ArticlePDF Available

CD8+ T Cell-Dependent Elimination of Dendritic Cells In Vivo Limits the Induction of Antitumor Immunity

Authors:
  • Flowjoanna

Abstract and Figures

The fate of dendritic cells (DC) after they have initiated a T cell immune response is still undefined. We have monitored the migration of DC labeled with a fluorescent tracer and injected s.c. into naive mice or into mice with an ongoing immune response. DC not loaded with Ag were detected in the draining lymph node in excess of 7 days after injection with maximum numbers detectable approximately 40 h after transfer. In contrast, DC that had been loaded with an MHC class I-binding peptide disappeared from the lymph node with kinetics that parallel the known kinetics of activation of CD8+ T cells to effector function. In the presence of high numbers of specific CTL precursors, as in TCR transgenic mice, DC numbers were significantly decreased by 72 h after injection. The rate of DC disappearance was extremely rapid and efficient in recently immunized mice and was slower in "memory" mice in which memory CD8+ cells needed to reacquire effector function before mediating DC elimination. We also show that CTL-mediated clearance of Ag-loaded DC has a notable effect on immune responses in vivo. Ag-specific CD8+ T cells failed to divide in response to Ag presented on a DC if the DC were targets of a pre-existing CTL response. The induction of antitumor immunity by tumor Ag-loaded DC was also impaired. Therefore, CTL-mediated clearance of Ag-loaded DC may serve as a negative feedback mechanism to limit the activity of DC within the lymph node.
Content may be subject to copyright.
RESEARCH ARTICLE | MARCH 15 2000
CD8
+
T Cell-Dependent Elimination of Dendritic Cells In Vivo Limits the
Induction of Antitumor Immunity
1
Ian F. Hermans; ... et. al
J Immunol (2000) 164 (6): 3095–3101.
https://doi.org/10.4049/jimmunol.164.6.3095
Related Content
Impaired Ability of MHC Class II−/− Dendritic Cells to Provide Tumor Protection is Rescued by CD40 Ligation
J Immunol (July,1999)
Tumor-Specic Tc1, But Not Tc2, Cells Deliver Protective Antitumor Immunity
J Immunol (December,2001)
Downloaded from http://journals.aai.org/jimmunol/article-pdf/164/6/3095/1115875/3095.pdf by guest on 21 July 2023
CD8
T Cell-Dependent Elimination of Dendritic Cells In Vivo
Limits the Induction of Antitumor Immunity
1
Ian F. Hermans, David S. Ritchie, Jianping Yang, Joanna M. Roberts, and Franca Ronchese
2
The fate of dendritic cells (DC) after they have initiated a T cell immune response is still undefined. We have monitored the
migration of DC labeled with a fluorescent tracer and injected s.c. into naive mice or into mice with an ongoing immune response.
DC not loaded with Ag were detected in the draining lymph node in excess of 7 days after injection with maximum numbers
detectable 40 h after transfer. In contrast, DC that had been loaded with an MHC class I-binding peptide disappeared from the
lymph node with kinetics that parallel the known kinetics of activation of CD8
T cells to effector function. In the presence of high
numbers of specific CTL precursors, as in TCR transgenic mice, DC numbers were significantly decreased by 72 h after injection.
The rate of DC disappearance was extremely rapid and efficient in recently immunized mice and was slower in “memory” mice
in which memory CD8
cells needed to reacquire effector function before mediating DC elimination. We also show that CTL-
mediated clearance of Ag-loaded DC has a notable effect on immune responses in vivo. Ag-specific CD8
T cells failed to divide
in response to Ag presented on a DC if the DC were targets of a pre-existing CTL response. The induction of antitumor immunity
by tumor Ag-loaded DC was also impaired. Therefore, CTL-mediated clearance of Ag-loaded DC may serve as a negative feedback
mechanism to limit the activity of DC within the lymph node. The Journal of Immunology, 2000, 164: 3095–3101.
Dendritic cells (DC)
3
are highly specialized APCs that
serve a sentinel function in immunity by internalizing
and processing peripheral Ags for presentation to naive
T cells (1). DC residing in the peripheral tissues are initially of an
“immature” phenotype characterized by a high propensity for Ag
capture but relatively poor ability to stimulate naive T cells (2).
However, upon exposure to cytokines associated with inflamma-
tion or tissue injury such as TNF-
or IL-1
, DC migrate from the
peripheral tissues to regional lymph nodes (3, 4) and undergo a
maturation process characterized by a reduction in Ag capture
function and up-regulation of MHC and costimulatory molecules
(5, 6). Final DC activation is induced by Ag-specific Th cells via
the interaction of CD40-ligand on the Th cell with CD40 on the
DC (7, 8). Activated DC can prime naive CD8
T cells to CTL
(9–11), which have the capacity to recognize and kill cells that
express specific Ag in the peripheral tissues. The fate of DC after
they have reached the lymph node is unknown. Because no cells
with the morphology of DC can be demonstrated in efferent lymph
(12), it has been proposed that DC die in situ, although the mech-
anism remains unclear.
Previous studies have shown that DC injected s.c. or i.v. have
the capacity to migrate to the regional lymph nodes or the spleen,
respectively (13). Fluorescent-labeled DC injected s.c. have been
shown to migrate to draining lymph nodes (DLN) where they in-
teract with Ag-specific CD4
T cells to form clusters in the para-
cortex (14). DC that had not been loaded with specific Ag failed to
form such clusters. Surprisingly, it was also observed that the num-
bers of Ag-loaded DC in the DLN had declined by 48 h, whereas
the non-Ag-loaded DC persisted for longer periods of time. This
suggested that the Ag-loaded DC may have been eliminated by an
immune-mediated mechanism.
We wished to examine in more detail the fate of peripherally
administered DC in a model of CTL-mediated immunity and to
establish how the immune response affected the persistence of DC
in the lymph node. Our results suggest that DC clearance occurs in
the presence of previously activated CTL and also during the ac-
tivation of naive Ag-specific CTL precursors. Furthermore, the
rapid clearance of tumor Ag-loaded DC was associated with im-
paired capacity to induce tumor immunity. Therefore, CTL-medi-
ated clearance of Ag-loaded DC may serve as a negative feedback
mechanism to limit the activity of DC within the lymph node.
Materials and Methods
Mice
C57BL/6 mice were from breeding pairs originally obtained from The
Jackson Laboratory (Bar Harbor, ME). Strain 318 mice (15), transgenic for
a TCR specific for H-2 D
b
plus fragment 33–41 of the lymphocytic cho-
riomeningitis virus glycoprotein (LCMV
33–41
),
3
were kindly provided by
Dr. H. Pircher (Institute of Medical Microbiology and Hygiene, University
of Freiburg, Freiburg, Germany). The B6Aa
0
/Aa
0
MHC class II
/
mice
(16) were supplied by Dr. H. Bluethmann (Hoffmann-LaRoche, Basel,
Switzerland). All mice were maintained at the Biomedical Research Unit of
the Wellington School of Medicine by brother sister mating; in vivo
experimental protocols were approved by the Wellington School of Med-
icine Animal Ethics Committee and were performed according to institu-
tional guidelines.
Tumor cell line
LL-LCMV is a derivative of the Lewis lung carcinoma LLTC (C57BL/6,
H-2
b
) that has been modified to express a minigene encoding LCMV
33–41
under the control of a CMV promoter (17).
Malaghan Institute of Medical Research, Wellington School of Medicine, Wellington,
New Zealand
Received for publication July 6, 1999. Accepted for publication January 13, 2000.
The costs of publication of this article were defrayed in part by the payment of page
charges. This article must therefore be hereby marked advertisement in accordance
with 18 U.S.C. Section 1734 solely to indicate this fact.
1
This work was supported by grants from the Cancer Society of New Zealand, New
Zealand Cancer Institute, and Otago University Research Fund and by a generous
donation by Sir Roy McKenzie. F.R. is the recipient of a Wellington Medical Re-
search Foundation Malaghan Senior Fellowship.
2
Address correspondence and reprint requests to Dr. Franca Ronchese, Malaghan
Institute of Medical Research, P.O. Box 7060, Wellington South, New Zealand. E-
mail address: fronchese@malaghan.org.nz
3
Abbreviations used in this paper: DC, dendritic cell; DLN, draining lymph node;
LCMV, lymphocytic choriomeningitis virus; CM, complete medium; CFSE, carboxy-
fluorescein succinimidyl ester.
