their environment, migrate on maturation, and cross-present cell-associated antigens enables these cells to carry antigen from tissue sites to lymph nodes, and thereby prime naïve T cells that cannot enter tissues. Despite being an infrequent cell type in tumors, we discuss how dendritic cells impact the immune environment of tumors and their response to cancer therapies. We review how radiation therapy of tumors can impact dendritic cells, through transfer of cell associated antigens to dendritic cells and the release of endogenous adjuvants, resulting in increased antigen presentation in the tumor-draining lymph nodes. We explore how tumor specific factors can result in negative regulation of dendritic cell function in the tumor, and the impact of direct radiation exposure to dendritic cells in the treatment field. These data suggest an important role for dendritic cell subpopulations in activating new T cell responses and boosting existing T cell responses to tumor associated antigens in tumor draining lymph nodes following radiation therapy. It further justifies a focus on the needs of the lymph node T cells to improve systemic anti-immunity following radiation therapy.
Chapter
Dendritic cells are cells of hematopoietic origin that are specialized in antigen presentation and instruction of innate and adaptive immune responses. They are a heterogenous group of cells populating lymphoid organs and most tissues. Dendritic cells are commonly separated in three main subsets that differ in their developmental paths, phenotype, and functions. Most studies on dendritic cells were done primarily in mice; therefore, in this chapter, we propose to summarize the current knowledge and recent progress on mouse dendritic cell subsets’ development, phenotype, and functions.
Article
Full-text available
CD40 signaling in classical type 1 dendritic cells (cDC1s) is required for CD8 T cell-mediated tumor rejection, but the underlying mechanisms are incompletely understood. Here, we identified CD40-induced genes in cDC1s, including Cd70, Tnfsf9, Ptgs2 and Bcl2l1, and examined their contributions to anti-tumor immunity. cDC1-specific inactivation of CD70 and COX-2, and global CD27 inactivation, only partially impaired tumor rejection or tumor-specific CD8 T cell expansion. Loss of 4-1BB, alone or in Cd27−/− mice, did not further impair anti-tumor immunity. However, cDC1-specific CD40 inactivation reduced cDC1 mitochondrial transmembrane potential and increased caspase activation in tumor-draining lymph nodes, reducing migratory cDC1 numbers in vivo. Similar impairments occurred during in vitro antigen presentation by Cd40−/− cDC1s to CD8⁺ T cells, which were reversed by re-expression of Bcl2l1. Thus, CD40 signaling in cDC1s not only induces costimulatory ligands for CD8⁺ T cells but also induces Bcl2l1 that sustains cDC1 survival during priming of anti-tumor responses.
Article
Full-text available
Both human cytomegaloviruses (HCMVs) and murine cytomegaloviruses (MCMVs) encode multiple genes that interfere with antigen presentation by major histocompatibility complex (MHC) class I, and thus protect infected targets from lysis by virus-specific cytotoxic T lymphocytes (CTLs). HCMV has been shown to encode four such genes and MCMV to encode two. MCMV m152 blocks the export of class I from a pre-Golgi compartment, and MCMV m6 directs class I to the lysosome for degradation. A third MCMV gene, m4, encodes a glycoprotein which is expressed at the cell surface in association with class I. Here we here show that m4 is a CTL-evasion gene which, unlike previously described immune-evasion genes, inhibited CTLs without blocking class I surface expression. m152 was necessary to block antigen presentation to both Kb- and Db-restricted CTL clones, while m4 was necessary to block presentation only to Kb-restricted clones. m152 caused complete retention of Db, but only partial retention of Kb, in a pre-Golgi compartment. Thus, while m152 effectively inhibited Db-restricted CTLs, m4 was required to completely inhibit Kb-restricted CTLs. We propose that cytomegaloviruses encode multiple immune-evasion genes in order to cope with the diversity of class I molecules in outbred host populations.
