ArticlePDF AvailableLiterature Review

Cross-priming in health and disease

Authors:

Abstract

Cross-priming is an important mechanism to activate cytotoxic T lymphocytes (CTLs) for immune defence against viruses and tumours. Although it was discovered more than 25 years ago, we have only recently gained insight into the underlying cellular and molecular mechanisms, and we are just beginning to understand its physiological importance in health and disease. Here we summarize current concepts on the cross-talk between the immune cells involved in CTL cross-priming and on its role in antimicrobial and antitumour defence, as well as in immune-mediated diseases.
Immune defence against intracellular pathogens or
tumours is the domain of CD8+ cytotoxic T lymphocytes
(CTLs). These cells recognize antigenic peptides asso-
ciated with MHC class I molecules, which are expressed
on the surface of all cells in the body. When patho-
gen- or tumour-specific effector CTLs detect antigen
being presented by a cell, they destroy the cell to avert
the spread of infection or cancer. To avoid the killing
of healthy bystander cells that have endocytosed viral
or tumour debris, this endocytosed material does not
usually enter the MHC class I-loading machinery in the
endoplasmic reticulum. The endogenous MHC class I
pathway is reserved for peptides derived from intra-
cellularly synthesized proteins. Therefore, CTL cytotoxic-
ity is focused on cells in which microorganisms replicate
or that are malignant.
However, this restriction of MHC class I loading is
insufficient in one situation: naive CTLs need to be acti-
vated by professional antigen-presenting cells (APCs),
usually dendritic cells (DCs), before they can exert their
cytotoxic effector functions. So when an intracellular
pathogen does not infect APCs or compromises their
endogenous MHC class I pathway, or when a tumour is
not APC-derived (which is the case for most tumours),
CTLs can only be activated if APCs present extracellular
antigen on their MHC class I molecules. This process,
termed cross-presentation, was named after the phenom-
enon of cross-priming that was discovered in the 1970s1,
in which antigens from intravenously injected cells
crossed’ into the MHC class I pathway of host APCs
for CTL priming. Cross-priming has since been shown
to be required for defence against many viruses and
tumours2,3, and it is essential for vaccinations with pro-
tein antigens, which must be cross-presented to activate
CTLs4. Self antigens are also cross-presented but rather
than cross-priming, this normally results in deletion of
autoreactive CTLs, in a process termed cross-tolerance5.
Technical and conceptual advances in recent years
have provided insights into the underlying mechanisms of
cross-priming, especially the associated intracellular anti-
gen processing mechanisms and DC subsets, which have
being reviewed elsewhere4,6,7. The physiological impor-
tance of cross-priming and cross-tolerance in health
and disease is less well understood. Here, we summarize
recent advances in our understanding of the regulation
of cross-priming and its implications for defence against
infections, CTL-mediated diseases and tumours.
Basic mechanisms of cross-priming
Roles of APC types in cross-priming. In viv o depletion or
functional inhibition of DCs compromises both cross-
priming and cross-tolerance8,9, identifying these cells as
the most relevant cross-presenting APCs in mice6,7,10,11.
Among the DCs, only those expressing the surface mark-
ers CD24, CD8α and CD103 (also known as αE integrin)
usually cross-present antigen6,7,10,11. Cross-presenting DC
subsets are usually identified by ex vivo analysis of DCs
isolated from antigen-injected mice, because selective
in v ivo targeting of DC subsets is difficult. The targeting of
DC subsets has become possible by the discovery that the
transcription factor BATF3 (basic leucine zipper tran-
scriptional factor ATF-like 3) is expressed by CD8α+ and
CD103+ DCs and is necessary for cross-presentation12.
Using BATF3-deficient mice, it has been proposed that
these DCs constitute a unified subset with a common
lineage13. Under inflammatory conditions, in mucosal
tissues and in tumours, DCs that lack expression of
CD24, CD8α and CD103 can also cross-present1418.
However, the human counterparts of cross-presenting
mouse DC subsets remain to be identified.
Classifying DC subsets on the basis of cell surface
markers is convenient for flow-cytometric analysis, but
does not mechanistically explain why a cell can cross-
present. Recent studies have proposed two mechanistic
*Institutes of Molecular
Medicine and Experimental
Immunology, University Clinic
of the Rheinische Friedrich-
Wilhelms-Universität,
Sigmund-Freud-Strasse 25,
53105 Bonn, Germany.
School of Medicine and
Pharmacology, Sir Charles
Gairdner Hospital, University
of Western Australia,
Nedlands, Perth 6009,
Australia.
Correspondence to C.K.
e-mail: ckurts@web.de
doi:10.1038/nri2780
Cross-presentation
The ability of certain
antigen-presenting cells to load
peptides that are derived from
exogenous antigens onto MHC
class I molecules. This property
is atypical, because most cells
exclusively present peptides
from their endogenous
proteins on MHC class I
molecules. Cross-presentation
can lead to cross-priming or
cross-tolerance.
Cross-priming
The initiation of an
immunogenic CD8+ T cell
response to an antigen that is
not synthesized by the
antigen-presenting cell.
Cross-presentation is essential
for the initiation of immune
responses to viruses that do
not infect antigen-presenting
cells.
Cross-priming in health and disease
Christian Kurts*, Bruce W. S. Robinson and Percy A. Knolle*
Abstract | Cross-priming is an important mechanism to activate cytotoxic T lymphocytes
(CTLs) for immune defence against viruses and tumours. Although it was discovered more
than 25 years ago, we have only recently gained insight into the underlying cellular and
molecular mechanisms, and we are just beginning to understand its physiological
importance in health and disease. Here we summarize current concepts on the cross-talk
between the immune cells involved in CTL cross-priming and on its role in antimicrobial
and antitumour defence, as well as in immune-mediated diseases.
REVIEWS
NATURE R EVIEWS
|
IMMUNOLOGY VOLUME 10
|
JUNE 2010
|
403
© 20 Macmillan Publishers Limited. All rights reserved10
Cross-tolerance
The antigen-specific
tolerization of CD8+ T cells by
antigen-presenting cells that
cross-present self or innocuous
antigens, which results in
BIM-mediated (BCL-2
inhibitable) deletion of
autoreactive CD8+ T cells.
C-type lectin receptors
A large family of receptors that
bind glycosylated ligands and
have several roles, such as in
cell adhesion, endocytosis,
pathogen recognition, natural
killer cell target recognition and
dendritic cell activation.
explanations, which are not mutually exclusive. The first
hypothesis proposed that only cross-presenting DCs pos-
sess the antigen processing machinery that loads endo-
cytosed antigen onto MHC class I molecules19. Therefore,
based on this hypothesis, cross-presentation must depend
on specific antigen processing mechanisms that can be
regulated separately, so that MHC class I-restricted pres-
entation of endogenous antigens remains operational.
Insulin-regulated aminopeptidase (IRAP; also known as
cystinyl aminopeptidase), a trimming peptidase located
in endosomal compartments, has been recently identified
as a processing molecule with such specificity20; how-
ever, its general role in cross-presentation remains to be
established. The second hypothesis proposed that cross-
presentation depends on distinct endocytosis mecha-
nisms that introduce antigen directly into the organelle
(or organelles) in which cross-presentation occurs21
(BOX 1). In support of this, uptake of antigen for cross-
presentation is restricted to distinct endocytosis receptors,
such as Fc receptors and certain members of the C-type
lectin receptor family, such as C-type lectin 9A (CLEC9A;
also known as DNGR1), CLEC7A (also known as
dectin 1), DC-SIGN (also known as CD209), DEC205
(also known as CD205) and mannose receptor 1 (also
known as CD206)19,2224. These receptors are expressed
by CD8α+ DCs21, providing a possible mechanistic
explanation for why this DC subset can cross-present.
If CTL-mediated immune responses depend on
cross-priming, one might ask why it is restricted to only
a few DC subsets. One explanation is that DCs capable
of cross-presentation automatically become targets for
CTL killing after endocytosis of viral or tumour debris.
Hence, during a systemic infection, all DCs might be
killed if they all could cross-present. Although DCs
possess mechanisms to avoid CTL-mediated killing to
some degree25, it is clear that cross-presenting DCs can
be effectively killed during viral infections26. However,
this killing does not seem to affect the ensuing immune
response as CTLs need less than one day of antigen
presentation by DCs for activation and a little longer for
developing cytolytic functionality27,28, and they can then
carry out their tasks without continual stimulation, at
least for some days. DCs that cannot cross-prime can
maintain other immune responses that depend on CD4+
T helper (TH) cells, such as antibody production and
macrophage stimulation, because they will not be killed
by CTLs, unless they are virally infected themselves.
Consistent with this notion, non-cross-presenting DCs
are superior at activating TH cells19,21.
Cross-priming CTLs specific for peripheral tissue
antigens involves the cooperation between the different
DC types. Tissue DCs transport antigen from tissues to
secondary lymphoid organs and transfer it to resident
CD8α+ DCs, which then cross-prime CTLs29,30. This
antigen transfer mechanism provides several theoretical
advantages7: first, migratory DCs may distribute antigen
to several resident DCs, which together can prime more
CTLs; second, antigen endocytosed by non-cross-present-
ing DCs can also be used for cross-priming; third, non-
cross-presenting DCs carrying viral antigens would not
be killed by viral-specific CTLs before reaching draining
lymph nodes; fourth, viruses may compromise endog-
enous antigen processing pathways in infected migratory
DCs, but not in the antigen-receiving DCs; and fifth, the
CTL will have to inspect only the few antigen-receiving
resident DCs rather than a large number of migratory
DCs, which may allow faster location of their cognate
antigen. However, this concept of antigen transfer also
contains some conceptual problems: even if antigen is
transported by non-cross-presenting DCs, eventually it
must reach a cross-presenting DC, which then might be
killed by CTLs. Furthermore, the information obtained
from direct encounters with microbial molecular patterns
must somehow be transferred to the cross-priming DCs.
Finally, the mechanism of antigen transfer between
DCs is unclear.
In addition to DCs, macrophages, neutrophilic
granulo cytes and mast cells can cross-present antigen but
cannot cross-prime CTLs31. B cells only cross-present
antigen recognized by their immunoglobulin recep-
tors and can cross-prime CTLs when Toll-like receptors
(TLRs) are also engaged32. Resting B cells can tolerize
CTLs when externally loaded with antigenic peptide33,
but evidence for a functional role of these cells in cross-
tolerance in vivo is lacking. By contrast, liver sinusoidal
endothelial cells (LSECs) can efficiently cross-tolerize
CTLs that are specific for food antigens or commensal
bacteria34. In summary, distinct APC types have different
roles in cross-priming and cross-tolerance.
DC activation and DC licensing. Immunogenic responses
require that DCs encounter antigen that is associated
with molecular patterns indicative of the presence of
microorganisms or other ‘danger’ signals35. Sensing such
patterns (for example, by activation of TLRs) leads to the
upregulation of co-stimulatory molecules and enhances
Box 1 | Distinct organelles for cross-presentation in dendritic cells
The cell biological mechanisms involved in cross-presentation have long remained
poorly understood. In particular, it was unclear how antigens may avoid lysosomal
degradation and enter the MHC class I-loading pathway, which is known to start in the
cytoplasm and to occur in the endoplasmic reticulum (ER)4. However, loading in the ER
would imply that the few peptides derived from endocytosed material would have to
compete with the high levels of endogenously produced peptides in the ER for loading
of MHC class I molecules. An elegant solution to this problem came from studies
showing that loading of peptides from particulate antigens occurs in phagosomes and
hence is sequestered from loading of endogenous peptides in the ER144,145. These
organelles were shown to contain the MHC class I-loading machinery and, after
isolation, were sufficient to import and cross-present exogenously added antigenic
peptides. However, it remains controversial how the loading machinery reaches
phagosomes146. During maturation, phagosomes fuse with lysosomes, lower their pH
and become competent for peptide loading onto MHC class II molecules147.