Copyright © 2000 by The American Association of Immunologists 0022-1767/00/$02.00
Downloaded from http://journals.aai.org/jimmunol/article-pdf/164/6/3095/1115875/3095.pdf by guest on 21 July 2023
In vitro culture media and reagents
Unless otherwise stated, all cultures were maintained in complete medium
(CM) comprising of IMDM with 2 mM glutamine, 1% penicillin-strepto-
mycin, 5 10
5
M 2-ME, and 5% FBS (all from Life Technologies,
Auckland, New Zealand). The synthetic peptides LCMV
33–41
(KAVYN
FATM) and OVA
257–264
(SIINFEKL) were from Chiron Mimotopes (Clay-
ton, Australia).
Culture of bone marrow-derived DC
Bone marrow cells from C57BL/6 mice or B6Aa
0
/Aa
0
mice were cultured
at 4 10
5
cells/ml in CM containing 20 ng/ml IL-4 and 20 ng/ml GM-CSF
as described previously (18). Cultures were provided fresh CM and cyto-
kines every 3 days and incubated at 37°C until the time of assay (68
days). Cultures typically contained 70–100% DC as determined by fluo-
rescent staining with the anti-CD11c Ab N418. DC (1 10
6
cells/ml) were
loaded with peptide Ag by incubation at 37°C in CM containing 10
M
synthetic peptide for 2 h and then were washed three times with IMDM to
remove excess peptide.
DC migration assay
DC were labeled with the fluorescent dye carboxyfluorescein succinimidyl
ester (CFSE; Molecular Probes, Eugene, OR) by incubation in PBS con-
taining 1
M CFSE for 10 min at 37°C before one wash in five volumes
of ice-cold PBS and two washes with IMDM. Unless otherwise stated,
mice were injected with 1 10
6
CFSE
DC by s.c. injection in the distal
forelimb (volar aspect). At the indicated times, both axillary and brachial
lymph nodes were removed from immunized mice and incubated in 2.4
mg/ml collagenase type II (Life Technologies) and 1 mg/ml DNase I (Sig-
ma, St. Louis, MO) for 90 min at 37°C. The tissue was then disrupted by
aspiration through an 18-gauge needle. The cell suspension was sieved
through gauze, washed in PBS, and resuspended for flow cytometric anal-
ysis in PBS containing 2% FBS and 0.01% sodium azide.
Lymph node suspensions were analyzed using a FACSort and CellQuest
software (both from Becton Dickinson, Mountain View, CA). The region
containing DC was identified on the basis of forward/side light scatter
profile. No CFSE
cells were found outside this region. In initial experi-
ments, the total number of DC within the DLN was calculated as follows:
% CFSE
cells in the DC region % of DLN cells in the DC region
number cells in DLN; data are expressed as average number of DC within
DLN for each experimental group. Because it was observed over repeated
experiments that the percentage of CFSE
cells in the DC region was
always proportional to the absolute number of CFSE
cells across the
experimental groups, data were expressed as average percentage of CFSE
cells thereafter. Only events falling within the gated DC region were col-
lected and stored (250,000 events for each DLN suspension).
Frozen sections (8
m) were prepared from the axillary lymph nodes of
animals injected with CFSE
DC 20 h earlier. The sections were stained
with hematoxylin and eosin and analyzed by standard and fluorescence
microscopy. CD8
T cells are depleted by one i.v. injection of 500
g
purified 2.43 mAb 24 h before DC immunization.
Ab staining
Ab staining was in PBS containing 2% FBS and 0.01% sodium azide. The
anti-Fc
RII mAb 2.4G2 was used at 10
g/ml to inhibit nonspecific stain-
ing. Anti-MHC class II (3JP) and anti-CD11c (N418) were affinity purified
from culture supernatants using protein G-Sepharose (Pharmacia Biotech,
Uppsala, Sweden), conjugated to biotin as described (19), and revealed
using streptavidin-PE (PharMingen, San Diego, CA). Instrument compen-
sation was set in each experiment using single color-stained samples.
Adoptive transfer and DC immunization
Pooled lymph node cell suspensions were prepared from strain 318 mice,
and the percentage of T cells expressing the transgenic TCR was deter-
mined on one sample of cells by staining with anti-V
2 and anti-V
8.1,
8.2 Abs and by FACS analysis. The cells were then labeled by incubation
in 1.25
M CFSE (Molecular Probes) in PBS for 10 min at room temper-
ature at a cell concentration of 2 10
7
cells/ml. The cells were pelleted by
centrifugation in the presence of 50% FCS, washed two times in CM, and
then washed again in IMDM. Suspensions containing 5 10
6
V
2
V
8
cells were transferred into C57BL/6 recipients by i.v. injection into the tail
vein. One day after adoptive transfer, the mice were immunized s.c. with
10
5
DC. After 66 h, the draining axillary and brachial lymph nodes were
removed, teased through gauze to prepare single-cell suspensions, and an-
alyzed by FACS.
Tumor immunity assay
Groups of C57BL/6 mice (n5) were immunized by s.c. injection into the
left flank with 10
5
DC. Secondary immunizations were by s.c. injection
into the contralateral flank 7 days later. One week after the last immuni-
zation, all animals were challenged with 10
6
LL-LCMV tumor cells in-
jected s.c. into the left flank. Mice were monitored for tumor growth every
3–4 days, and tumor size for each group was calculated as the mean of the
products of bisecting diameters (SE). Measurements were terminated for
each group when the first animal developed a tumor in excess of 200 mm
2
.
Results
CFSE
DC injected s.c. are detected in the DLN
We examined the migration of injected DC to the secondary lym-
phoid organs of mice. DC were cultured from bone marrow pre-
cursors in the presence of GM-CSF and IL-4 for 7 days and labeled
with CFSE, an amine-reactive dye that can be retained in live cells
for many generations. DC were administered s.c. into the distal
forelimb of mice without any deliberate preincubation with Ag. As
can be seen in Fig. 1, 20 h after injection CFSE
cells could be
detected in the DLN by fluorescence microscopy on frozen tissue
sections and were located in the paracortical region of the lymph
node. Analysis of DLN cell preparations by flow cytometry also
revealed the presence of CFSE
cells in the DLN (Fig. 2). No
CFSE
cells could be detected in the non-DLN. In addition,
CFSE
cells could not be detected in the DLN if the DC were heat
killed before injection. These results imply that the CFSE
cells
had reached the DLN via an active migratory process.
The possibility that the CFSE
cells found in the DLN were in
fact endogenous cells that had acquired CFSE from the injected
cells was examined by injecting MHC class II
/
DC and then
using MHC class II expression to distinguish the endogenous from
the injected DC. C57BL/6 mice were injected with CFSE
DC
cultured from either C57BL/6 mice or MHC class II
/
B6Aa
0
/
Aa
0
mice. MHC class II expression was then examined on CFSE
cells in the DLN. As shown in Fig. 2B, MHC class II expression
could be demonstrated on CFSE
cells obtained from mice that
had received C57BL/6 DC. In contrast, no expression of MHC
class II was found on CFSE
cells from mice that received MHC
class II
/
DC. We conclude that the CFSE
cells detected in the
DLN were the same DC that had been injected peripherally and
that these cells had actively migrated from the site of injection to
the DLN.
The proportion of CFSE
DC that reached the DLN and their
kinetics of appearance were also examined. As shown in Fig. 3A,
the absolute number of CFSE
DC found within the DLN repre-
sented only a small proportion (0.1%) of the original number of
DC injected. The injected DC appeared in the DLN within 12 h of
injection, reaching maximum numbers before 48 h. Considerable
numbers of CFSE
DC could still be demonstrated in the DLN at
172 h postinjection (Fig. 3B), the latest time point examined.
In summary, CFSE
DC can be demonstrated to actively mi-
grate from the site of s.c. injection to the DLN and to persist in
vivo in excess of 7 days from the time of injection.