Article
Full-text available
Although CD103-expressing dendritic cells (DCs) are widely present in nonlymphoid tissues, the transcription factors controlling their development and their relationship to other DC subsets remain unclear. Mice lacking the transcription factor Batf3 have a defect in the development of CD8alpha+ conventional DCs (cDCs) within lymphoid tissues. We demonstrate that Batf3(-/-) mice also lack CD103+CD11b- DCs in the lung, intestine, mesenteric lymph nodes (MLNs), dermis, and skin-draining lymph nodes. Notably, Batf3(-/-) mice displayed reduced priming of CD8 T cells after pulmonary Sendai virus infection, with increased pulmonary inflammation. In the MLNs and intestine, Batf3 deficiency resulted in the specific lack of CD103+CD11b- DCs, with the population of CD103+CD11b+ DCs remaining intact. Batf3(-/-) mice showed no evidence of spontaneous gastrointestinal inflammation and had a normal contact hypersensitivity (CHS) response, despite previous suggestions that CD103+ DCs were required for immune homeostasis in the gut and CHS. The relationship between CD8alpha+ cDCs and nonlymphoid CD103+ DCs implied by their shared dependence on Batf3 was further supported by similar patterns of gene expression and their shared developmental dependence on the transcription factor Irf8. These data provide evidence for a developmental relationship between lymphoid organ-resident CD8alpha+ cDCs and nonlymphoid CD103+ DCs.
Article
Full-text available
CD8+ T cells can be primed by peptides derived from endogenous proteins (direct presentation), or exogenously acquired protein (cross-presentation). However, the relative ability of these two pathways to prime CD8+ T cells during a viral infection remains controversial. Cytomegaloviruses (CMVs) can infect professional antigen presenting cells (APCs), including dendritic cells, thus providing peptides for direct presentation. However, the viral immune evasion genes profoundly impair recognition of infected cells by CD8+ T cells. Nevertheless, CMV infection elicits a very strong CD8+ T cell response, prompting its recent use as a vaccine vector. We have shown previously that deleting the immune evasion genes from murine cytomegalovirus (MCMV) that target class I MHC presentation, has no impact on the size or breadth of the CD8+ T cell response elicited by infection, suggesting that the majority of MCMV-specific CD8+ T cells in vivo are not directly primed by infected professional APCs. Here we use a novel spread-defective mutant of MCMV, lacking the essential glycoprotein gL, to show that cross-presentation alone can account for the majority of MCMV-specific CD8+ T cell responses to the virus. Our data support the conclusion that cross-presentation is the primary mode of antigen presentation by which CD8+ T cells are primed during MCMV infection.
Article
Full-text available
In the respiratory tract, different dendritic cell (DC) populations guard a tight balance between tolerance and immunity to infectious or harmless materials to which the airways are continuously exposed. For infectious and noninfectious antigens administered via different routes, different subsets of DC might contribute during the induction of T-cell tolerance and immunity. We studied the impact of primary respiratory syncytial virus (RSV) infection on respiratory DC composition in C57BL/6 mice. We also tracked the migration of respiratory DC to the lymph nodes and studied antigen presentation by lung-derived and lymph node-resident DC to CD4+ and CD8+ T cells. We observed a massive influx of mainly CD103− CD11bhigh CD11c+ conventional DC (cDC) and plasmacytoid DC during the first 7 days of RSV infection, while CD103+ CD11blow CD11c+ cDC disappeared from the lung. The two major subsets of lung tissue DC, CD103+ CD11blow CD11c+ and CD103− CD11bhigh CD11c+ cDC, both transported RSV RNA to the lung-draining lymph node. Furthermore, these lung-derived cDC subsets as well as resident LN DC, which did not contain viral RNA, displayed viral antigen by major histocompatibility complex class I and class II to CD8+ and CD4+ T cells. Taken together, our data indicate that during RSV infections, at least three DC subsets might be involved during the activation of lymph node-homing naïve and memory CD4+ and CD8+ T cells.
Article
Full-text available
Skin-derived dendritic cells (DCs) include Langerhans cells, classical dermal DCs and a langerin-positive CD103(+) dermal subset. We examined their involvement in the presentation of skin-associated viral and self antigens. Only the CD103(+) subset efficiently presented antigens of herpes simplex virus type 1 to naive CD8(+) T cells, although all subsets presented these antigens to CD4(+) T cells. This showed that CD103(+) DCs were the migratory subset most efficient at processing viral antigens into the major histocompatibility complex class I pathway, potentially through cross-presentation. This was supported by data showing only CD103(+) DCs efficiently cross-presented skin-derived self antigens. This indicates CD103(+) DCs are the main migratory subtype able to cross-present viral and self antigens, which identifies another level of specialization for skin DCs.