Cross-presentation of soluble antigens can occur in stable early or recycling
endosomes that do not mature to late endosomes, in which peptides are loaded on
MHC class II molecules21,38,148,149. These early endosomes are supplied with antigen by
distinct endocytic mechanisms, such as that mediated by the mannose receptor,
whereas late endosomes received antigen by other means. According to this model,
cross-presentation and MHC class II-restricted presentation of soluble antigens are
physically separated, whereas MHC class I and II loading of particulate antigens is
temporally separated in the same phagosomes. For both antigen classes,
cross-presentation is sequestered from MHC class I loading of endogenous peptides,
avoiding competition of peptides for MHC class I loading.
REVIEWS
404
|
JUNE 2010
|
VOLUME 10 www.nature.com/reviews/immunol
© 20 Macmillan Publishers Limited. All rights reserved10
NKT cell
Nature Reviews | Immunology
MHC class II
MHC
class II
MHC class I
TCR
TCR
CD40L
IL-2
CD80 or
CD86 CD28
CD27
PDL1
TRAIL
expression
TLR agonist
CD70
CD4+ TH cell
CD4+ TH cell
CD8+ CTL
CD8+ CTL
CCL3, CCL4
and CCL5 CCR5
Cross-presenting
DC
TLR
Antigen
a
b
CD40
TLR
CD1d
TCR
α-GalCer
Antigen
CCR4
CCL22
TCR
CCL17
cross-priming; the enhancement of cross-priming being
especially effective after TLR3 and TLR9 stimulation36,37.
Furthermore, TLR signalling enhances peptide loading
onto MHC class I molecules by inducing the relocation
of components of the peptide-loading machinery to the
endosomes in which cross-presentation occurs38. Many
C-type lectin receptors preferentially recognize foreign
glycosylation structures characteristic of microorganisms
and thereby allow some degree of self–foreign discrimi-
nation21,39. Engaging these receptors may synergize with
TLR signals to promote upregulation of co-stimulatory
molecules because distinct signalling pathways are used40,
and may theoretically enhance cross-priming. However,
this cannot be extrapolated from studies targeting anti-
gens to C-type lectin receptors, because such targeting
enhances cross-priming by increasing antigen uptake,
which is difficult to discriminate from C-type lectin recep-
tor signalling. Also, activation of RIG-I-like cytoplasmic
receptors for nucleic acids and the inflammasome boost
CTL responses41,42 and indirect evidence suggests that
these cytoplasmic sensors can stimulate cross-priming43,44,
but this has not yet been formally shown.
Although co-stimulatory signals are necessary for
immunogenic cross-priming, they are insufficient in
some models to break cross-tolerance45,46, unless specific
TH cells are present45,47. This has led to the realization that
DCs require signals from specific TH cells for immuno-
genic cross-priming. CTLs activated without T cell help
(termed ‘helpless’ CTLs) lack expression of specific anti-
apoptotic molecules, have a short life-span and cannot
carry out cytotoxic effector functions5,48–50. The neces-
sity for specific T cell help can be viewed as requesting
a ‘second opinion’ from TH cells before programming
CTLs for cytotoxic functions. Both autoreactive TH cells
and CTLs are present at a low frequency in a normal
repertoire, but the likelihood that clones specific for the
same autoantigen coexist will be much lower. Hence,
autoimmune responses become less likely when antigen
must be recognized by two lymphocyte subsets that had
been negatively selected for self-reactivity. An analogous
safety strategy is implemented in antibody production, in
which specific B cells and TH cells need to both recognize
the foreign antigen. However, in contrast to B cells, CTLs
lack MHC class II molecules and cannot directly receive
specific help from TH cells. Therefore, cross-presenting
DCs act as a temporal bridge between these cells. The
TH cells convert the DCs to a transient state in which they
can programme the CTL for sustained cytotoxic effec-
tor functions and memory differentiation (FIG. 1a)51,52.
When such DCs are adoptively transferred into naive
mice, they retain the ability to cross-prime CTLs, show-
ing that they store the information received from the
TH cells. This has been termed ‘DC licensing’.
DC licensing is usually facilitated by TH cell-expressed
CD40 ligand (also known as CD154), which interacts
with CD40 on DCs31. It remains unclear to what extent
signalling events induced by TH cells or by TLRs over-
lap in cross-priming DCs. Some differences must exist,
because TH cells and pattern-recognition receptors
synergistically activate DCs and both are necessary for
optimal cross-priming4,45.
Figure 1 | Molecular mechanisms of licensing dendritic cells for classical
cross-priming. a | The molecular mechanisms involved in classical cross-priming
are illustrated. Dendritic cells (DCs) take up antigen by distinct endocytosis
mechanisms (not shown) and present it to CD4+ T helper (TH) cells through MHC
class II molecules and cross-present it to CD8+ cytotoxic T lymphocytes (CTLs)
through MHC class I molecules. Activated CD4+ TH cells can stimulate CTLs through
the production of interleukin-2 (IL-2) and license DCs for cross-priming through
CD40 ligand (CD40L)–CD40 interactions. Licensed DCs upregulate expression of
co-stimulatory molecules, such as CD70, CD80 and CD86, and downregulate
inhibitory molecules, such as programmed cell death ligand (PDL1). Toll-like
receptor (TLR) ligands further activate DCs and increase their cross-presentation
activity. Cross-primed CTLs are programmed for survival and cease TNF-related
apoptosis-inducing ligand (TRAIL) production. ‘Helpless’ CTLs activated by
unlicensed DCs die following secondary encounter with antigen in their effector
phase (not shown). b | Chemokine-mediated regulation of cross-priming is
illustrated. CD4+ TH cells, and the DCs they license, produce CC-chemokine ligand 3
(CCL3), CCL4 and CCL5 in the presence of TLR ligands, which recruit naive CTLs for
classical cross-priming. Alternatively, DCs licensed by natural killer T (NKT) cells
produce the CC-chemokine receptor (CCR4) ligand CCL17, and NKT cells
themselves produce the CCR4 ligand CCL22, which recruit naive CCR4+ CTLs for
cross-priming. The CCR4- and CCR5-mediated recruitment pathways are
synergistic. In this figure, dashed arrows indicate antigen routing for cross-
presentation. α-GalCer, α-galactosylceramide; TCR, T cell receptor.
REVIEWS
NATURE R EVIEWS
|
IMMUNOLOGY VOLUME 10
|
JUNE 2010
|
405
© 20 Macmillan Publishers Limited. All rights reserved10
Inflammasome
A large multiprotein complex
formed by a nucleotide-binding
domain (NBD)-, leucine-rich
repeat (LRR)-containing family
(NLR) protein, the adaptor
protein apoptosis-associated
speck-like protein containing a
caspase recruitment domain
(ASC) and pro-caspase 1. The
assembly of the inflammasome
leads to the activation of
caspase 1, which cleaves
pro-IL-1β and pro-IL-18 to
generate the active
pro-inflammatory cytokines.
DC licensing
A concept that DCs must
be converted by an
antigen-specific T helper cell
into a functional state required
for immunogenic activation of
CTLs, which decreases the
likelihood of autoimmunity, but
requires coordination of cell
encounters by chemokines.
CTL programming
A concept that CTLs receive
additional information during
their activation that affects
their effector functions,
life-span, memory
differentiation or migratory
properties. These signals
determine whether
cross-presentation leads
to cross-priming or
cross-tolerance.
CTL programming. Licensed cross-presenting DCs
provide signals that dictate whether cross-priming or
cross-tolerance ensues, a process termed ‘CTL program-
ming27,28,53. Understanding the identity and regulation
of these programming signals is important for optimiz-
ing vaccinations and understanding autoimmunity, and
hence they have been intensely studied. As candidate
signals, altered antigen presentation and modified co-
stimulatory and/or co-inhibitory signals have been pro-
posed. The duration of T cell receptor (TCR) signalling
affects whether activated CTL survive or not27,28, whereas
the TCR affinity dictated the level of CTL expansion
and ensured that only high affinity CD8+ T cell clones
develop into memory CTLs54. It is unknown, however,
how these qualities of TCR signalling may be regulated
by the DCs and by licensing TH cells.
CD70 is a candidate co-stimulatory signal for CTL
programming. This activation-induced tumour necro-
sis factor (TNF) receptor family molecule interacts
with CD27, which is expressed by CTLs, and increases
their survival17,55. However, the expression of CD70 by
DCs is not regulated by TH cells56. Downregulation of
programmed cell death ligands (PDLs), which bind to
the co-inhibitory receptor PD1 of the CD28 family and
promote CTL tolerance, might also contribute to CTL
programming57. It is unclear whether these ligands are
regulated by TH cells. The expression of interleukin-12
(IL-12) by DCs is regulated by TH cells and TLR ligands,
and promotes CTL effector functions58; however, its
ability to programme CTLs for survival is unclear.
CTL survival can be programmed by IL-2 during
priming59. However, IL-2 is not thought to be produced
by DCs and therefore cannot be the elusive survival
signal that licensed DCs provide. Theoretically, it may
originate from TH cells, but this would require their pres-
ence during CTL priming, which is inconsistent with
DCs storing the licensing information. Furthermore,
TH cell-derived IL-2 seems to have a distinct, probably
antigen-nonspecific, role in CTL memory maintenance53.
Alternatively, IL-2 may be produced by the CTL them-
selves. Indeed, prolonged TCR signalling during priming
induces autocrine IL-2 production60, and such IL-2 can
sustain CTL survival, at least when the CTLs are present
at high numbers61. Also, CD70–CD27 signals upregulate
autocrine IL-2 secretion by CTLs62, whereas it is down-
regulated by PD1 ligation63, suggesting that these two
candidates for the DC-derived CTL survival signal may
operate indirectly by regulating autocrine IL-2 produc-
tion. A converse effect on CTL survival has been reported
for autocrine TNF-related apoptosis-inducing ligand
(TRAIL), which CTLs produce by default and which is
shut off by specific TH cells64. Thus, helpless CTLs may
use TRAIL to kill themselves or each other if they also
express the TRAIL receptor. However, IL-2 does not
inhibit TRAIL production65, and causal connections to
CD70 and PD1 ligands have not yet been established. In
conclusion, many mechanistic steps important in cross-
priming have been described, but a comprehensive theory
of the molecular mechanisms by which licensed DCs pro-
gramme CTLs for survival, cytotoxic effector function and
memory generation that links these steps is lacking.
The cellular encounters for cross-priming
CTL activation in lymphoid organs. The need for DC
licensing by TH cells has an important limitation: three
rare immune cells — the cross-presenting DC, the anti-
gen-specific TH cell and the CTL — need to all interact.
However, because the licensing information is stored
within DCs, these interactions may not necessarily occur
at the same time. The CTL may theoretically interact
with the licensed DC after the DC has interacted with
the licensing TH cell51. Nevertheless, relying on random
encounters during normal T cell recirculation would
probably be too time-consuming to guarantee timely
defence against rapidly replicating microorganisms.
Recent studies showed that the encounters necessary
for CTL activation are greatly enhanced by chemokines.
In particular, CC-chemokine receptor 5 (CCR5) ligands
are produced following DC–TH cell interactions under
inflammatory conditions, which were mimicked by the
injection of TLR ligands66 (FIG. 1b). These chemokines
allowed CCR5-expressing naive CTLs to be attracted
to licensed DCs, where they could scan for cognate
antigen. Although TH cells and DCs first need to find
each other, the subsequent attraction of the third rare
cell type is thereby greatly enhanced and this decreases
the time spent by naive CTLs scanning unlicensed DCs.
Once activated, CTLs also produce CCR5 ligands67,
which may guide other naive CTLs towards DCs that
have successfully cross-primed. CTLs also produce
XC-chemokine ligand 1 (XCL1; al so known as lympho-
tactin) that binds to XC-chemokine receptor 1 (XCR1),
which is selectively expressed by CD8α+ DCs, and this
interaction supports cross-talk between these cell types
and enhances cross-priming68.