DC loaded with MHC class I-binding peptide Ag are cleared
from the DLN
Next, we sought to determine the fate of Ag-loaded DC after s.c.
injection into mice. CFSE
DC were loaded with LCMV
33–41
,an
MHC class I-binding peptide from the LCMV glycoprotein, and
injected into the forelimbs of C57BL/6 recipients. The numbers of
Ag-loaded DC in the DLN were compared with the numbers in the
DLN of mice that received DC not loaded with Ag. As can be seen
in Fig. 4A, loading with Ag had no effect on the number of CFSE
DC recovered from the DLN at 16 h after injection. By 72 h, a
small reduction in the number of DC in the DLN was observed in
3096 CTL-MEDIATED ELIMINATION OF DC IN VIVO
Downloaded from http://journals.aai.org/jimmunol/article-pdf/164/6/3095/1115875/3095.pdf by guest on 21 July 2023
the experiment shown, but this reduction was not always observed
in repeated experiments. In contrast, when the number of CD8
T
cells specific for the Ag presented on DC was increased as in TCR
transgenic recipients, a significant and highly reproducible reduc-
tion in the number of Ag-loaded DC in the DLN was observed 72 h
after injection. No reduction was apparent at 16 h. Depletion of
CD8
T cells from TCR transgenic recipients before DC injection
prevented the reduction in number of Ag-loaded DC in the DLN
FIGURE 3. Quantitation and kinetics of DC migration to the DLN. A,
Groups of mice were given s.c. injections in both distal forelimbs with the
indicated numbers of CFSE
DC; DLN were removed for FACS analysis
20 h later. Each group contained three animals with each injected forelimb
treated as an independent event. B, Mice were injected with 10
6
CFSE
DC, and the DLN were removed for FACS analysis at the indicated time
points. Three animals were analyzed at each time point with each injected
forelimb treated independently (n6). Data are presented as the mean
percentage of DC collected in a gate encompassing only large cells (SE),
as outlined in Materials and Methods.
FIGURE 1. DC injected s.c. migrate to the paracortex of the DLN. Mice were injected s.c. with 5 10
5
CFSE
DC in the distal forelimb, and axillary
and brachial lymph nodes were removed for histological analysis 20 h after injection. Frozen tissue sections of the DLN were prepared and stained with
hematoxylin and eosin. CFSE
cells were detected by fluorescence microscopy. A, Low-power magnification with a representative CFSE
cell indicated
(arrowhead). B, High-power magnification showing three CFSE
cells.
FIGURE 2. DC that have migrated to the DLN can be detected by
FACS analysis. Mice were injected s.c. with 5 10
5
CFSE
DC in the
distal forelimb, and axillary and brachial lymph nodes were removed for
FACS analysis 20 h after injection. Data were collected in a gate encom-
passing only large cells as based on the known size of cultured DC. No
CFSE
cells were found outside this gate. A, Results are shown for a
noninjected animal, an animal injected with CFSE
DC (DLN and con-
tralateral nodes), and an animal that was injected with CFSE
DC that had
previously been heated to 80°C for 10 min. B, DLN from C57BL/6 mice
injected with either C57BL/6 DC (MHC class II
/
DC) or B6Aa
0
/Aa
0
DC
(MHC class II
/
DC). DLN cell suspensions were stained with anti-MHC
class II to determine MHC class II expression on CFSE
cells.
3097The Journal of Immunology
Downloaded from http://journals.aai.org/jimmunol/article-pdf/164/6/3095/1115875/3095.pdf by guest on 21 July 2023
(Fig. 4B). These results suggest that activation of TCR transgenic
CD8
T cells to cytotoxic effector cells is associated with the
disappearance of DC expressing specific Ag from the DLN. DC
clearance occurs between 16 and 72 h after injection, a period
consistent with the acquisition of effector function by CD8
T
cells as reported by other investigators (20, 21). The minimal
clearance of Ag-loaded DC demonstrated in the DLN of naive,
non-TCR transgenic mice probably reflects the smaller number of
Ag-specific T cells that become activated in these mice and hence
the lower number of interactions that result in DC clearance.
Clearance of Ag-loaded DC is accelerated in the presence of an
Ag-specific immune response
Next, we next investigated the fate of Ag-loaded DC in the context
of an ongoing immune response. To this end, CFSE
DC loaded
with the MHC class I-binding peptide LCMV
33–41
were adminis-
tered to mice that had been injected s.c. in the flank 7 days before
with 10
5
DC alone or with 10
5
DC loaded with the same
LCMV
33–41
peptide. The 7-day interval was chosen because our
previous studies have shown that Ag-specific T cell expansion is
maximal at this time (22); also, resistance to challenge with
LCMV
33–41
-expressing tumors can be demonstrated (17). In ad-
dition, “memory” recipients were also used. These animals had
received LCMV
33–41
-loaded DC 6 mo before and had subse-
quently rejected a challenge with tumor cells expressing the
LCMV
33–41
epitope. All mice received the LCMV
33–41
-loaded,
CFSE
DC by s.c. injection into the anterior forelimb. As can be
seen in Fig. 5, similar numbers of CFSE
DC were observed in the
DLN of mice that were either previously immunized with DC
without Ag or in those of mice that had received no previous
treatment. In contrast, a large reduction in the number of CFSE
DC was observed in the DLN of animals immunized with
LCMV
33–41
peptide-loaded DC 7 days earlier. The reduction was
clearly detectable as early as 20 h after injection, suggesting that it
was related to the presence of peptide-specific effector cells in the
recipient mice. Similar results were also obtained with MHC class
II
/
DC (data not shown), indicating that CD4
T cells were not
involved in DC elimination. A decrease in the number of CFSE
DC was also observed in the DLN of mice from the “memory”
group. However, in this latter case the decrease in CFSE
DC was
observed only at 72 h and not at 20 h after in vivo injection. This
result suggests that the reduction in the number of CFSE
DC was
related to the acquisition of effector activity by pre-existing mem-
ory CD8
T cells in the recipient mice.
Taken together, the data presented above indicate that the dis-
appearance of CFSE
DC from the DLN of recipient mice is as-
sociated with the presence or the development of effector CTL. We
suggest that in the course of an immune response, Ag-loaded DC
become targets for CTL activity and are eliminated from the
lymph node.
CTL-mediated clearance of DC from the lymph node impairs
induction of further immune responses
We wished to establish whether elimination of Ag-loaded DC dur-
ing an immune response impacts upon T cell activation initiated by
the same DC. As an in vivo readout of T cell activation, we ex-
amined the proliferation of CFSE-labeled, LCMV
33–41
-specific
CD8
T cells adoptively transferred into syngeneic recipients.
Proliferation of CFSE
T cells can be monitored in the DLN of
immunized mice by flow cytometry and is detected as progressive
halving of cellular fluorescence with every cell division completed
(23). Thus, we examined the specific proliferation induced by DC
that were or were not the targets of a pre-existing immune re-
sponse. Mice were preimmunized s.c. in the flank with DC
OVA
257–264
peptide to elicit an OVA-specific CTL immune re-
sponse or with DC only as a control. One week later, both groups
of mice received an adoptive transfer of CFSE
LCMV
33–41
-spe-
cific T cells and were immunized s.c. in the forelimb with DC
loaded with both the OVA
257–264
and LCMV
33–41
peptides. These
FIGURE 4. DC loaded with MHC class I-binding peptide Ag are
cleared from the DLN. Groups of C57BL/6 or TCR transgenic mice were
injected s.c. in the forelimb with either 10
6
CFSE
DC or 10
6
CFSE
DC
that had been loaded with the LCMV
33–41
peptide recognized by the trans-
genic TCR. A, DLN were removed for FACS analysis 16 or 72 h after
CFSE
DC injection. B, TCR transgenic recipients were depleted of CD8
T cells or were left untreated before injection with CFSE
DC. DLN were
removed for FACS analysis 72 h later. All data are presented as in Fig. 3B.
FIGURE 5. Clearance of Ag-loaded DC is accelerated in the presence
of an Ag-specific immune response. The presence of CFSE
, LCMV
33–41
peptide-loaded DC was monitored in the DLN of mice that had been im-
munized 1 wk before with 10
5
LCMV
33–41
-loaded DC injected s.c. in the
flank or with 10
5
DC not loaded with Ag or in mice that had received no
previous treatment. A fourth group, termed the “memory” group, was also
assessed. This group had been immunized with 10
5
LCMV
33–41
peptide-
loaded DC 6 mo before and had also successfully rejected a challenge with
10
6
LL-LCMV tumor cells 7 days after immunization. All groups were
injected s.c. with 10
6
CFSE
, LCMV
33–41
peptide-loaded DC in each fore-
limb. Three animals from each group were analyzed at the indicated time
points after CFSE
DC administration. Data are presented as in Fig. 3B.