Article
Full-text available
Dendritic cells (DC), which can be subdivided into different phenotypic and functional subsets, play a pivotal role in the generation of cytotoxic T cell immunity against viral infections. Understanding the modes of Ag acquisition, processing and presentation by DC is essential for the design of effective antiviral vaccines. We aimed to assess the contribution of direct vs cross-presentation for the induction of HSV1-specific CD8(+) T lymphocyte responses in mice. Using HSV1 strains expressing fluorescence proteins, we provide evidence for the ability of HSV1 to induce viral transcription. Using HSV1-wild-type as well as gB- or gH-deficient mutants to either directly inoculate DC or to infect target cells, which then were given to DC, we show that DC acquired viral Ag via phagocytosis of target cells and via direct inoculation of virus being released from target cells. Our study corroborates the function of the CD8(+) DC specialized in Ag cross-presentation and confirms this specific feature for Ags that these DC acquire directly from HSV1. However, although infection of cross-presenting DC amplified T cell responses, it was not a requirement for presentation of viral Ags, both in vitro and in vivo. Finally, we provide evidence that direct presentation did not contribute to the Ag presentation capacity of CD8(+) DC after phagocytosis of infected target cells. We conclude that cross-presentation is of major importance for the induction of CTL immunity in mice.
Article
HSV infection blocks G1 events in the cell cycle and arrests host cell growth in the G1 phase. To further define the mechanism of the effect and determine the viral gene product(s) responsible, we examined various mutant viruses for their effects on cell cycle regulatory proteins (pRb, cyclin D1, and cdk4) and on cell cycle progression into S phase. Unlike the wild-type virus, the ICP27 mutant virus was defective for blocking the phosphorylation of pRb proteins, and the normal pRb pattern was restored in cells infected with a rescued virus. The virion host shutoff (vhs) function, DNA replication, and late gene functions were not required for the virus-induced effects on pRb protein. BrdU incorporation in synchronized HSV-infected cells showed that ICP27 was required for blocking the cell cycle in the G1 phase. Furthermore, ICP27, ICP4, ICP0, and vhs were required for blocking the induction of the G1 cell cycle regulators cyclin D1 and cdk4 in HSV-infected cells. Both ICP27 and the vhs function contributed to the reduction of cyclin D1 mRNA levels in HSV-infected cells. These results provide evidence that HSV-1 ICP27 protein is essential for viral inhibition of G1-phase functions and that certain other HSV proteins are required for some of the viral effects on the cell cycle. Finally, these results show that HSV-1 ICP27 and vhs act jointly to reduce host mRNA levels in infected cells.
Article
Priming of CD8(+) T cells specific for viruses that interfere with the MHC class I presentation pathway is a challenge for the immune system and is believed to rely on cross-presentation. Cytomegalovirus (CMV) infection induces vigorous CD8(+) T-cell responses despite its potent immune evasion strategies. Furthermore, CD8(+) T cells specific for a subset of viral epitopes accumulate and are maintained at high levels exhibiting an activated phenotype - referred to as "inflationary T cells". Taking advantage Batf3(-/-) mice in which the development of cross-presenting CD8α(+) and CD103(+) DCs is severely compromised, we analyzed their role in the induction and inflation of murine (M)CMV-specific CD8(+) T-cell responses. We found that priming of MCMV-specific CD8(+) T cells was severely impaired in the absence of cross-presenting DCs. However, inflation of two immuno-dominant MCMV-specific CD8(+) T-cell populations was largely normal in the absence of cross-presenting DCs, indicating that inflation during latency was mainly dependent on direct antigen presentation. These results highlight differential antigen presentation requirements during acute and latent MCMV infection.
Article
Mouse lymphoid tissues contain a subset of dendritic cells (DCs) expressing CD8 alpha together with a pattern of other surface molecules that distinguishes them from other DCs. These molecules include particular Toll-like receptor and C-type lectin pattern recognition receptors. A similar DC subset, although lacking CD8 expression, exists in humans. The mouse CD8(+) DCs are non-migrating resident DCs derived from a precursor, distinct from monocytes, that continuously seeds the lymphoid organs from bone marrow. They differ in several key functions from their CD8(-) DC neighbors. They efficiently cross-present exogenous cell-bound and soluble antigens on major histocompatibility complex class I. On activation, they are major producers of interleukin-12 and stimulate inflammatory responses. In steady state, they have immune regulatory properties and help maintain tolerance to self-tissues. During infection with intracellular pathogens, they become major presenters of pathogen antigens, promoting CD8(+) T-cell responses to the invading pathogens. Targeting vaccine antigens to the CD8(+) DCs has proved an effective way to induce cytotoxic T lymphocytes and antibody responses.