Natural killer T (NKT) cells, which recognize glyco-
lipid antigens presented by CD1d molecules, can also
enhance cross-priming, for example by upregulating
co-stimulatory molecules on DCs69. This process has
been recently shown to involve cognate DC licensing70
and consequently a tripartite immune cell encounter.
Chemokines, specifically CCR4 ligands and espe-
cially CCL17 (also known as TARC), produced by
cross-priming DCs are instrumental in this setting70
(FIG. 1b). Thus, at least two chemokine systems recruit
naive CTLs towards DCs that have been licensed by
TH cells or NKT cells. The CTL chemotaxis mediated
by CCR4 and CCR5 is synergistic, providing a mecha-
nistic rationale for combining ligands for NKT cells
with TLR ligands plus TH cell epitopes in vaccination
strategies. However, it remains unclear how naive
CTLs are induced to express CCR4 or CCR5 before
they interact with DCs that were licensed by NKT or
TH cells, respectively.
Regulation of effector CTLs in tissues. DCs can also
regulate the CTL effector phase in tissues50,71,72. If DCs
are infected, they may do so by using the endogenous
MHC class I pathway, unless it has been compromised by
the infection. If tissue DCs are not infected, they would
need to cross-present the antigen to ensure the antigen
specificity of this regulation. However, tissue DCs are
usually of the CD8αCD11b+ subset, which has been
REVIEWS
406
|
JUNE 2010
|
VOLUME 10 www.nature.com/reviews/immunol
© 20 Macmillan Publishers Limited. All rights reserved10
Nature Reviews | Immunology
CD8+
CTL
CXCR3
or CCR5
CXCR3 or
CCR5 ligand
Blood vessel
Endothelial
cell
LFA1
ICAM1
Virus-infected
tissue cell
Cytokines
and interferons
Virus
CD8+
CTL
CD4+
T cell
MHC
class I
MHC
class II
TCR
Tissue
DC
shown to be unable of cross-presentation, at least under
steady-state conditions and in v itro6,7. If such DCs could
acquire this ability by inflammation-induced redifferen-
tiation into cross-presenting cells (for example CD8α+
DCs), the ability to cross-present would essentially be
a functional state that can be regulated, rather than a
property of DC subsets as entities with distinct line-
age. Recently, CD8αCD103+ DCs have been detected
in certain tissues, which are developmentally related
to CD8α+ DCs13, and can cross-present tissue antigens
after migrating into draining lymph nodes11. However,
it is unclear if these cells can cross-present antigen while
in tissues. Thus, it is currently undecided whether DCs
within non-lymphoid tissues can regulate effector CTLs
in an antigen-specific manner.
An alternative mechanism by which tissue DCs could
regulate infiltrating effector CTLs in an antigen-specific,
but non-MHC class I-restricted, manner involves the
presentation of antigen on MHC class II molecules to TH
cells, which results in the production of cytokines and
chemokines that recruit and/or regulate CTLs (FIG. 2). This
has been described in mouse models of mesotheli oma73,
immune-mediated kidney disease50 and genital herpes
simplex virus (HSV) infection72.
There is also an antigen-specific and MHC
class I-restricted CTL recruitment mechanism. This is
not facilitated by DCs, but instead by cross-presenting
endothelial cells. Such cells have been detected in pancre-
atic islets74, the central nervous system75 and the liver76,
and have been shown to establish firm T cell contact
and activation, which in turn will increase lymphocyte
function-associated antigen 1 (LFA1; also known as αLβ2
integrin)–intercellular adhesion molecule 1 (ICAM1)-
mediated transmigration thorough endothelial barriers
into tissues. Subsequently, tissue-resident DCs may then
regulate the incoming CTLs; for example, by cross-talk
wit h TH cells and chemokine production. However, cross-
presenting endothelial cells might be killed by CTLs,
causing unwanted side-effects. This may be avoided by
high levels of co-inhibitory molecule expression, such as
PDL1, by the endothelial cells, which could help prevent
CTL-mediated damage77. Thus, in addition to the classical
paradigm of CTL recruitment into tissues by attachment
to adhesion molecules on inflamed endo thelial cells,
two specific pathways exist that are facilitated by cross-
presenting endothelial cells or by chemokines resulting
from specific TH cell–DC crosstalk (FIG. 2).
Cross-priming during infections
Viral infections. Cross-priming is necessary for CTL-
mediated immune responses against microorgan-
isms that do not target DCs, such as Epstein–Barr
virus (EBV), hepatitis B virus (HBV) or poliovirus
(TABLE 1). Furthermore, cross-priming is important for
DC-infecting viruses that evade the endogenous MHC
class I pathway, such as members of the Herpes virus
family, especially murine cytomegalovirus (MCMV).
However, many viruses infect DCs, suggesting that
avoiding DCs is not a survival advantage for the virus,
perhaps because cross-priming exists.
MCMV infects various cells, including DCs, and
produces immune evasins that inhibit MHC class I-
restricted antigen presentation78. Hence, MCMV-infected
DCs fail to induce a virus-specific CTL-mediated
immune response79. Despite escaping from the
endogenous antigen presentation pathway, MCMV-
specific CTLs eventually develop, demonstrating
that cross-priming by uninfected DCs must occur80.
This pathway also induces CTL-mediated immunity
against other immune evasin-producing viruses, such
as EBV81 and HSV-1 (REF. 7). But even when effective
cross-priming occurs, MCMV-infected cells still
escape CTL-mediated killing because these cells present
viral epitopes that are different from those that the
CTLs are cross-primed against80. Interestingly, ablation
of immune evasins from MCMV results in decreased
cross-priming and antiviral immunity82. This paradoxi-
cal observation can be explained by the rapid control
of viral replication and decreased viral antigen burden
in the absence of these molecules. Other viruses also
directly avoid cross-priming: vaccinia viruses escape DC
cross-priming by restricting late viral antigens to a partic-
ular cellular compartment that is inaccessible for cross-
presentation but accessible for MHC class II-restricted
antigen presentation83.
Figure 2 | Recruitment of cross-primed effector cytotoxic T lymphocytes into
non-lymphoid tissues. Viral infection of tissue cells leads to their secretion of
pro-inflammatory cytokines and interferons that upregulate the expression of adhesion
molecules, such as intercellular adhesion molecule 1 (ICAM1), by endothelial cells.
Effector cytotoxic T lymphocytes (CTLs) attach to these molecules and are
nonspecifically recruited from the bloodstream into non-lymphoid tissues. In addition,
recent studies have revealed two antigen-specific recruitment mechanisms: first,
endothelial cells in certain tissues, such as the liver, pancreatic islets or the brain, can
cross-present microbial antigen, which allows them to selectively recruit antigen-specific
CTLs. Second, when specific CD4+ T helper (TH) cells detect microbial (or self) antigen on
non-cross-presenting tissue DCs, ligands for CC-chemokine receptor 5 (CCR5) or
CXC-chemokine receptor 3 (CXCR3) are produced that recruit CTLs into the infected
tissue. LFA1, lymphocyte function-associated antigen 1; TCR, T cell receptor.
REVIEWS
NATURE R EVIEWS
|
IMMUNOLOGY VOLUME 10
|
JUNE 2010
|
407
© 20 Macmillan Publishers Limited. All rights reserved10
Table 1 | Cross-priming of CTLs in viral infections, tumours and CTL-mediated diseases
Disease Finding Refs
Viral infections
Influenza virus Cross-priming by distinct DC subsets at different time points after infection 17,55
EBV Cross-priming induces CTLs against latency-associated antigens expressed in B cells 81
Poliovirus Virus-specific immunity requires cross-priming by bone marrow-derived APCs 3
Lymphocytic choriomeningitis virus Type I IFNs induces virus-specific cross-priming 84
HIV Cross-priming of vaccines induces CTLs specific for HIV components 153
Herpes simplex virus 1 Requirement of DC licensing; division of labour between migratory DCs, lymph
node-resident cross-priming CD8α+ DCs and CD103+ DCs that migrate and
cross-present
7,11,29,51
Coxsackie virus Viral evasion of DC cross-priming 154
Murine cytomegalovirus Presentation of distinct epitopes in infected tissue allows for immune escape despite
successful cross-priming
80
HCV NS3-specific cross-priming following uptake of apoptotic infected cells 155
HBV Cross-priming of circulating viral antigens 86,87
Vaccinia virus Antigen transfer from infected cells enables cross-priming 156
Adenovirus Antigen transfer from infected macrophages to CD8α+ DCs in cross-priming 30
Non-viral infections
Listeria monocytogenes Essential role of cross-priming for bacteria-specific immune responses 9
Plasmodium spp. DCs cross-priming contributes to CTL-mediated immunity; also promoting
experimental cerebral malaria after brain infiltration of cross-primed CTLs
157
Trypanosoma cruzi DC cross-priming confers protection 158
Mycobacteria DC cross-priming after uptake of apoptotic infected cells 94
Chlamydia trachomatis Cross-priming requires antigen from infected cells 159
CTL-mediated diseases
Type 1 diabetes Central and peripheral cross-tolerance; cross-priming of diabetogenic CTLs 5,97,103,104
Multiple sclerosis Central and peripheral cross-tolerance; cross-priming of encephalitogenic CTLs; role of
cross-presenting endothelial cells
75,111,112,160
Glomerulonephritis and interstitial
nephritis
Activation of nephritogenic CTLs 50,108
Psoriasis vulgaris Activation of CTLs specific for skin autoantigens 109
Ankylosing spondylitis Epidemiological link to EBV infection 114
Autoimmune hepatitis Potential mimicry of EBV and HCV epitopes that are cross-primed; potential role of
cross-presenting endothelial cells
34,115,116
Thyroiditis Vaccination of mice with thyroglobulin caused CTL-mediated thyroiditis 161
Transplant rejection Cross-priming of host CTL specific for graft antigens 117
Tumours
Mesothelioma Weak cross-priming of localized CTLs 124
Melanoma and thymoma Cross-priming restricted by poor TH cell response 123
Leukaemia and lymphoma Cross-tolerance induction 2,162
Transformed embryo cells Cross-presenting DCs can be activated to cross-prime 52
Melanoma Cross-priming is autophagy dependent 163
Hepatocellular carcinoma Cross-priming in an autochthonous mouse model 122
Lung cancer Efficient cross-priming but no tumour eradication 125
Glioma of the brain Cross-primed CTLs enter and are retained in brain tumours 75
Insulinoma Weak cross-priming increasing with tumour size 127
APC, antigen-presenting cell; CTL, cytotoxic T lymphocyte; DC, dendritic cell; EBV, Epstein–Barr virus; HBV, hepatitis B virus; HCV, hepatitis C virus; IFN, interferon;
TH cell, T helper cell.
REVIEWS
408
|
JUNE 2010
|
VOLUME 10 www.nature.com/reviews/immunol
© 20 Macmillan Publishers Limited. All rights reserved10
Central tolerance
Self-tolerance that is created at
the level of the central
lymphoid organs. Developing
T cells in the thymus and
B cells in the bone marrow that
strongly recognize self antigen
face deletion or marked
suppression.
Antigenic mimicry
A mechanism for the induction
of autoimmunity, in which a
pathogen expresses a protein
or peptide that is similar to a
self protein. After the induction
of a pathogen-specific immune
response, a cross-reactive
response to self results in
autoimmune pathology.
Type I interferons (IFNs) are released from virus-
infected cells or sentinel cells such as plasmacytoid DCs,
and they can induce DC maturation and support cross-
priming. This may be important when neither viral nor
microbial-derived signals are available to induce the
maturation of cross-priming DCs84. Type I IFNs are effi-
ciently induced by triggering TLR3, which recognizes
double-stranded viral RNA and effectively stimulates
cross-priming37. TLR3-induced type I IFN expression
is associated with control of viral infection, albeit at the
expense of immune-mediated organ damage85. Despite
these mechanisms, some viruses still do not elicit effi-
cient CTL responses despite abundant antigen produc-
tion. For example, hepatitis C virus (HCV) is recognized
by RIG-I, but blocks the subsequent induction of type I
IFNs, and HBV induces the production of IL-6 but not
type I IFNs by infected cells86,87. The efficacy of type I
IFN treatment in patients with chronic viral hepatitis
may involve improved cross-priming, in addition to any
direct antiviral effect.