3098 CTL-MEDIATED ELIMINATION OF DC IN VIVO
Downloaded from http://journals.aai.org/jimmunol/article-pdf/164/6/3095/1115875/3095.pdf by guest on 21 July 2023
mice were used to examine the proliferation of LCMV
33–41
-spe-
cific T cells. In addition, separate mice that had also been immu-
nized in the flank with DC OVA
257–264
or with DC only re-
ceived CFSE
DC that had been loaded with both the OVA
257–264
and LCMV
33–41
peptides by s.c. injection in the forelimb. These
latter mice were used to evaluate the numbers of CFSE
DC in the
DLN. As shown in Fig. 6, OVA
257–264
LCMV
33–41
-loaded DC
were rapidly eliminated in mice that had previously been immu-
nized with DC OVA
257–264
, most likely due to the induced
OVA-specific CTL response. In contrast, a considerable propor-
tion of OVA
257–264
LCMV
33–41
-loaded DC could be demon-
strated in the DLN of mice previously immunized with DC only.
Fig. 6 also shows the proliferative response of LCMV
33–41
-spe-
cific T cells in mice that had received the same DC immunizations.
In mice in which DC could be demonstrated in the DLN,
LCMV
33–41
-specific T cells underwent several cycles of cell di-
vision, indicating the onset of an active immune response. In con-
trast, in mice in which DC had disappeared from the DLN, only
limited proliferation of LCMV
33–41
-specific T cells could be dem-
onstrated. We conclude from these results that CTL-mediated
elimination of DC may have a profound impact on T cell immune
responses and may prevent the initiation of immune responses to
other Ags that are simultaneously presented on the same DC.
CTL-mediated clearance of tumor Ag-loaded DC from the lymph
node impairs antitumor immune responses
We wished to extend our findings by examining whether elimina-
tion of Ag-loaded DC during the induction of an antitumor im-
mune response has a significant negative impact on antitumor im-
munity. For these studies we used the tumor cell line LL-LCMV,
a Lewis lung carcinoma expressing the LCMV
33–41
epitope (17).
All mice were preimmunized s.c. in the flank with DC loaded with
OVA
257–264
to induce OVA
257–264
-specific CTL responses. One
week later, mice received a second s.c. immunization in the op-
posite flank with MHC class II
/
DC that were loaded with both
OVA
257–264
and the “tumor Ag” LCMV
33–41
or with MHC class
II
/
DC loaded with the “tumor Ag” LCMV
33–41
only. MHC
class II
/
DC were used in this experiment to rule out any effects
mediated by CD4
T cells. Similar results were obtained when
MHC class II
/
DC were used (data not shown), because DC are
eliminated by pre-existing CTL responses regardless of MHC class
II expression. DC presenting OVA
257–264
and tumor Ag became
targets of the anti-OVA
257–264
CTL response and were eliminated
(data not shown). In contrast, DC presenting tumor Ag alone re-
mained unaffected. One week after the second DC immunization,
mice were challenged with LL-LCMV, and tumor size was mea-
sured. In Fig. 7, we show that DC presenting both OVA
257–264
and LCMV
33–41
were less effective at inducing antitumor immu-
nity than DC presenting the LCMV
33–41
Ag alone were. This re-
duction in efficacy was most likely due to the rapid clearance of
OVA
257–264
-loaded DC by OVA
257–264
-specific CTL, which pre-
vented the productive initiation of an LCMV
33–41
-specific im-
mune response. However, a limited degree of antitumor immunity
was reproducibly induced by OVA
257–264
-loaded DC in these ex-
periments, suggesting that the DC were able to induce some degree
of T cell activation before being eliminated. Alternatively, DC-
associated Ag had been released and taken up by other APC, al-
lowing a specific immune response to be initiated.
Discussion
The capacity of DC to migrate from peripheral tissues to lymphoid
organs and to initiate immune responses has been the subject of a
number of investigations (reviewed in Ref. 1). In contrast, the sub-
sequent fate of DC has received little attention. DC are not ob-
served in the efferent lymph (12), suggesting that they may not be
FIGURE 6. CTL-mediated clearance of Ag-loaded DC from the DLN
limits the induction of further immune responses. Groups of mice were
immunized s.c. in the flank with either 10
5
OVA
257–264
-loaded DC or with
10
5
DC alone. All mice received a second injection of DC loaded with
OVA
257–264
and LCMV
33–41
s.c. in the forelimb. Survival of the DC ad-
ministered with the latter injection and their induction of a T cell response
were examined. Left panel, CFSE
DC simultaneously loaded with
OVA
257–264
and LCMV
33–41
are eliminated in OVA-immunized mice.
Data were collected 66 h after injection of CFSE
DC and are presented as
in Fig. 3B.Right panel, Adoptively transferred CFSE
TCR transgenic
cells specific for LCMV
33–41
fail to divide in response to specific peptide
presented on DC if the DC are eliminated by an existing CTL response.
Data were collected 66 h after DC immunization; only CFSE
,V
2
V
8.1,8.2
events are shown. FACS profiles shown are of one represen-
tative mouse of the six in each group.
FIGURE 7. CTL-mediated clearance of tumor Ag-loaded DC from the
DLN impairs the generation of antitumor immune responses. Two groups
of mice (n5) were immunized with 10
5
OVA
257–264
peptide-loaded DC
by s.c. injection in the flank, and then 1 wk later, one group was immunized
with LCMV
33–41
peptide-loaded MHC class II
/
DC in the contralateral
flank, and the second group was immunized with MHC class II
/
DC
loaded with both LCMV
33–41
and OVA
257–264
peptides. After 7 days, both
of these groups and an additional control group were challenged with 10
6
LL-LCMV tumor cells presenting the LCMV
33–41
epitope. Mice were
monitored for tumor growth every 3–4 days, and tumor size for each group
was calculated as the mean of the products of bisecting diameters (SE).
Measurements were terminated for each group when the first animal de-
veloped a tumor in excess of 200 mm
2
.
3099The Journal of Immunology
Downloaded from http://journals.aai.org/jimmunol/article-pdf/164/6/3095/1115875/3095.pdf by guest on 21 July 2023
capable of leaving the lymph node and may die in situ. It has been
speculated that DC may be eliminated by the very immune re-
sponses they elicit. This would allow the activated T cells to pro-
liferate freely and to migrate away from the lymph node to pe-
ripheral tissues or, in the case of CD4
T cells, it would allow the
T cells to migrate to the B cell areas of the lymph node to deliver
help for Ab production (14, 24). Furthermore, elimination of DC
could function as a negative feedback mechanism to limit T cell
responses and also could prevent excessive accumulation in the
lymph node of DC that carry Ags to which the immune system has
already been sensitized.
In this paper we have monitored the migration of CFSE
DC
injected s.c. into mice. The effect of Ag presented on MHC class
I on DC survival in the DLN was also examined. We show that
Ag-loaded DC disappear from the lymph node during the course of
a CD8
T cell response, with kinetics that parallel the known
kinetics of activation of CD8
T cells to effector function (20, 25).
Ag-loaded DC were immediately cleared in recently immunized
mice with a reduction in DC numbers observed in the DLN within
20 h of injection, suggesting that some DC may have been elim-
inated before reaching the lymph node. The kinetics of clearance
were somewhat slower in “memory” mice, in which labeled DC
were clearly demonstrated in the DLN at 20 h after injection but
not at 72 h after injection. Memory CD8
cells presumably would
need to reacquire effector function before mediating elimination of
DC in these mice (21). Lastly, Ag-loaded DC were cleared from
the DLN between 16 h and 72 h after s.c. injection into naive, TCR
transgenic mice. This indicates that Ag-loaded DC may be elim-
inated as a consequence of a primary CD8
T cell immune re-
sponse, perhaps as a mechanism to allow turnover of DC within
the lymph node. This process may contribute to the restriction of
the number of T cell specificities generated in a given immune
response, a phenomenon that has been reported for viral infections
(26–28). High-avidity T cells that become successfully activated
in response to Ag on a given DC may eliminate that DC, thereby
preventing the full activation of lower-avidity T cells that recog-
nize different antigenic epitopes presented on the same DC.
The close parallel of the kinetics of DC clearance with the de-
velopment of a CTL response suggests that the failure to demon-
strate CFSE
DC in the DLN is most likely due to active elimi-
nation of labeled DC rather than to their migration to another site
or to selective loss of the CFSE label from otherwise viable DC.
This hypothesis is also supported by the observation that DC elim-
ination was inhibited by anti-CD8 Ab treatment and did not require
MHC class II expression by the DC. Elimination of DC was clearly
a systemic event in that it was observed at sites distant from the
site of original immunization, as would be expected on the basis of
the CTL’s ability to recirculate to different sites. However, the
mechanism of DC clearance, although clearly Ag-specific, is as yet
undefined. We cannot exclude the possibility that there was a con-
stant rapid turnover of DC in the DLN and that the clearance ob-
served represented a failure of DC to migrate to the DLN in the
presence of activated CTL. However, the fact that Ag-loaded DC
were present in the DLN of naive TCR transgenic mice at 16 h and
then were lost by 72 h is highly suggestive of a cytolytic mecha-
nism operating within the node. Furthermore, the kinetics of DC
clearance in naive animals are consistent with the reported time
required for CTL to gain cytolytic function upon activation (20,
25). Preliminary experiments indicated that DC clearance is not
impaired in IFN-
-deficient mice (data not shown), suggesting that
this cytokine is not critical to the clearance process and that other
effector mechanisms, presumably cytotoxicity, have a greater role.