Viruses such as HIV, influenza virus or HCV can
escape established CTL responses by mutating their
antigens (TABLE 1). Furthermore, during the course of
an infection, different viral antigens are expressed, such
as the early and late antigens of HSV. Thus, continuous
delivery of viral antigens is necessary to cross-prime CTL
clones specific for new antigens in lymph nodes. Such
antigen delivery can be mediated by migratory DCs29 or
by the endocytosis of viral antigens that drained though
lymph vessels by lymph node-resident DCs88. In HSV
infection of the skin or lung, antigen is transported by
migratory CD103CD11bhi tissue DCs (or by Langerhans
cells in the skin) to draining lymph nodes and trans-
ferred to cross-presenting CD8α+ DCs17, or is conveyed
by migratory CD103+CD11b DCs that directly cross-
present in draining lymph nodes7. By contrast, migratory
CD103CD11bhi DCs transported viral antigen during
influenza virus infection from the lung to the bronchial
lymph nodes and directly cross-primed CTLs without the
need to transfer antigen to CD8α+ DCs 17. The role of these
DC subsets in other viral infections is unclear.
Bacterial infections. The importance of cross-priming
for bacterial infection has been mostly studied using
Listeria monocytogenes. Following infection, L. mono-
cytogenes gains access to the cytoplasm of splenic mac-
rophages and hepatocytes, which subsequently undergo
apoptosis. DCs acquire antigen for cross-priming fol-
lowing uptake of debris of infected cells, which is cru-
cial for defence against L. monocytogenes infection9.
Cross-priming can also occur in the absence of protein
synthesis, on the condition that the innate immune
stimulatory function of L. monocytogenes is preserved89.
Surprisingly, cross-priming DCs are also important for
bacterial dissemination into the spleen, as they take up
circulating bacteria and allow their initial intracellular
survival, thereby promoting subsequent bacterial pro-
liferation and spread to other cell populations90. This
may localize infections to sites where DCs can later
orchestrate innate and adaptive immune responses91.
Shortening the time of bacterial exposure curtailed
specific immunity by decreasing TH cell responses92,
showing the importance of DC licensing, which requires
time-consuming establishment of physical contact with
licensing TH or NKT cells. Terminating CTL immunity
against infections that do not provide stimulation for a
minimum time (that is, are eliminated by innate immune
mechanisms) may focus adaptive immune responses
to those infections requiring CTL immunity but may
also allow pathogen escape mechanisms to establish
persistent infection.
Acquisition of bacterial antigen by DCs has been
most intensively studied during mycobacterial infection,
in which mycobacteria persist in phagosomal vesicles
and dispersion of bacterial antigens into the cytoplasm
does not occur. Here, apoptosis of infected cells, facili-
tating the uptake of bacterial antigens, is indispensable
for cross-priming by DCs and induction of efficient
CTL-mediated immunity93. Similar to viral infections,
transport of bacterial antigens by DC into lymph nodes
was also required for cross-priming94. However, process-
ing of bacterial antigens from apoptotic vesicles in DCs
differs from that of viral antigens by requiring saposins,
which open vesicles and release bacterial antigens into
the cytoplasm for processing94. Taken together, DC
cross-priming supports the induction of CTL-mediated
immunity against pathogens with a narrow host cell
tropism that avoid infecting DCs or possess immune
escape mechanisms.
Cross-priming in immune-mediated diseases
Although central tolerance is an efficient process, some
autoreactive CTLs escape negative selection and enter
the circulation. In secondary lymphoid organs, cross-
tolerance serves as a second checkpoint that can eliminate
such escapees5,8. However, cross-tolerance occurs only
when the autoantigen dose and/or CTL affinity are suffi-
ciently high31,49. If not, then autoreactive CTLs may escape
cross-tolerance and cause disease when cross-primed95.
CTLs are the main effector cells of human type 1
diabetes96. Cross-priming is necessary and sufficient for
islet infiltration and destruction in transgenic diabetes
models31,49 and in non-obese diabetic (NOD) mice97,
which have multigenic disease susceptibility similar to
human illness, including DC defects that compromise
cross-tolerance98. However, the identity of the antigens
that are cross-presented is not clear. Theoretically, these
antigens could be pancreatic self antigens or microbial
antigens mimicking such antigens. The suggestion of a
role for microbial antigens is supported by studies with
transgenic mice expressing viral components in islets,
which developed CTL-mediated diabetes after virus
infection99. However, the lymphocytic choriomenin-
gitis virus used in these studies infects DCs, which
then directly prime CTLs, and therefore cross-priming
becomes unnecessary. Antigenic mimicry was suggested
to be supported by the epidemiological association of
diabetes with coxsackie virus infections, but this is now
thought to result from changes in the micromilieu of
infected islets rather than viral antigens99,100. Thus, the
evidence for antigenic mimicry as a cause of type 1
diabetes is unconvincing101.
REVIEWS
NATURE R EVIEWS
|
IMMUNOLOGY VOLUME 10
|
JUNE 2010
|
409
© 20 Macmillan Publishers Limited. All rights reserved10
CD8+
CTL
DC
Lymph node
Cross-priming
by CD40
agonist and TLR7
Cross-presentation
by chemotherapy, monoclonal
antibodies and vaccines
Circulation
Antigen
release
Traffic into
tumour
Tumour attack
MHC
class I
Tumour
Blood vessel
Tumour
TCR
Tumour-derived
antigen
Tumour-specific CTL
Nature Reviews | Immunology
Oligodendrocyte
A type of glial cell that creates
the myelin sheath that
insulates axons and improves
the speed and reliability of
signal transmission by neurons.
If so, then diabetogenic CTLs are likely to be cross-
primed by pancreatic islet autoantigens. One candidate
is an MHC class I-binding peptide from the insulin
B chain102, an antigen also relevant in NOD mice103.
Pancreatic autoantigens are cross-presented in the pan-
creatic lymph nodes and this normally leads to cross-tol-
erance5,104, unless specific TH cells are present31. Notably,
TH cells specific for pancreatic autoantigens are also
activated at this site105 and might license DCs for cross-
priming. This may be important in patients with type 1
diabetes, because many of them possess particularities
in MHC class II-restricted antigen presentation and in
the responding TH cells106. Diabetes has long been known
to be closely associated with the MHC class II loci107,
although MHC class I-restricted CTLs are the effectors
of disease. The abnormal TH cells of patients with diabetes
might function by licensing DCs in the pancreatic lymph
node, which then cross-prime diabetogenic CTLs.
Most concepts on the role of CTLs and cross-priming
in autoimmunity have been extrapolated from type 1
diabetes models. However, the rapid destruction of
pancreatic islets has hampered investigating intra-organ
cross-talk between immune cells during the CTL effector
phase. Work in autoimmunity models of the kidney and
skin, which are more resistant to CTL-mediated killing of
tissue cells, has provided new insights. Autoreactive CTLs
specific for kidney glomerular antigens are cross-primed
in the renal lymph node and are recruited by chemo-
kines into the kidney, where they inflict damage50. These
chemokines were produced following cross-talk of auto-
reactive TH cells with tubulointerstitial DCs presenting
renal autoantigen, explaining the long-known ability of
TH cells to facilitate kidney access of autoreactive CTLs;
for example, in interstitial nephritis108. TH cell-dependent
recruitment of autoreactive effector CTLs was also noted
in psoriasis109.
In multiple sclerosis research, CTLs are being redis-
covered as important effectors110. Myelin-specific CTLs
that escape thymic tolerance and cross-tolerance mech-
anisms can damage oligodendrocytes111,112. Recent work
in humanized mice has shown that several classes of
bacteria contain an MHC class I-binding peptide that
mimics myelin basic protein, a candidate autoantigen
in multiple sclerosis113. But as with type 1 diabetes, the
exact autoantigens in multiple sclerosis are unknown,
hampering elucidation of the role of cross-priming and
of antigenic mimicry.
CTL-mediated responses against EBV have been linked
with ankylosing spondylitis114, long known to be associated
with HLA-B27 and with autoimmune hepatitis115. Hepatic
autoantigens are also recognized by HLA-A-restricted
CTLs specific for HCV116. In organ transplantation, host
CTLs cross-primed against graft antigens will be restricted
to the host haplotype, but nevertheless can damage graft
cells, either by alloreactivity or by killing host endothelial
cells growing in the graft117. There are further examples for
associations between disease entities and CTLs (TABLE 1),
but in nearly all cases, a causal role of cross-priming
remains to be formally shown.
Cross-priming and tumours
There are notionally three different types of tumour
antigens: ‘self’ antigens, to which a person is partially
tolerant (for example, normal embryonic or differen-
tiation antigens that also happen to be expressed in
tumours), ‘neo-antigens’ to which the host is not toler-
ant (for example, those due to mutations and viruses)
and ‘potential’ antigens, which are epitopes that may be
unmasked by treatments such as chemotherapy, which
increases antigen delivery into the cross-presentation
pathway118. Tumours contain thousands of potentially
strongly immunogenic neo-antigens caused by muta-
tions119. Several studies have shown that cross-presen-
tation of these antigens is an efficient and continuous
process120; therefore, other reasons as to why the tumour
cells are not rejected must exist. Several experimental
procedures have been used to analyse cross-priming in
tumour models2,120123, and these studies have confirmed
that cross-presentation of tumour antigens occurs, it per-
sists for the dominate antigen and, interestingly, tends to
remain localized to the tumour-draining lymph node120.
Within these lymph nodes, both CD8α+ and CD8α DCs
cross-present tumour antigens12,123, although whether
this reflects multiple mechanisms for delivery of tumour
antigen into the cross-presentation pathway, such as
through Fc receptor-mediated uptake of endogenous
antibodies that have opsonized tumour cells, or other
aspects of tumour antigen delivery, is not known.
Figure 3 | Cross-priming and immunotherapy in an effective antitumour immune
response. The first step in any successful antitumour immune attack usually involves
cross-presentation of antigens released from tumours, a process that can be boosted by
chemotherapies, monoclonal antibodies and vaccines (with or without dendritic cells
(DCs) or heat shock proteins). However, this cross-presentation of tumour antigen is not
sufficient to eradicate tumours; DC activation is required to turn cross-presentation into
a cross-priming event, a process boosted by agents such as CD40 agonists and Toll-like
receptor 7 (TLR7). Additional steps are then required; for example, expansion and
circulation of antitumour-specific effector cytotoxic T lymphocytes (CTLs), entry of those
effector cells into the tumour site and attack of the tumour — each step being subject to
regulation. Therefore, boosting cross-presentation and/or cross-priming alone would be
insufficient for tumour eradication: scientifically validated therapeutic combinations will
be required to turn cross-priming into successful anticancer immunotherapy.
REVIEWS
410
|
JUNE 2010
|
VOLUME 10 www.nature.com/reviews/immunol
© 20 Macmillan Publishers Limited. All rights reserved10
Although in some tumour models cross-tolerance
occurs, in others a weak, ineffective and localized CTL
response is induced124. A strong CTL response rarely
results from tumour antigen cross-priming without
further manipulation125. These weak responses are
understandable, as many tumour tissues lack intense
inflammation and/or danger signals and do not contain
pathogen-associated molecular patterns that usually
drive a strong CTL response126. The efficiency of cross-
priming to tumour antigens probably depends on the
cell line and/or tissue of origin plus other factors, such
as antigen dose127 (TABLE 1).
In summary, tumour antigens are cross-presented
efficiently in draining lymph nodes but fail to elicit a
strong antitumour CTL response.
Cross-priming and cancer treatment
It is in cancer therapy that cross-priming has triggered
the most interest. For example, one of the main mecha-
nisms whereby chemotherapy works is by stimulating
antitumour immunity by increased cross-presentation
and cross-priming of antitumour CTLs128. Importantly,
different cancer chemotherapies kill tumour cells
by different mechanisms, not all of which are immuno-
genic. Certainly it seems that the induction of apoptosis
by cancer chemotherapies can increase the amount of
tumour antigen delivered into the cross-presentation
pathway128. The outcome, however, depends on the
effects of chemotherapy on the host immune response
and importantly, the way in which chemotherapy
kills the tumour cell129131. Increased cross-priming of
tumour-specific CTLs induced by chemotherapy has
been shown to facilitate long term cures of advanced
solid, non-immunogenic tumours when combined with
appropriate immunotherapies132 (FIG. 3).
Our knowledge of cross-presentation will also affect
cancer surgery. In almost all published studies, the
cross-presentation of tumour antigens is restricted to
the lymph node that drains the tumour. Indeed, this may
generate a level of CTL activity within the local ‘sentinel’
lymph node that represents a first ‘checkpoint’ in stop-
ping tumour spread124. Therefore, it has been proposed
that the removal of tumour-draining lymph nodes, even
if they do not show evidence of tumour spread, may
have a negative effect on antitumour immunity.
It has been shown that stronger antitumour effects
are seen if cross-priming of CTLs to tumour antigens is
increased, for example by using adjuvants such as isco-
matrix133, viral vectors134, heat shock proteins135, endog-
enous danger signals such as high-mobility group box 1
(HMGB1)136 or uric acid crystals43,44, or by using mono-
clonal antibodies. Indeed, it is possible that one way in
which monoclonal antibodies work in cancer immuno-
therapy is by opsonizing tumour cells and increasing their
delivery into the cross-priming pathway137. New strate-
gies are also being developed for linking antigens with
DC-targeting molecules which favour particular uptake
receptors and hence deliver the tumour antigen into the
appropriate cross-presentation pathway19,21,35.
One new concept that evolved from studies of
tumour antigen cross-priming is the use of a tumour
as its own vaccine132. Because the multitude of tumour-
specific mutations cannot be easily identified, strat-
egies that destroy tumours in a way that delivers
increased antigen doses into the cross-presentation
pathway may well increase the loading of these neo-
antigens and boost cross-priming128. This will not gen-
erate a strong and prolonged antitumour response; if
that were the case, then partial responses to chemo-
therapy would lead to immune-based tumour eradica-
tion after chemotherapy has ceased. However, it does
predict that immunotherapies that target DCs follow-
ing chemotherapy are more likely to be effective than
those that do not.
In cases in which tumour antigens or oncogenic
viruses have been identified, various vaccination strat-
egies have been used to induce antitumour responses
through the cross-presentation pathway. One promis-
ing approach is the use of synthetic long peptides. These
seem to be more efficient than full-length proteins at
entering the cross-presentation pathway138. Synthetic
long peptides can induce measurable CTL and TH cell
responses against potentially oncogenic viruses in
humans139,140.
Box 2 | Why was cross-priming so controversial?
Cross-priming has long been controversial, which today may sound surprising given the conceptual advantages it offers.
One reason is that several of the principal experimental models used in this field disfavour cross-presentation: for
example, many viruses — including the widely studied lymphocytic choriomeningitis virus (LCMV) — can infect dendritic
cells (DCs), and the associated antigens are presented by the endogenous MHC class I pathway, bypassing the need for
cross-priming150. Second, antigens that were originally thought not to be cross-presented were found to be so when
more sensitive methods were used127. Third, some antigenic epitopes, such as those located in signal sequences of viral
proteins, are rapidly degraded and therefore are not effectively cross-presented151. Fourth, immortalized cell lines, which
do not mimic all of the functions of bona fide APCs in vivo, were used as surrogate DCs. Fifth, among primary DCs, not all
subsets can efficiently cross-present6,7. Sixth, collagenase-based isolation of primary DCs inhibits endocytosis receptors
that internalize antigen for cross-presentation152.
There were also conceptual problems: some scientists were sceptical about cross-presentation because the underlying
cell biological mechanisms had long remained undefined. Finally, the ability to cross-present implies that DCs that have
taken up viral or tumour components might be killed by effector cytotoxic T lymphocytes (CTLs) (not during priming
because cytotoxic activity requires time to be established), and it was hard to imagine that the immune system should be
targeted against itself. This contradiction has been resolved by the independence of activated CTLs from continual
stimulation by DCs27,28 and by the existence of non-cross-presenting DC subsets6,7 that are not killed and hence can
maintain non-CTL-mediated immune responses.
REVIEWS
NATURE R EVIEWS
|
IMMUNOLOGY VOLUME 10
|
JUNE 2010
|
411
© 20 Macmillan Publishers Limited. All rights reserved10
Signals 1 and 2
T cells require presentation of
antigen (signal 1) and
co-stimulatory signals (signal 2)
for immunogenic activation;
cytokines have been proposed
to be a third signal that
dictates T cell differentiation.
Concluding remarks
Antiviral vaccinations have long been taking advantage
of cross-priming4. Recent findings have clarified some
of the underlying mechanisms, such as the endocytosis
mechanisms in DCs that facilitate cross-presentation,
the organelles in which cross-presentation occurs, DC
licensing, CTL programming, chemokine-mediated
CTL recruitment and the interplay of cross-priming with
chemotherapeutic agents. Translating these advances
into the clinic may allow for the design of more effec-
tive vaccination strategies, for example by optimized
antigen targeting to endocytosis receptors that engage
the cross-presentation machinery in suitable DC sub-
sets. Adjuvants can be chosen that not only augment co-
stimulation, but also improve the cross-presentation of
antigen. Furthermore, the use of adjuvants that induce
different chemokines might further improve cross-prim-
ing. These chemokines may be viewed as a discrete signal
that profoundly affects CTL priming by acting before the
antigen- and co-stimulation-derived signals (also termed
signals 1 and 2, respectively, for T cell activation)141 and
DC-derived cytokines (proposed to be a third signal)58.
Hence, chemokines have been interpreted as ‘signal 0’
in T cell priming142.
Improvements are especially needed in tumour vac-
cination, as recent clinical studies on DC-based tumour
vaccination have been disappointing132,143. Future stud-
ies need to focus on how to harness cross-presenta-
tion to generate chemo-immunotherapy strategies for
therapeutic benefit. Understanding which current
chemotherapies work with, rather than against, the
anticancer immune response may facilitate new cancer
immunotherapies.
We are only beginning to understand in which infec-
tions and in which CTL-mediated diseases cross-prim-
ing is involved or crucial. Certainly, clarifying its role
in human disease is far more difficult than doing so in
mouse disease models. In vitro models have long been
insufficient (BOX 2), but are now improved and supple-
mented by knockdown techniques, and hence will prob-
ably provide further mechanistic insight into the role of
cross-priming in many disease entities.
1. Bevan, M. J. Cross-priming for a secondary cytotoxic
response to minor H antigens with H-2 congenic cells
which do not cross-react in the cytotoxic assay.
J. Exp. Med. 143, 1283–1288 (1976).
This is a seminal paper describing the
phenomenon of cross-priming.
2. Huang, A. Y. et al. Role of bone marrow-derived cells
in presenting MHC class I-restricted tumor antigens.
Science 264, 961–965 (1994).
3. Sigal, L. J., Crotty, S., Andino, R. & Rock, K. L.
Cytotoxic T-cell immunity to virus-infected non-
haematopoietic cells requires presentation of
exogenous antigen. Nature 398, 77–80 (1999).
4. Yewdell, J. W. & Haeryfar, S. M. Understanding
presentation of viral antigens to CD8+ T cells in vivo:
the key to rational vaccine design. Annu. Rev.
Immunol. 23, 651–682 (2005).
5. Kurts, C., Kosaka, H., Carbone, F. R., Miller, J. F. &
Heath, W. R. Class I-restricted cross-presentation of
exogenous self-antigens leads to deletion of
autoreactive CD8+T cells. J. Exp. Med. 186,
239–245 (1997).
This paper showed the phenomenon of
cross-tolerance in a transgenic model, in which
cross-presentation of autoantigen causes
peripheral deletion of autoreactive CTLs.
6. Shortman, K. & Naik, S. H. Steady-state and
inflammatory dendritic-cell development. Nature Rev.
Immunol. 7, 19–30 (2007).
7. Heath, W. R. & Carbone, F. R. Dendritic cell subsets
in primary and secondary T cell responses at body
surfaces. Nature Immunol. 10, 1237–1244
(2009).
8. Kurts, C., Cannarile, M., Klebba, I. & Brocker, T.
Dendritic cells are sufficient to cross-present self-
antigens to CD8 T cells in vivo. J. Immunol. 166,
1439–1442 (2001).
9. Jung, S. et al. In vivo depletion of CD11c+ dendritic
cells abrogates priming of CD8+ T cells by exogenous
cell-associated antigens. Immunity 17, 211–220
(2002).
10. del Rio, M. L., Rodriguez-Barbosa, J. I., Kremmer, E.
& Forster, R. CD103 and CD103+ bronchial lymph
node dendritic cells are specialized in presenting and
cross-presenting innocuous antigen to CD4+ and
CD8+ T cells. J. Immunol. 178, 6861–6866 (2007).
11. Bedoui, S. et al. Cross-presentation of viral and self
antigens by skin-derived CD103+ dendritic cells.
Nature Immunol. 10, 488–495 (2009).
12. Hildner, K. et al. Batf3 deficiency reveals a critical role
for CD8α+ dendritic cells in cytotoxic T cell immunity.
Science 322, 1097–1100 (2008).
13. Edelson, B. T. et al. Peripheral CD103+ dendritic cells
form a unified subset developmentally related to
CD8α+ conventional dendritic cells. J. Exp. Med. 207,
823–836 (2010).
14. den Haan, J. M. & Bevan, M. J. Constitutive versus
activation-dependent cross-presentation of immune
complexes by CD8+ and CD8 dendritic cells in vivo.
J. Exp. Med. 196, 817–827 (2002).
15. Chung, Y., Chang, J. H., Kweon, M. N., Rennert, P. D.
& Kang, C. Y. CD8α11b+ dendritic cells but not
CD8α+ dendritic cells mediate cross-tolerance
toward intestinal antigens. Blood 106, 201–206
(2005).
16. Lutz, M. B. & Kurts, C. Induction of peripheral CD4+
T-cell tolerance and CD8+ T-cell cross-tolerance by
dendritic cells. Eur. J. Immunol. 39, 2325–2330
(2009).
17. Ballesteros-Tato, A., Leon, B., Lund, F. E. & Randall,
T. D. Temporal changes in dendritic cell subsets, cross-
priming and costimulation via CD70 control CD8+
T cell responses to influenza. Nature Immunol. 11,
216–224 (2010).
18. McDonnell, A. M., Prosser, A. C., van Bruggen, I.,
Robinson, B. W. & Currie, A. J. CD8α+ DC are not the
sole subset cross-presenting cell-associated tumor
antigens from a solid tumor. Eur. J. Immunol.
1 Apr 2010 (doi:10.1002/eji.200940153).
19. Dudziak, D. et al. Differential antigen processing by
dendritic cell subsets in vivo. Science 315, 107–111
(2007).
20. Saveanu, L. et al. IRAP identifies an endosomal
compartment required for MHC class I cross-
presentation. Science 325, 213–217 (2009).
21. Burgdorf, S., Kautz, A., Bohnert, V., Knolle, P. A. &
Kurts, C. Distinct pathways of antigen uptake and
intracellular routing in CD4 and CD8 T cell activation.
Science 316, 612–616 (2007).
22. Burgdorf, S. & Kurts, C. Endocytosis mechanisms and
the cell biology of antigen presentation. Curr. Opin.
Immunol. 20, 89–95 (2008).