Other authors have also reported a decrease in the number of Ag-
loaded DC in the lymph node during a CD4
T cell immune re-
sponse (14). That decrease appeared less profound than the one we
describe here, probably reflecting different mechanisms of DC
elimination during CD4
T cell responses compared with CD8
T
cell responses. DC elimination has also been demonstrated by his-
tological means in mice exposed to viruses that induce strong CTL
immune responses (29).
We also show here that CTL-mediated clearance of Ag-loaded
DC can have significant consequences on immune responses. Ag-
specific T cell activation and proliferation, and induction of anti-
tumor immunity were severely impaired when the DC were
cleared by an existing CTL response. However, it should be noted
that a limited degree of T cell proliferation and antitumor immu-
nity was reproducibly demonstrated in these experiments, suggest-
ing that some T cell activation was occurring despite the rapid
clearance of DC. This weakened T cell response may have resulted
from suboptimal stimulation by DC in the process of being elim-
inated or may have been induced by a low number of healthy DC
that had escaped elimination.
It has been reported that Ag from short-lived migratory DC can
be processed and presented by recipient DC within the lymph node
(30). If DC are cleared from the DLN by a cytolytic mechanism,
these dying cells may provide a ready source of Ag for lymph
node-resident DC. The recipient DC may be of the “lymphoid”
subclass that has been proposed to serve a regulatory, tolerogenic
function (31, 32). This process could account for the reduced an-
titumor responses observed when the DC were “cleared” by an
existing CTL response. Alternatively, it is possible that transfer of
Ag from migratory DC to lymph node-resident DC, in fact, may be
stimulatory but that this process was inefficient in our experimental
system.
One surprising finding in our study is that a proportion of the
injected DC appeared to persist in the DLN for a long time (in
excess of 7 days from the in vivo transfer). This suggests that DC
may have the ability to “wait” for T cells of the appropriate spec-
ificity to migrate through the lymph node and to recognize Ag on
their surface. The prolonged survival of some DC may be simply
a stochastic process or alternatively may reflect heterogeneity in
the degree of activation of our DC before injection in vivo. It is
also possible that the DC become activated in vivo by host CD4
T cells to become the “temporal bridge” hypothesized by Ridge et
al. (10), thereby becoming “conditioned” to stimulate further im-
mune responses. Our experiments do not distinguish among these
possibilities. In addition, we cannot exclude the possibility that this
apparent prolonged persistence of DC in the DLN may in fact
represent a slow turnover of DC that are continuously replaced by
fresh DC migrating from the site of injection. We find this possi-
bility less attractive in that it would appear that the lymph node
should offer a more favorable environment for DC survival com-
pared with that of the periphery. However, additional experiments
are required to clarify this issue.
The process of DC clearance has implications for the design of
DC-based immunotherapy regimes. From our results it can be con-
cluded that repeated immunizations at short intervals with DC
loaded with a given MHC class I-binding peptide may not be ef-
fective at enhancing responses to this Ag because the DC used in
the secondary injections would be rapidly cleared by existing CTL.
Indeed, our own experiments have suggested that repeated immu-
nization with LCMV
33–41
-loaded DC does not lead to enhanced
CTL responses against LL-LCMV challenge (data not shown).
However, repeated DC injections may be effective in maintaining
effector function in “memory” CD8
T cells that may have lost
activity due to suboptimal presentation of Ag in the context of
tumor tissue.
3100 CTL-MEDIATED ELIMINATION OF DC IN VIVO
Downloaded from http://journals.aai.org/jimmunol/article-pdf/164/6/3095/1115875/3095.pdf by guest on 21 July 2023
Acknowledgments
We thank the personnel of the Wellington Medical School Biomedical
Research Unit for animal husbandry and Drs. H. Pircher and H. Bluethman
for the modified mouse strains. The expert help of Dr. H. H. Teoh,
A. Richardson, and R. Irlam of the Department of Laboratory Services
(Anatomic Pathology) at Wellington Hospital in the preparation and anal-
ysis of lymph node tissue sections is also gratefully acknowledged.
References
1. Banchereau, J., and R. M. Steinman. 1998. Dendritic cells and the control of
immunity. Nature 392:245.
2. Romani, N., S. Koide, M. Crowley, M. Witmer-Pack, A. M. Livingstone,
C. G. Fathman, K. Inaba, and R. M. Steinman. 1989. Presentation of exogenous
protein antigens by dendritic cells to T cell clones: intact protein is presented best
by immature, epidermal Langerhans cells. J. Exp. Med. 169:1169.
3. Cumberbatch, M., and I. Kimber. 1992. Dermal tumour necrosis factor-
induces
dendritic cell migration to draining lymph nodes, and possibly provides one stim-
ulus for Langerhans’ cell migration. Immunology 75:257.
4. Cumberbatch, M., and I. Kimber. 1995. Tumour necrosis factor-
is required for
accumulation of dendritic cells in draining lymph nodes and for optimal contact
sensitization. Immunology 84:31.
5. Sallusto, F., and A. Lanzavecchia. 1994. Efficient presentation of soluble antigen
by human dendritic cells is maintained by granulocyte/macrophage colony stim-
ulating factor plus interleukin-4 and downregulated by tumor necrosis factor-
.
J. Exp. Med. 179:1109.
6. Sallusto, F., M. Cella, C. Danieli, and A. Lanzavecchia. 1995. Dendritic cells use
macropinocytosis and the mannose receptor to concentrate macromolecules in the
major histocompatibility complex class II compartment: downregulation by cy-
tokines and bacterial products. J. Exp. Med. 182:389.
7. Caux, C., C. Massacrier, B. Vanbervliet, B. Dubois, C. Van Kooten, I. Durand,
and J. Banchereau. 1994. Activation of human dendritic cells through CD40
crosslinking. J. Exp. Med. 180:1263.
8. Cella, M., D. Scheidegger, K. Palmer-Lehmann, P. Lane, A. Lanzavecchia, and
G. Alber. 1996. Ligation of CD40 on dendritic cells triggers production of high
levels of interleukin-12 and enhances T cell stimulatory capacity: T-T help via
APC activation. J. Exp. Med. 184:747.
9. Bennett, S. R. M., F. R. Carbone, F. Karamalis, R. A. Flavell, J. F. A. P. Miller,
and W. R. Heath. 1998. Help for cytotoxic-T-cell responses is mediated by CD40
signalling. Nature 393:478.
10. Ridge, J. P., F. Di Rosa, and P. Matzinger. 1998. A conditioned dendritic cell can
be a temporal bridge between a CD4
T-helper and a T-killer cell. Nature 393:
474.
11. Schoenberger, S. P., R. E. M. Toes, E. I. H. Vandervoort, R. Offringa, and
C. J. M. Melief. 1998. T-cell help for cytotoxic T lymphocytes is mediated by
CD40-CD40L interactions. Nature 393:480.
12. Pugh, C. W., G. G. MacPherson, and H. W. Steer. 1983. Characterization of
nonlymphoid cells derived from rat peripheral lymph. J. Exp. Med. 157:1758.
13. Kupiec-Weginski, J. W., J. M. Austyn, and P. J. Morris. 1988. Migration patterns
of dendritic cells in the mouse. J. Exp. Med. 167:632.
14. Ingulli, E., A. Mondino, A. Khoruts, and M. K. Jenkins. 1997. In vivo detection
of dendritic cell antigen presentation to CD4
T cells. J. Exp. Med. 185:2133.
15. Pircher, H., K. Buerki, R. Lang, H. Hengartner, and R. M. Zinkernagel. 1989.
Tolerance induction in double specific T-cell receptor transgenic mice varies with
antigen. Nature 342:559.
16. Ko¨ntgen, F., G. Su¨ss, C. Stewart, M. Steinmetz, and H. Bluethmann. 1993. Tar-
geted disruption of the MHC class II Aa gene in C57BL/6 mice. Int. Immunol.
5:957.