23. Caminschi, I. et al. The dendritic cell subtype-
restricted C-type lectin Clec9A is a target for vaccine
enhancement. Blood 112 , 3264–3273 (2008).
24. Weck, M. M. et al. hDectin-1 is involved in uptake and
cross-presentation of cellular antigens. Blood 111,
4264–4272 (2008).
25. Medema, J. P. et al. Expression of the serpin serine
protease inhibitor 6 protects dendritic cells from
cytotoxic T lymphocyte-induced apoptosis: differential
modulation by T helper type 1 and type 2 cells.
J. Exp. Med. 194, 657–667 (2001).
26. Bocharov, G., Ford, N. J. & Ludewig, B.
A mathematical approach for optimizing dendritic
cell-based immunotherapy. Methods Mol. Med. 109 ,
19–34 (2005).
27. van Stipdonk, M. J., Lemmens, E. E. &
Schoenberger, S. P. Naive CTLs require a single brief
period of antigenic stimulation for clonal expansion
and differentiation. Nature Immunol. 2, 423–429
(2001).
28. Kaech, S. M. & Ahmed, R. Memory CD8+ T cell
differentiation: initial antigen encounter triggers a
developmental program in naive cells. Nature
Immunol. 2, 415–422 (2001).
29. Allan, R. S. et al. Migratory dendritic cells transfer
antigen to a lymph node-resident dendritic cell
population for efficient CTL priming. Immunity 25,
153–162 (2006).
30. Backer, R. et al. Effective collaboration between
marginal metallophilic macrophages and CD8+
dendritic cells in the generation of cytotoxic T cells.
Proc. Natl Acad. Sci. USA 107, 216–221 (2010).
31. Carbone, F. R., Kurts, C., Bennett, S. R., Miller, J. F. &
Heath, W. R. Cross-presentation: a general mechanism
for CTL immunity and tolerance. Immunol. Today 19,
368–373 (1998).
32. Heit, A. et al. CpG-DNA aided cross-priming by cross-
presenting B cells. J. Immunol. 172, 1501–1507
(2004).
33. Bennett, S. R., Carbone, F. R., Toy, T., Miller, J. F. &
Heath, W. R. B cells directly tolerize CD8+ T cells.
J. Exp. Med. 188, 1977–1983 (1998).
34. Limmer, A. et al. Efficient presentation of exogenous
antigen by liver endothelial cells to CD8+ T cells
results in antigen-specific T-cell tolerance. Nature Med.
6, 1348–1354 (2000).
35. Steinman, R. M. & Pope, M. Exploiting dendritic cells
to improve vaccine efficacy. J. Clin. Invest. 109,
1519–1526 (2002).
36. Maurer, T. et al. CpG-DNA aided cross-presentation of
soluble antigens by dendritic cells. Eur. J. Immunol.
32, 2356–2364 (2002).
37. Schulz, O. et al. Toll-like receptor 3 promotes cross-
priming to virus-infected cells. Nature 433, 887–892
(2005).
38. Burgdorf, S., Scholz, C., Kautz, A., Tampe, R. & Kurts,
C. Spatial and mechanistic separation of cross-
presentation and endogenous antigen presentation.
Nature Immunol. 9, 558–566 (2008).
39. van Kooyk, Y. & Rabinovich, G. A. Protein-glycan
interactions in the control of innate and adaptive
immune responses. Nature Immunol. 9, 593–601
(2008).
40. Rogers, N. C. et al. Syk-dependent cytokine induction
by Dectin-1 reveals a novel pattern recognition
pathway for C type lectins. Immunity 22, 507–517
(2005).
41. Barchet, W., Wimmenauer, V., Schlee, M. &
Hartmann, G. Accessing the therapeutic potential of
immunostimulatory nucleic acids. Curr. Opin.
Immunol. 20, 389–395 (2008).
42. Latz, E. The inflammasomes: mechanisms of activation
and function. Curr. Opin. Immunol. 22, 28–23 (2010).
43. Shi, Y., Evans, J. E. & Rock, K. L. Molecular
identification of a danger signal that alerts the immune
system to dying cells. Nature 425, 516–521 (2003).
REVIEWS
412
|
JUNE 2010
|
VOLUME 10 www.nature.com/reviews/immunol
© 20 Macmillan Publishers Limited. All rights reserved10
44. Martinon, F., Petrilli, V., Mayor, A., Tardivel, A. &
Tschopp, J. Gout-associated uric acid crystals activate
the NALP3 inflammasome. Nature 440, 237–241
(2006).
45. Hamilton-Williams, E. E. et al. Cutting edge: TLR
ligands are not sufficient to break cross-tolerance to
self-antigens. J. Immunol. 174, 1159–1163 (2005).
46. Qiu, F. & Cui, Z. CD4+ T helper cell response is
required for memory in CD8+ T lymphocytes induced
by a poly(I:C)-adjuvanted MHC I-restricted peptide
epitope. J. Immunother. 30, 180–189 (2007).
47. Melief, C. J. Mini-review: Regulation of cytotoxic
T lymphocyte responses by dendritic cells: peaceful
coexistence of cross-priming and direct priming?
Eur. J. Immunol. 33, 2645–2654 (2003).
48. Janssen, E. M. et al. CD4+ T cells are required for
secondary expansion and memory in CD8+
T lymphocytes. Nature 421, 852–856 (2003).
49. Redmond, W. L. & Sherman, L. A. Peripheral tolerance
of CD8 T lymphocytes. Immunity 22, 275–284 (2005).
50. Heymann, F. et al. Kidney dendritic cell activation is
required for progression of renal disease in a mouse
model of glomerular injury. J. Clin. Invest. 119,
1286–1297 (2009).
This report shows that cross-talk between DCs
and autoreactive TH cells in non-lymphoid tissues
causes chemokine production and recruitment of
cross-primed autoreactive CTLs that cause
immunopathology.
51. Smith, C. M. et al. Cognate CD4+ T cell licensing of
dendritic cells in CD8+ T cell immunity. Nature
Immunol. 5, 1143–1148 (2004).
This paper formally showed that T cells can license
DCs for cross-priming of CTLs.
52. van Mierlo, G. J. et al. Activation of dendritic cells that
cross-present tumor-derived antigen licenses CD8+
CTL to cause tumor eradication. J. Immunol. 173,
6753–6759 (2004).
53. Sun, J. C., Williams, M. A. & Bevan, M. J. CD4+ T cells
are required for the maintenance, not programming,
of memory CD8+ T cells after acute infection.
Nature Immunol. 5, 927–933 (2004).
54. Zehn, D., Lee, S. Y. & Bevan, M. J. Complete but
curtailed T-cell response to very low-affinity antigen.
Nature 458, 211–214 (2009).
55. Belz, G. T., Bedoui, S., Kupresanin, F., Carbone, F. R. &
Heath, W. R. Minimal activation of memory CD8+
T cell by tissue-derived dendritic cells favors the
stimulation of naive CD8+ T cells. Nature Immunol.
8, 1060–1066 (2007).
56. Keller, A. M., Xiao, Y., Peperzak, V., Naik, S. H. &
Borst, J. Costimulatory ligand CD70 allows induction
of CD8+ T-cell immunity by immature dendritic cells in
a vaccination setting. Blood 113 , 5167–5175 (2009).
57. Keir, M. E., Freeman, G. J. & Sharpe, A. H. PD-1
regulates self-reactive CD8+ T cell responses to
antigen in lymph nodes and tissues. J. Immunol.
179, 5064–5070 (2007).
58. Mescher, M. F. et al. Signals required for programming
effector and memory development by CD8+ T cells.
Immunol. Rev. 211 , 81–92 (2006).
59. Williams, M. A., Tyznik, A. J. & Bevan, M. J.
Interleukin-2 signals during priming are required for
secondary expansion of CD8+ memory T cells.
Nature 441, 890–893 (2006).
60. Spierings, D. C., Lemmens, E. E., Grewal, K.,
Schoenberger, S. P. & Green, D. R. Duration of CTL
activation regulates IL-2 production required for
autonomous clonal expansion. Eur. J. Immunol. 36,
1707–1717 (2006).
61. Mintern, J. D., Davey, G. M., Belz, G. T., Carbone, F. R.
& Heath, W. R. Cutting edge: precursor frequency
affects the helper dependence of cytotoxic T cells.
J. Immunol. 168, 977–980 (2002).
62. Fuse, S. et al. Recall responses by helpless memory
CD8+ T cells are restricted by the up-regulation of
PD-1. J. Immunol. 182, 4244–4254 (2009).
63. Peperzak, V., Xiao, Y., Veraar, E. A. & Borst, J.
CD27 sustains survival of CTLs in virus-infected
nonlymphoid tissue in mice by inducing autocrine IL-2
production. J. Clin. Invest. 120, 168–178 (2009).
64. Janssen, E. M. et al. CD4+ T-cell help controls CD8+
T-cell memory via TRAIL-mediated activation-induced
cell death. Nature 434, 88–93 (2005).
65. Sacks, J. A. & Bevan, M. J. TRAIL deficiency does not
rescue impaired CD8+ T cell memory generated in the
absence of CD4+ T cell help. J. Immunol. 180,
4570–4576 (2008).
66. Castellino, F. et al. Chemokines enhance immunity by
guiding naive CD8+ T cells to sites of CD4+ T cell-
dendritic cell interaction. Nature 440, 890–895 (2006).
This paper showed that TH cell-mediated licensing
of DCs causes CCR5 ligand-mediated recruitment
of naive CTLs for cross-priming.
67. Hugues, S. et al. Dynamic imaging of chemokine-
dependent CD8+ T cell help for CD8+ T cell responses.
Nature Immunol. 8, 921–930 (2007).
68. Dorner, B. G. et al. Selective expression of the
chemokine receptor XCR1 on cross-presenting
dendritic cells determines cooperation with CD8+
T cells. Immunity 31, 823–833 (2009).
69. Fujii, S., Shimizu, K., Hemmi, H. & Steinman, R. M.
Innate Vα14+ natural killer T cells mature dendritic
cells, leading to strong adaptive immunity. Immunol.
Re v. 220, 183–198 (2007).
70. Semmling, V. et al. Alternative cross-priming through
CCL17–CCR4-mediated attraction of CTLs toward
NKT cell-licensed DCs. Nature Immunol. 11, 313–320
(2010).
This paper showed that NKT cells can license DCs
for cross-priming and recruit naive CTLs by CCR4
ligands.
71. Wakim, L. M., Waithman, J., van Rooijen, N., Heath,
W. R. & Carbone, F. R. Dendritic cell-induced memory
T cell activation in nonlymphoid tissues. Science 319,
198–202 (2008).
72. Nakanishi, Y., Lu, B., Gerard, C. & Iwasaki, A. CD8+
T lymphocyte mobilization to virus-infected tissue
requires CD4+ T-cell help. Nature 462, 510–513
(2009).
This report showed that cross-talk between DCs
and TH cells in viral infections of mucosal tissue
causes chemokine-mediated tissue recruitment of
cross-primed CTLs.
73. Marzo, A. L. et al. Tumor-specific CD4+ T cells have
a major “post-licensing” role in CTL mediated anti-
tumor immunity. J. Immunol. 165, 6047–6055
(2000).
74. Savinov, A. Y., Wong, F. S., Stonebraker, A. C. &
Chervonsky, A. V. Presentation of antigen by
endothelial cells and chemoattraction are required for
homing of insulin-specific CD8+ T cells. J. Exp. Med.
197, 643–656 (2003).
75. Galea, I. et al. An antigen-specific pathway for CD8
T cells across the blood-brain barrier. J. Exp. Med.
204, 2023–2030 (2007).
76. von Oppen, N. et al. Systemic antigen cross-presented
by liver sinusoidal endothelial cells induces liver-
specific CD8 T-cell retention and tolerization.
Hepatology 49, 1664–1672 (2009).
77. Dong, H. et al. B7-H1 determines accumulation and
deletion of intrahepatic CD8+ T lymphocytes.
Immunity 20, 327–336 (2004).
78. Reddehase, M. J. Antigens and immunoevasins:
opponents in cytomegalovirus immune surveillance.
Nature Rev. Immunol. 2, 831–844 (2002).
79. Andrews, D. M., Andoniou, C. E., Granucci, F.,
Ricciardi-Castagnoli, P. & Degli-Esposti, M. A.
Infection of dendritic cells by murine cytomegalovirus
induces functional paralysis. Nature Immunol.
2, 1077–1084 (2001).
80. Holtappels, R. et al. Cytomegalovirus misleads its host
by priming of CD8 T cells specific for an epitope not
presented in infected tissues. J. Exp. Med. 199,
131–136 (2004).
81. Bickham, K. et al. Dendritic cells initiate immune
control of Epstein-Barr virus transformation of B
lymphocytes in vitro. J. Exp. Med. 198, 1653–1663
(2003).
82. Bohm, V. et al. Epitope-specific in vivo protection
against cytomegalovirus disease by CD8 T cells in
the murine model of preemptive immunotherapy.
Med. Microbiol. Immunol. 197, 135–144 (2008).
83. Tewalt, E. F. et al. Viral sequestration of antigen
subverts cross presentation to CD8+ T cells.
PLoS Pathog. 5, e1000457 (2009).
84. Le Bon, A. et al. Cross-priming of CD8+ T cells
stimulated by virus-induced type I interferon.
Nature Immunol. 4, 1009–1015 (2003).
This paper revealed that type I IFN stimulates
cross-priming.
85. Lang, K. S. et al. Immunoprivileged status of the
liver is controlled by Toll-like receptor 3 signaling.
J. Clin. Invest. 116, 2456–2463 (2006).
86. Rehermann, B. & Nascimbeni, M. Immunology of
hepatitis B virus and hepatitis C virus infection.
Nature Rev. Immunol. 5, 215–229 (2005).
87. Hosel, M. et al. Not interferon, but interleukin-6
controls early gene expression in hepatitis B virus
infection. Hepatology 50, 1773–1782 (2009).
88. Sixt, M. et al. The conduit system transports soluble
antigens from the afferent lymph to resident dendritic
cells in the T cell area of the lymph node. Immunity
22, 19–29 (2005).
89. Datta, S. K. et al. Vaccination with irradiated Listeria
induces protective T cell immunity. Immunity 25,
143–152 (2006).
90. Neuenhahn, M. et al. CD8α+ dendritic cells are
required for efficient entry of Listeria monocytogenes
into the spleen. Immunity 25, 619–630 (2006).
91. Kang, S. J., Liang, H. E., Reizis, B. & Locksley, R. M.
Regulation of hierarchical clustering and activation of
innate immune cells by dendritic cells. Immunity 29,
819–833 (2008).
92. Tseng, K. E., Chung, C. Y., H’Ng, W., S. & Wang, S. L.
Early infection termination affects number of CD8+
memory T cells and protective capacities in Listeria
monocytogenes-infected mice upon rechallenge.
J. Immunol. 182, 4590–4600 (2009).
93. Schaible, U. E. et al. Apoptosis facilitates antigen
presentation to T lymphocytes through MHC-I and
CD1 in tuberculosis. Nature Med. 9, 1039–1046
(2003).
94. Winau, F. et al. Apoptotic vesicles crossprime CD8
T cells and protect against tuberculosis. Immunity 24,
105–117 (2006).
95. Zehn, D. & Bevan, M. J. T cells with low avidity for a
tissue-restricted antigen routinely evade central and
peripheral tolerance and cause autoimmunity.
Immunity 25, 261–270 (2006).
96. McDevitt, H. O. & Unanue, E. R. Autoimmune
diabetes mellitus — much progress, but many
challenges. Adv. Immunol. 100, 1–12 (2008).
97. de Jersey, J. et al. β cells cannot directly prime
diabetogenic CD8 T cells in nonobese diabetic mice.
Proc. Natl Acad. Sci. USA 104, 1295–1300 (2007).
This paper showed that cross-priming is required
for diabetes in the NOD mouse model.
98. Hamilton-Williams, E. E. et al. Expression of diabetes-
associated genes by dendritic cells and CD4 T cells
drives the loss of tolerance in nonobese diabetic mice.
J. Immunol. 183, 1533–1541 (2009).
99. von Herrath, M. Diabetes: A virus-gene collaboration.
Nature 459, 518–519 (2009).
100. Richer, M. J. & Horwitz, M. S. Coxsackievirus infection
as an environmental factor in the etiology of type 1
diabetes. Autoimmun Rev. 8, 611–615 (2009).
101. Benoist, C. Autoimmunity provoked by infection:
how good is the case for T cell epitope mimicry?
Nature Immunol. 2, 797–801 (2001).
102. Pinkse, G. G. et al. Autoreactive CD8 T cells associated
with β cell destruction in type 1 diabetes. Proc. Natl
Acad. Sci. USA 102, 18425–18430 (2005).
103. Nakayama, M. et al. Priming and effector dependence
on insulin B: 9–23 peptide in NOD islet autoimmunity.
J. Clin. Invest. 117, 1835–1843 (2007).
104. Luckashenak, N. et al. Constitutive crosspresentation of
tissue antigens by dendritic cells controls CD8+ T cell
tolerance in vivo. Immunity 28, 521–532 (2008).
This report showed that non-transgenic self
antigens can induce cross-tolerance.
105. Hoglund, P. et al. Initiation of autoimmune diabetes
by developmentally regulated presentation of islet cell
antigens in the pancreatic lymph nodes. J. Exp. Med.
189, 331–339 (1999).
106. Mohan, J. F. et al. Unique autoreactive T cells
recognize insulin peptides generated within the islets
of Langerhans in autoimmune diabetes. Nature
Immunol. 4, 350–354 (2010).
107. Jahromi, M. M. & Eisenbarth, G. S. Cellular and
molecular pathogenesis of type 1A diabetes. Cell.
Mol. Life Sci. 64, 865–72 (2007).
108. Neilson, E. G., McCafferty, E., Mann, R., Michaud, L.
& Clayman, M. Murine interstitial nephritis. III. The
selection of phenotypic (Lyt and L3T4) and idiotypic
(RE-Id) T cell preferences by genes in Igh-1 and
H-XXXXX2K characterizes the cell-mediated potential
for disease expression: susceptible mice provide a
unique effector T cell repertoire in response to tubular
antigen. J. Immunol. 134, 2375–2382 (1985).
109. Gudjonsson, J. E., Johnston, A., Sigmundsdottir, H.
& Valdimarsson, H. Immunopathogenic mechanisms
in psoriasis. Clin. Exp. Immunol. 135, 1–8 (2004).
110. Goverman, J. Autoimmune T cell responses in the
central nervous system. Nature Rev. Immunol.
9, 393–407 (2009).
111. Perchellet, A., Brabb, T. & Goverman, J. M.
Crosspresentation by nonhematopoietic and direct
presentation by hematopoietic cells induce central
tolerance to myelin basic protein. Proc. Natl Acad. Sci.
USA 105 , 14040–14045 (2008).
This paper implicated cross-tolerance in the
control of encephalitogenic CTLs.
REVIEWS
NATURE R EVIEWS
|
IMMUNOLOGY VOLUME 10
|
JUNE 2010
|
413
© 20 Macmillan Publishers Limited. All rights reserved10
112. Huseby, E. S. et al. A pathogenic role for myelin-
specific CD8+ T cells in a model for multiple sclerosis.
J. Exp. Med. 194, 669–676 (2001).
113. Harkiolaki, M. et al. T cell-mediated autoimmune
disease due to low-affinity crossreactivity to
common microbial peptides. Immunity 30,
348–357 (2009).
114. Fiorillo, M. T. & Sorrentino, R. T-cell responses against
viral and self-epitopes and HLA-B27 subtypes
differentially associated with ankylosing spondylitis.
Adv. Exp. Med. Biol. 649, 255–262 (2009).
115. Ichiki, Y. et al. T cell immunity in autoimmune
hepatitis. Autoimmun. Rev. 4, 315–321 (2005).
116. Kammer, A. R. et al. Molecular mimicry of human
cytochrome P450 by hepatitis C virus at the level of
cytotoxic T cell recognition. J. Exp. Med. 190,
169–176 (1999).
117. Gokmen, M. R., Lombardi, G. & Lechler, R. I. The
importance of the indirect pathway of allorecognition
in clinical transplantation. Curr. Opin. Immunol. 20,
568–574 (2008).
118. Boon, T., Cerottini, J. C., Van den Eynde, B., van der
Bruggen, P. & Van Pel, A. Tumor antigens recognized
by T lymphocytes. Annu. Rev. Immunol. 12, 337–365
(1994).
119. Boland, C. R. & Ricciardiello, L. How many mutations
does it take to make a tumor? Proc. Natl Acad. Sci.
USA 96, 14675–14677 (1999).
120. Marzo, A. L. et al. Tumor antigens are constitutively
presented in the draining lymph nodes. J. Immunol.
162, 5838–5845 (1999).
121. Cuenca, A. et al. Extra-lymphatic solid tumor growth is
not immunologically ignored and results in early
induction of antigen-specific T-cell anergy: dominant
role of cross-tolerance to tumor antigens. Cancer Res.
63, 9007–9015 (2003).
122. Ney, J. T. et al. Autochthonous liver tumors induce
systemic T cell tolerance associated with T cell
receptor down-modulation. Hepatology 49, 471–481
(2009).
123. Gerner, M. Y., Casey, K. A. & Mescher, M. F. Defective
MHC class II presentation by dendritic cells limits
CD4 T cell help for antitumor CD8 T cell responses.
J. Immunol. 181, 155–164 (2008).
124. Stumbles, P. A. et al. Cutting edge: tumor-specific
CTL are constitutively cross-armed in draining lymph
nodes and transiently disseminate to mediate tumor
regression following systemic CD40 activation.
J. Immunol. 173, 5923–5928 (2004).
125. Nelson, D. J. et al. Tumor progression despite efficient
tumor antigen cross-presentation and effective
“arming” of tumor antigen-specific CTL. J. Immunol.
166, 5557–5566 (2001).
126. Lyman, M. A., Aung, S., Biggs, J. A. & Sherman, L. A.
A spontaneously arising pancreatic tumor does not
promote the differentiation of naive CD8+ T
lymphocytes into effector CTL. J. Immunol. 172,
6558–6567 (2004).
127. Nguyen, L. T. et al. Tumor growth enhances cross-
presentation leading to limited T cell activation
without tolerance. J. Exp. Med. 195, 423–435
(2002).
128. Nowak, A. K. et al. Induction of tumor cell apoptosis
in vivo increases tumor antigen cross-presentation,
cross-priming rather than cross-tolerizing host tumor-
specific CD8 T cells. J. Immunol. 170, 4905–4913
(2003).
129. Sauter, B. et al. Consequences of cell death: exposure
to necrotic tumor cells, but not primary tissue cells or
apoptotic cells, induces the maturation of
immunostimulatory dendritic cells. J. Exp. Med. 191,
423–434 (2000).
130. Schnurr, M. et al. Apoptotic pancreatic tumor cells are
superior to cell lysates in promoting cross-priming of
cytotoxic T cells and activate NK and γδ T cells.
Cancer Res. 62, 2347–2352 (2002).
131. Obeid, M. et al. Calreticulin exposure dictates the
immunogenicity of cancer cell death. Nature Med. 13,
54–61 (2007).
132. Lake, R. A. & Robinson, B. W. Immunotherapy and
chemotherapy — a practical partnership. Nature Rev.
Cancer 5, 397–405 (2005).
133. Schnurr, M. et al. ISCOMATRIX adjuvant induces
efficient cross-presentation of tumor antigen by
dendritic cells via rapid cytosolic antigen delivery and
processing via tripeptidyl peptidase II. J. Immunol.
182, 1253–1259 (2009).