17. Hermans, I. F., A. Daish, P. Moroni-Rawson, and F. Ronchese. 1997. Tumor
peptide-pulsed dendritic cells isolated from spleen or cultured in vitro from bone
marrow precursors can provide protection against tumor challenge. Cancer Im-
munol. Immunother. 44:341.
18. Garrigan, K., P. Moroni-Rawson, C. McMurray, I. Hermans, N. Abernethy,
J. Watson, and F. Ronchese. 1996. Functional comparison of spleen dendritic
cells and dendritic cells cultured in vitro from bone marrow precursors. Blood
88:3508.
19. Coligan, J. E., A. M. Kruisbeek, D. H. Margulies, E. M. Shevach, and W. Strober.
1991. Current Protocols in Immunology. Wiley, New York.
20. Oehen, S., and K. Brduscha-Riem. 1998. Differentiation of naive CTL to effector
and memory CTL: correlation of effector function with phenotype and cell divi-
sion. J. Immunol. 161:5338.
21. Ehl, S., P. Klenerman, P. Aichele, H. Hengartner, and R. M. Zinkernagel. 1997.
A functional and kinetic comparison of antiviral effector and memory cytotoxic
T lymphocyte populations in vivo and in vitro. Eur. J. Immunol. 27:3404.
22. Hermans, I. F., D. S. Ritchie, A. Daish, J. Yang, M. R. Kehry, and F. Ronchese.
1999. Impaired ability of MHC class II
/
dendritic cells to provide tumor pro-
tection is rescued by CD40 ligation. J. Immunol. 163:77.
23. Lyons, A. B., and C. R. Parish. 1994. Determination of lymphocyte division by
flow cytometry. J. Immunol. Methods 171:131.
24. Garside, P., E. Ingulli, R. R. Merica, J. G. Johnson, R. J. Noelle, and
M. K. Jenkins. 1998. Visualization of specific B and T lymphocyte interactions
in the lymph node. Science 281:96.
25. Zimmermann, C., A. Prevost-Blondel, C. Blaser, and H. Pircher. 1999. Kinetics
of the response of naive and memory CD8 T cell antigen: similarities and dif-
ferences. Eur. J. Immunol. 29:284.
26. Pantaleo, G., J. F. Demarest, H. Soudeyns, C. Graziosi, F. Denis,
J. W. Adelsberger, P. Borrow, M. S. Saag, G. M. Shaw, R. P. Sekaly, and
A. S. Fauci. 1994. Major expansion of CD8
T cells with a predominant V
usage during the primary immune response to HIV. Nature 370:463.
27. Vitiello, A., L. Yuan, R. W. Chesnut, J. Sidney, S. Southwood, P. Farness,
M. R. Jackson, P. A. Peterson, and A. Sette. 1995. Immunodominance analysis of
CTL responses to influenza PR8 virus reveals two new dominant and subdomi-
nant K
b
-restricted epitopes. J. Immunol. 157:5555.
28. Callan, M. F., N. Steven, P. Krausa, J. D. Wilson, P. A. Moss, G. M. Gillespie,
J. I. Bell, A. B. Rickinson, and A. J. McMichael. 1996. Large clonal expansions
of CD8
T cells in acute infectious mononucleosis. Nat. Med. 2:906.
29. Odermatt, B., M. Eppler, T. P. Leist, H. Hengertner, and R. M. Zinkernagel.
1991. Virus-triggered acquired immunodeficiency by cytotoxic T-cell-dependent
destruction of antigen-presenting cells and lymph follicle structure. Proc. Natl.
Acad. Sci. USA 88:8252.
30. Inaba, K., S. Turley, F. Yamaide, T. Iyoda, K. Mahnke, M. Inaba, M. Pack,
M. Subklewe, B. Sauter, D. Sheff, et al. 1998. Efficient presentation of phago-
cytosed cellular fragments on the major histocompatibility complex class II prod-
ucts of dendritic cells. J. Exp. Med. 188:2163.
31. Kronin, V., K. Winkel, G. Suss, A. Kelso, W. Heath, J. Kirberg, H. von Boehmer,
and K. Shortman. 1996. A subclass of dendritic cells regulates the response of
naive CD8 T cells by limiting their IL-2 production. J. Immunol. 157:3819.
32. Suss, G., and K. Shortman. 1996. A subclass of dendritic cells kills CD4 T cells
via Fas/Fas-ligand-induced apoptosis. J. Exp. Med. 183:1789.
3101The Journal of Immunology
Downloaded from http://journals.aai.org/jimmunol/article-pdf/164/6/3095/1115875/3095.pdf by guest on 21 July 2023
... But due to the low life span of DCs its use is limited. 130 When compared to antigen presenting DCs, the exosomes isolated from the dendritic cells consists of complexes made from major histocompatibility factors and peptides and some co-stimulatory molecules on their membrane, that allows them to continue with the antigen presentation and enhance the process of immunisation in murine models. 131 Survivin, an anti-apoptotic protein, suppresses the activation of cell death in large number of cancer cells. ...
Article
Full-text available
Extracellular vesicles (EVs) are lipid-bound vesicles produced into the extracellular space by cells. Apoptotic bodies (ApoBD), microvesicles (MVs), and exosomes are examples of EVs, which act as essential regulators in cell-cell communication in both normal and diseased conditions. Natural cargo molecules such as miRNA, messenger RNA, and proteins are carried by EVs and transferred to nearby cells or distant cells through the process of circulation. Different signalling cascades are then influenced by these functionally active molecules. The information to be delivered to the target cells depends on the substances within the EVs that also includes synthesis method. EVs have attracted interest as potential delivery vehicles for therapies due to their features such as improved circulation stability, biocompatibility, reduced immunogenicity, and toxicity. Therefore, EVs are being regarded as potent carriers of therapeutics that can be used as a therapeutic agent for diseases like cancer. This review focuses on the exosome-mediated drug delivery to cancer cells and the advantages and challenges of using exosomes as a carrier molecule.
... When Hermans et al. injected non-antigen-loaded DCs into mice, they could identify these cells in draining lymph nodes several days following injection. Interestingly, the use of antigen-loaded DCs resulted in significantly fewer lymph node DCs, a finding that was reversed following CD8+ T cell depletion, suggesting that antigen-specific CD8+ T cells are responsible for restricting the size of the DC population (17). This same group went on to evaluate the importance of an intact cytotoxicity pathway for this response. ...
Article
Full-text available
Familial hemophagocytic lymphohistiocytosis (fHLH) encompasses a group of rare inherited immune dysregulation disorders characterized by loss-of-function mutations in one of several genes involved in the assembly, exocytosis, and function of cytotoxic granules within CD8+ T cells and natural killer (NK) cells. The resulting defect in cytotoxicity allows these cells to be appropriately stimulated in response to an antigenic trigger, and also impairs their ability to effectively mediate and terminate the immune response. Consequently, there is sustained lymphocyte activation, resulting in the secretion of excessive amounts of pro-inflammatory cytokines that further activate other cells of the innate and adaptive immune systems. Together, these activated cells and pro-inflammatory cytokines mediate tissue damage that leads to multi-organ failure in the absence of treatment aimed at controlling hyperinflammation. In this article, we review these mechanisms of hyperinflammation in fHLH at the cellular level, focusing primarily on studies performed in murine models of fHLH that have provided insight into how defects in the lymphocyte cytotoxicity pathway mediate rampant and sustained immune dysregulation.
... Therefore, DC-based vaccine delivery sys- DC vaccines still faces many challenges. For example, the body's immune response is very complex, the activities of DC vaccines vary greatly in vivo and in vitro, little DCs migrate to the draining lymph nodes, and the efficacy against pathogens or abnormal cells is still insufficient, which greatly limits the CTL response [9]. Therefore, further improving the efficacy of DC vaccines is always an issue that researchers need to explore. ...
Chapter
Graft-versus-host disease (GVHD) develops when stem cell graft containing immune cells is transplanted from donor to the recipient resulting in characteristic immune-mediated reactions in the recipient. Acute GVHD is one of the major causes of morbidity and mortality in allogeneic hematopoietic stem cell transplantation (allo SCT). Alloreactive T cells of the donor are involved in the pathophysiology of acute GVHD. They react to the host antigens presented by antigen presenting cells (APCs) to donor T cells. The cytokines released following tissue injury from conditioning regimens further amplify the alloimmune response. Acute GVHD not only damages the skin, liver and gut but also affects the thymus and bone marrow which have significant clinical relevance. First line treatment for acute GVHD consists of corticosteroids. Response to corticosteroids is seen in approximately 50% of patients. Patients with steroid-resistant acute GVHD have a very poor prognosis, with mortality rates in excess of 90%. If GVHD prophylaxis is not used, serious acute GVHD can affect almost every recipient.
Article
Unlabelled: Oral squamous cell carcinoma (OSCC) is one of the most common malignant tumors of the head and neck with poor prognosis. This study aimed to explore the role of lnc-METRNL-1 in occurrence and prognosis of OSCC patients. Expression of lnc-METRNL-1 was compared between OSCC samples and paracancerous samples from The Cancer Genome Atlas (TCGA) database. Additionally, the lnc-METRNL-1 expression in cell lines was detected by using qRT-PCR. The overall survival (OS) was estimated based on the Kaplan-Meier and the immune cell infiltration was evaluated using CIBERSORT. Significantly enriched biological pathways were identified by Gene-set enrichment analysis (GSEA). Differential expression analysis was done in edgeR package. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways of differential expression genes were conducted using DAVID version 6.8. The lnc-METRNL-1 expression in OSCC was significantly lower than that in paracancerous samples, and patients with low lnc-METRNL-1 expression had poorer OS. Additionally, lnc-METRNL-1 was significantly down-regulated in OSCC cell lines compared with normal cell line. High expression of lnc-METRNL-1 was closely associated with the activation of several tumor metabolic and metabolism-related pathways. Besides, aberrant lnc-METRNL-1 expression was found to be related to the differential infiltration of immune cells in tumor tissue, such as regulatory T cells, and Macrophages. Low lnc-METRNL-1 expression was probably a poor prognostic biomarker for OSCC patients. Moreover, the potential role of lnc-METRNL-1 in the onset of OSCC was partly revealed. Supplementary information: The online version contains supplementary material available at 10.1007/s13205-023-03674-0.
Article
Extracellular vesicles (EVs) can affect immune responses through antigen presentation and costimulation or coinhibition. We generated designer EVs to modulate T cells in the context of type 1 diabetes, a T cell-mediated autoimmune disease, by engineering a lymphoblast cell line, K562, to express HLA-A*02 (HLA-A2) alongside costimulatory CD80 and/or coinhibitory programmed death ligand 1 (PD-L1). EVs presenting HLA-A2 and CD80 activated CD8+ T cells in a dose, antigen, and HLA-specific manner. Adding PD-L1 to these EVs produced an immunoregulatory response, reducing CD8+ T cell activation and cytotoxicity in vitro. EVs alone could not stimulate T cells without antigen-presenting cells. EVs lacking CD80 were ineffective at modulating CD8+ T cell activation, suggesting that both peptide-HLA complex and costimulation are required for EV-mediated immune modulation. These results provide mechanistic insight into the rational design of EVs as a cell-free approach to immunotherapy that can be tailored to promote inflammatory or tolerogenic immune responses.
Article
Immunotherapy has become a rapidly developing approach in the treatment of cancer. Cancer immunotherapy aims at promoting the immune system response to react against the tumor. In view of this, we develop a mathematical model for immune-tumor interplays with immunotherapeutic drug, and strategies for optimally administering treatment. The tumor-immune dynamics are given by a system of five coupled nonlinear ordinary differential equations which represent the interaction among tumor-specific CD4+T cells, tumor-specific CD8+T cells, tumor cells, dendritic cells and the immuno-stimulatory cytokine interleukin-2 (IL-2), extended through the addition of a control function describing the application of a dendritic cell vaccination. Dynamical behavior of the system is studied from the analytical as well numerical points of views. The main aim is to investigate the treatment regimens which minimize the tumor cell burden and the toxicity of dendritic cell vaccination. Our numerical simulations demonstrate that the optimal treatment strategies using dendritic cell vaccination reduce the tumor cell burden and increase the cell count of CD4+T cells, CD8+T cells, dendritic cells and IL-2. The most influential parameters having significant impacts on the tumor cells are identified by employing the approach of global sensitivity analysis.
Article
ABSTRACT: The onset and spread of the SARS-CoV-2 virus have created an unprecedented universal crisis. Although vaccines have been developed against the parental SARS-CoV-2, outbreaks of the disease still occur through the appearance of different variants, suggesting a continuous need for improved and effective therapeutic strategies. Therefore, we developed a novel nanovesicle presenting Spike protein on the surface of the dendritic cell- derived extracellular vesicles (DEVs) for use as a potential vaccine platform against SARS-CoV-2. DEVs express peptide/MHC-I (pMHC-I) complexes, CCR-7, on their surface. The immunogenicity and efficacy of the Spike- activated DEVs were tested in mice and compared with free Spike protein. A 1/10 Spike equivalent dose of DEVs showed a superior potency in inducing anti-Spike IgG titers in blood of mice when compared to dendritic cells or free Spike protein treatment. Moreover, DEV-induced sera effectively reduced viral infection by 55−60% within days of booster dose administration. Furthermore, a 1/10 Spike equivalent dose of DEV-treated mice was found to be equally effective in inducing CD19 + CD38 + T-cells in the spleen and lymph node; CD8 cells in the bone marrow, spleen, and lymph node; and CD4 + CD25 + T-cells in the spleen and lymph node after 90 days of treatment. Thus, our results support the immunogenic nature of DEVs, demonstrating that a low dose of DEVs induces antibodies to inhibit SARS-CoV-2 infection in vitro, therefore warranting further investigations.
Article
Full-text available
Exosomes, nano-sized extracellular vesicles for intercellular communications, are gaining rapid momentum as a novel strategy for the diagnosis and therapeutics of a spectrum of diseases including cancers. Secreted by various cell sources, exosomes pertain numerous functionalities from their parental cells and have enhanced stability that enable them with many features favorable for clinical use and commercialization. This paper focuses on the possible roles of exosomes in cancer therapeutics and reviews current exosome-based innovations toward enhanced cancer management and challenges that limit their clinical translation. Importantly, this paper casts insights on how cold atmospheric plasma, an emerging anticancer strategy, may aid in innovations on exosome-based onco-therapeutics toward improved control over cancers.
Article
Full-text available
We investigated the possibility that T helper cells might enhance the stimulatory function of dendritic cells (DCs). We found that ligation of CD40 by CD40L triggers the production of extremely high levels ofbioactive IL-12. Other stimuli such as microbial agents, TNF-cx or LPS are much less effective or not at all. In addition, CD40L is the most potent stimulus in upregu-lating the expression ofICAM-1, CD80, and CD86 molecules on DCs. These effects of CD40 ligation result in an increased capacity of DCs to trigger proliferative responses and IFN-~/pro-duction by T cells. These findings reveal a new role for CD40-CD40L interaction in regulating DC function and are relevant to design therapeutic strategies using cultured DCs.
Article
Full-text available
Dendritic cells, the professional antigen-presenting cells (APC) involved in T cell priming, express CD40, a molecule which triggering plays a key role in B cell growth and differentiation as well as monocyte activation. Herein we demonstrate that dendritic Langerhans cells (D-Lc) generated by culturing cord blood CD34+ progenitor cells with granulocyte/macrophage colony-stimulating and tumor necrosis factor alpha (TNF-alpha) express functional CD40 at a density higher than that found on B cells. Culturing D-Lc on CD40-ligand (CD40L) transfected L cells allowed D-Lc survival as 50 +/- 15% of seeded cells were recovered after 4 d while only 5% survived over control L cells. CD40 activation induced important morphological changes with a reduction of cytoplasmic content and a remarkable increase of dendrite development as well as an altered phenotype. In particular, CD40 triggering induced maintenance of high levels of major histocompatibility complex class II antigens and upregulation of accessory molecules such as CD58, CD80 (B7-1) and CD86 (B7-2). CD40 engagement also seems to turn on D-Lc maturation as illustrated by upregulation of CD25, a molecule usually expressed on interdigitating dendritic cells of secondary lymphoid organs. Finally, CD40 activated D-Lc secreted a limited set of cytokines (TNF-alpha, IL-8, and macrophage inflammatory protein 1 alpha [MIP-1 alpha]) whereas a similar activation induced elutriated monocytes to secrete IL-1 alpha, IL-1 beta, IL-6, IL-8, IL-10, TNF-alpha, and MIP-1 alpha. As D-Lc activated T cells upregulated CD40L, it is likely that CD40 activation of D-Lc observed herein with a fibroblast cell line stably expressing CD40L, mimics physiological interactions between dendritic cells and T cells.
Article
Full-text available
Although lymphoid dendritic cells (DC) are thought to play an essential role in T cell activation, the initial physical interaction between antigen-bearing DC and antigen-specific T cells has never been directly observed in vivo under conditions where the specificity of the responding T cells for the relevant antigen could be unambiguously assessed. We used confocal microscopy to track the in vivo location of fluorescent dye-labeled DC and naive TCR transgenic CD4+ T cells specific for an OVA peptide–I-Ad complex after adoptive transfer into syngeneic recipients. DC that were not exposed to the OVA peptide, homed to the paracortical regions of the lymph nodes but did not interact with the OVA peptide-specific T cells. In contrast, the OVA peptide-specific T cells formed large clusters around paracortical DC that were pulsed in vitro with the OVA peptide before injection. Interactions were also observed between paracortical DC of the recipient and OVA peptide-specific T cells after administration of intact OVA. Injection of OVA peptide-pulsed DC caused the specific T cells to produce IL-2 in vivo, proliferate, and differentiate into effector cells capable of causing a delayed-type hypersensitivity reaction. Surprisingly, by 48 h after injection, OVA peptide-pulsed, but not unpulsed DC disappeared from the lymph nodes of mice that contained the transferred TCR transgenic population. These results demonstrate that antigen-bearing DC directly interact with naive antigen-specific T cells within the T cell–rich regions of lymph nodes. This interaction results in T cell activation and disappearance of the DC.
Article
Full-text available
Summary We have previously demonstrated that human peripheral blood low density mononuclear cells cultured in granulocyte/macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)-4 develop into dendritic cells (DCs) that are extremely efficient in presenting soluble antigens to T ceils. To identify the mechanisms responsible for efficient antigen capture, we studied the endocytic capacity of DCs using fluorescein isothiocyanate--dextran, horseradish peroxidase, and lucifer yellow. We found that DCs use two distinct mechanisms for antigen capture. The first is a high level of fluid phase uptake via macropinocytosis. In contrast to what has been found with other cell types, macropinocytosis in DCs is constitutive and allows continuous internalization of large volumes of fluid. The second mechanism of capture is mediated via the mannose receptor (MR), which is expressed at high levels on DCs. At low ligand concentrations, the MR can deliver a large number of ligands to the cell in successive rounds. Thus, while macropinocytosis endows DCs with a high capacity, nonsaturable mechanism for capture of any soluble antigen, the MR gives an extra capacity for antigen capture with some degree of selectivity for non-self molecules. In addition to their high endocytic capacity, DCs from GM-CSF+ IL-4-dependent cultures are characterized by the presence of a large intracellular compartment that contains high levels of class II molecules, cathepsin D, and lysosomal-~issociated membrane protein-I, and is rapidly accessible to endocytic markers. We investigated whether the capacity of DCs to capture and process antigen could be modulated by exogenous stimuli. We found that DCs respond to tumor necrosis factor or, CD40 ligand, IL-1, and lipopolysaccharide with a coordinate series of changes that include downregulation of macropinocytosis and Fc receptors, disappearance of the class II compartment, and upregulation of adhesion and costimulatory molecules. These changes occur within 1-2 d and are irreversible, since neither pinocytosis nor the class II compartment are recovered when the maturation-inducing stimulus is removed. The specificity of the MR and the capacity to respond to inflammatory stimuli maximize the capacity of DCs to present infectious non-self antigens to T cells.
Article
Full-text available
The capacity of dendritic cells to present protein antigens has been studied with two MHC class II-restricted, myoglobin-specific, T cell clones. Spleen dendritic cells and cultured epidermal Langerhans cells (LC) presented native myoglobin weakly and often not at all. These same populations were powerful stimulators of allogeneic T cells in the primary MLR. Freshly isolated LC were in contrast very active in presenting proteins to T cell clones but were weak stimulators of the MLR. Both fresh and cultured LC could present specific peptide fragments of myoglobin to the clones. These results suggest that dendritic cells in nonlymphoid tissues like skin can act as sentinels for presenting antigens in situ, their accessory function developing in two phases. First antigens are captured and appropriately presented. Further handling of antigen then is downregulated while the cells acquire strong sensitizing activity for the growth and function of resting T lymphocytes. The potent MLR stimulating activity of cultured epidermal LC and lymphoid dendritic cells probably reflects prior handling of antigens leading to the formation of allogeneic MHC-peptide complexes.
Article
We have previously demonstrated that human peripheral blood low density mononuclear cells cultured in granulocyte/macrophage colony-stimulating factor (GM-CSF) and interleukin (IL)-4 develop into dendritic cells (DCs) that are extremely efficient in presenting soluble antigens to T cells. To identify the mechanisms responsible for efficient antigen capture, we studied the endocytic capacity of DCs using fluorescein isothiocyanate-dextran, horseradish peroxidase, and lucifer yellow. We found that DCs use two distinct mechanisms for antigen capture. The first is a high level of fluid phase uptake via macropinocytosis. In contrast to what has been found with other cell types, macropinocytosis in DCs is constitutive and allows continuous internalization of large volumes of fluid. The second mechanism of capture is mediated via the mannose receptor (MR), which is expressed at high levels on DCs. At low ligand concentrations, the MR can deliver a large number of ligands to the cell in successive rounds. Thus, while macropinocytosis endows DCs with a high capacity, nonsaturable mechanism for capture of any soluble antigen, the MR gives an extra capacity for antigen capture with some degree of selectivity for non-self molecules. In addition to their high endocytic capacity, DCs from GM-CSF + IL-4-dependent cultures are characterized by the presence of a large intracellular compartment that contains high levels of class II molecules, cathepsin D, and lysosomal-associated membrane protein-1, and is rapidly accessible to endocytic markers. We investigated whether the capacity of DCs to capture and process antigen could be modulated by exogenous stimuli. We found that DCs respond to tumor necrosis factor alpha, CD40 ligand, IL-1, and lipopolysaccharide with a coordinate series of changes that include downregulation of macropinocytosis and Fc receptors, disappearance of the class II compartment, and upregulation of adhesion and costimulatory molecules. These changes occur within 1-2 d and are irreversible, since neither pinocytosis nor the class II compartment are recovered when the maturation-inducing stimulus is removed. The specificity of the MR and the capacity to respond to inflammatory stimuli maximize the capacity of DCs to present infectious non-self antigens to T cells.
Article
Previous studies have shown that following skin sensitization there is an accumulation of dendritic cells (DC) in lymph nodes draining the site of exposure. A significant number of the DC which arrive in the lymph nodes bear high levels of antigen, and the available evidence indicates that they are derived from epidermal Langerhans' cells (LC). Although freshly isolated LC are relatively inefficient antigen-presenting cells, the antigen-bearing DC which are found within draining nodes following skin sensitization are highly immunostimulatory. Recent investigations indicate that the functional maturation of LC as they migrate from the skin is reflected by an enhanced capacity to form stable clusters with lymphocytes, and is associated with an increased expression of membrane major histocompatibility complex (MHC) class II (Ia) antigen. By analogy with in vitro studies of LC maturation, it is possible that such changes are effected by granulocyte/macrophage colony-stimulating factor (GM-CSF) and interleukin-1 (IL-1), both of which are products of epidermal cells. The question remains as to the nature of the stimulus that initiates LC migration. In the present study we have examined in mice the effects of intradermal injection of tumour necrosis factor-alpha (TNF-alpha), another epidermal cytokine, on the accumulation of DC in lymph nodes. Murine recombinant TNF-alpha was found to cause a concentration- and time-dependent increase in the number of DC within draining nodes. Under the same conditions of exposure murine recombinant GM-CSF was without effect. Heat treatment of mouse TNF-alpha resulted in an equivalent inhibition of both DC accumulation and cytotoxic activity measured by in vitro bioassay. An interesting observation was that equal concentrations of human TNF-alpha, of equivalent specific activity, failed to influence the frequency of lymph node DC. These data demonstrate that TNF-alpha induces DC accumulation in draining lymph nodes, and we propose that this cytokine may provide one stimulus for LC migration during cutaneous immune responses.
Article
Virus-induced acquired immune suppression in mice infected with lymphocytic choriomeningitis virus is shown here to be caused by the CD8+-T-cell-dependent elimination of macrophages/antigen-presenting cells. Surprisingly, this is associated with severe destruction of the follicular organization of lymphoid organs, indicating a crucial role for dendritic cells and marginal zone macrophages in maintaining follicular structure. Once established, this immunopathology cannot be readily reversed by the elimination of CD8+ effector cells. Such a T-cell-mediated pathogenesis may play a pivotal role in acquired virus-induced immunosuppression and may represent one strategy by which virus escapes immune surveillance and establishes persistent infections in initially immunocompetent hosts.