134. Motta, I. et al. Cross-presentation by dendritic cells of
tumor antigen expressed in apoptotic recombinant
canarypox virus-infected dendritic cells. J. Immunol.
167, 1795–1802 (2001).
135. Bendz, H. et al. Human heat shock protein 70
enhances tumor antigen presentation through
complex formation and intracellular antigen delivery
without innate immune signaling. J. Biol. Chem.
282, 31688–31702 (2007).
136. Apetoh, L. et al. Toll-like receptor 4-dependent
contribution of the immune system to anticancer
chemotherapy and radiotherapy. Nature Med. 13,
1050–1059 (2007).
137. Dhodapkar, K. M., Krasovsky, J., Williamson, B. &
Dhodapkar, M. V. Antitumor monoclonal antibodies
enhance cross-presentation of cellular antigens and
the generation of myeloma-specific killer T cells by
dendritic cells. J. Exp. Med. 195, 125–133 (2002).
138. Zhang, H. et al. Comparing pooled peptides with
intact protein for accessing cross-presentation
pathways for protective CD8+ and CD4+ T cells.
J. Biol. Chem. 284, 9184–9191 (2009).
139. Melief, C. J. & van der Burg, S. H. Immunotherapy of
established (pre)malignant disease by synthetic long
peptide vaccines. Nature Rev. Cancer 8, 351–360
(2008).
140. Kenter, G. G. et al. Vaccination against HPV-16
oncoproteins for vulvar intraepithelial neoplasia.
N. Engl. J. Med. 361, 1838–1847 (2009).
141. Bretscher, P. & Cohn, M. A theory of self–nonself
discrimination. Science 169, 1042–1049 (1970).
142. Bousso, P. & Albert, M. L. Signal 0 for guided priming
of CTLs: NKT cells do it too. Nature Immunol. 11,
284–286 (2010).
143. Robson, N. C., Hoves, S., Maraskovsky, E. & Schnurr,
M. Presentation of tumour antigens by dendritic cells
and challenges faced. Curr. Opin. Immunol. 22,
137–144 (2010).
144. Guermonprez, P. et al. ER-phagosome fusion defines
an MHC class I cross-presentation compartment in
dendritic cells. Nature 425, 397–402 (2003).
145. Houde, M. et al. Phagosomes are competent
organelles for antigen cross-presentation. Nature
425, 402–406 (2003).
146. Touret, N. et al. Quantitative and dynamic assessment
of the contribution of the ER to phagosome formation.
Cell 123, 157–170 (2005).
147. Savina, A. et al. NOX2 controls phagosomal pH to
regulate antigen processing during crosspresentation
by dendritic cells. Cell 126, 205–218 (2006).
148. Di Pucchio, T. et al. Direct proteasome-independent
cross-presentation of viral antigen by plasmacytoid
dendritic cells on major histocompatibility complex
class I. Nature Immunol. 9, 551–557 (2008).
149. Belizaire, R. & Unanue, E. R. Targeting proteins to
distinct subcellular compartments reveals unique
requirements for MHC class I and II presentation.
Proc. Natl Acad. Sci. USA 106, 17463–17468
(2009).
150. Zinkernagel, R. M. On cross-priming of MHC class
I-specific CTL: rule or exception? Eur. J. Immunol. 32,
2385–2392 (2002).
151. Wolkers, M. C., Brouwenstijn, N., Bakker, A. H.,
Toebes, M. & Schumacher, T. N. Antigen bias in T cell
cross-priming. Science 304, 1314–1317 (2004).
152. Burgdorf, S. et al. Steady-state cross-presentation of
OVA is mannose receptor-dependent but inhibitable
by collagen fragments. Proc. Natl Acad. Sci. USA 107,
E48–E49 (2010).
153. Di Bonito, P. et al. Anti-tumor CD8+ T cell immunity
elicited by HIV-1-based virus-like particles incorporating
HPV-16 E7 protein. Virology 395, 45–55 (2009).
154. Kemball, C. C. et al. Coxsackievirus B3 inhibits
antigen presentation in vivo, exerting a profound
and selective effect on the MHC class I pathway.
PLoS Pathog. 5, e1000618 (2009).
155. Racanelli, V., Behrens, S. E., Aliberti, J. & Rehermann, B.
Dendritic cells transfected with cytopathic self-
replicating RNA induce crosspriming of CD8+ T cells
and antiviral immunity. Immunity 20, 47–58 (2004).
156. Ramirez, M. C. & Sigal, L. J. Macrophages and
dendritic cells use the cytosolic pathway to rapidly
cross-present antigen from live, vaccinia-infected cells.
J. Immunol. 169, 6733–6742 (2002).
157. Lundie, R. J. et al. Blood-stage Plasmodium infection
induces CD8+ T lymphocytes to parasite-expressed
antigens, largely regulated by CD8α+ dendritic cells.
Proc. Natl Acad. Sci. USA 105, 14509–14514 (2008).
158. Araujo, A. F. et al. CD8+-T-cell-dependent control of
Trypanosoma cruzi infection in a highly susceptible
mouse strain after immunization with recombinant
proteins based on amastigote surface protein 2.
Infect. Immun. 73, 6017–6025 (2005).
159. Steele, L. N., Balsara, Z. R. & Starnbach, M. N.
Hematopoietic cells are required to initiate a
Chlamydia trachomatis-specific CD8+ T cell
response. J. Immunol. 173, 6327–6337 (2004).
160. Calzascia, T. et al. Cutting edge: cross-presentation as a
mechanism for efficient recruitment of tumor-specific
CTL to the brain. J. Immunol. 171, 2187–2191 (2003).
161. Brazillet, M. P., Batteux, F., Abehsira-Amar, O.,
Nicoletti, F. & Charreire, J. Induction of experimental
autoimmune thyroiditis by heat-denatured porcine
thyroglobulin: a Tc1-mediated disease. Eur. J. Immunol.
29, 1342–1352 (1999).
162. Sotomayor, E. M. et al. Cross-presentation of tumor
antigens by bone marrow-derived antigen-presenting
cells is the dominant mechanism in the induction of
T-cell tolerance during B-cell lymphoma progression.
Blood 98, 1070–1077 (2001).
163. Li, Y. et al. Efficient cross-presentation depends on
autophagy in tumor cells. Cancer Res. 68,
6889–6895 (2008).
Acknowledgments
The authors thank W. Kolanus (LIMES Institute Bonn), J.
Yewdell (NIH), M. von Herrath (LIAI) and R. Slattery (Monash
University Melbourne) for comments and discussions. We
apologize to all colleagues whose work could not be cited
owing to space restrictions. The authors are supported by the
German Research foundation (DFG Sonderforschungsbereiche
704, 670 and 645, Transregio 57, Klinische Forschergruppe
228), the German Academic Exchange service (DAAD) and
the Group of Eight, Australia.
Competing interests statement
The authors declare no competing financial interests.
DATABASES
UniProtKB: http://www.uniprot.org
BATF3 | CCL17 | CCR4 | CCR5 | CD8α | CD24 | CD27 |
CD40 ligand | CD70 | CD103 | CLEC7A | CLEC9A | DC-SIGN |
DEC205 | IRAP | mannose receptor 1 | PD1 | RIG-I | TRAIL |
XCL1 | XCR1
FURTHER INFORMATION
Christian Kurts’s homepage: http://www.immei.uni-bonn.de
ALL LINKS ARE ACTIVE IN THE ONLINE PDF
REVIEWS
414
|
JUNE 2010
|
VOLUME 10 www.nature.com/reviews/immunol
© 20 Macmillan Publishers Limited. All rights reserved10
... There was comparable cytokine producing capacity of CD4 + T cells and similar frequency of CD4 + CD25 + FOXP3 + Treg in the PLN between the genotypes ( Supplementary Fig 3g, h). Although these observations in CD8 + T cells were restricted to the initiation phase of autoimmune T1D when the PLN was crucial for the development of diabetes (~10w) 39 , these results suggest that the defective proliferation and/or reduced number of CD8 + CTLs, which are the primary effector immune cells for β-cell Reduced proliferation of CD8 + T cells in the PLN of Tyk2deficient mice Antigen encountered naïve T cells become activated, proliferate, and develop into effector and memory T cells that exhibit the CD44 hi phenotype 40 . To investigate the role of TYK2 in the priming of naïve CD8 + T cells, we used islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)-specific CD8 + T cells (8.3 CD8 + T cells) from NY8.3-NOD transgenic mice, which express a T cell receptor (TCR) specific for the islet autoantigen IGRP 206-214 peptide 41 . ...
... A previous study reported the importance of TYK2 in antigen presentation by dendritic cells (DCs) for the expansion of IFN-γ + CD8 + T cells following Listeria monocytogenes infection 43 . Next, we analyzed DCs, which have a central role in the adaptive immune system via antigen presentation to T cells ( Supplementary Fig. 5a, b) 40 . In the PLN, the frequency of CD11c + MHC II mid resident DCs (rDCs), including CD8 + and CD11b + subsets, and CD11c + MHC II hi migratory DCs (mDCs), including the CD103 + CD11b -, CD103 + CD11b + , and CD103 -CD11b + subsets, were comparable irrespective of Tyk2 genotypes ( Fig. 4c and Supplementary Fig. 5c). ...
... Comparable expression levels of MHC II (I-A g7 ) were observed in these DC subsets from all genotypes ( Supplementary Fig. 5d). However, we found that Tyk2 deficiency reduced the expression of MHC I (H-2K d and H-2D b ), a crucial molecule for presenting antigens to CD8 + T cells 40 , in CD8 + rDC but not other subsets of DCs ( Fig. 4d and Supplementary Fig. 5e, f). This MHC I reduction in CD8 + rDC was not specific for the PLN as comparable results were observed in the iLN and spleen ( Supplementary Fig. 5g, h). ...
Article
Full-text available
Tyrosine kinase 2 (TYK2), a member of the JAK family, has attracted attention as a potential therapeutic target for autoimmune diseases. However, the role of TYK2 in CD8⁺ T cells and autoimmune type 1 diabetes (T1D) is poorly understood. In this study, we generate Tyk2 gene knockout non-obese diabetes (NOD) mice and demonstrate that the loss of Tyk2 inhibits the development of autoreactive CD8⁺ T-BET⁺ cytotoxic T lymphocytes (CTLs) by impairing IL-12 signaling in CD8⁺ T cells and the CD8⁺ resident dendritic cell-driven cross-priming of CTLs in the pancreatic lymph node (PLN). Tyk2-deficient CTLs display reduced cytotoxicity. Increased inflammatory responses in β-cells with aging are dampened by Tyk2 deficiency. Furthermore, treatment with BMS-986165, a selective TYK2 inhibitor, inhibits the expansion of T-BET⁺ CTLs, inflammation in β-cells and the onset of autoimmune T1D in NOD mice. Thus, our study reveals the diverse roles of TYK2 in driving the pathogenesis of T1D.
... Dendritic cells are among the most potent antigen-presenting cells in the immune system [54]. In a cancer context, dendritic cells participate in a process called cross-priming, wherein they activate CD8 + T cells by presenting cancer antigens [55,56]. Subsequently, these CD8 + T cells undergo training and activation against the presented antigens, thereby initiating a cytotoxic immune response against tumor components [55,56]. ...
... In a cancer context, dendritic cells participate in a process called cross-priming, wherein they activate CD8 + T cells by presenting cancer antigens [55,56]. Subsequently, these CD8 + T cells undergo training and activation against the presented antigens, thereby initiating a cytotoxic immune response against tumor components [55,56]. This response constitutes a cornerstone of immunity against cancer antigens. ...
Article
Full-text available
Simple Summary This study aimed to pinpoint immune-related genes that show heightened activity in cancerous tissue and explore their correlation with cell infiltration in the tumor microenvironment using bioinformatics analysis. By examining gene expression from both stomach cancer and adjacent healthy tissues, we aimed to uncover their significance in cancer development and their impact on the body’s immune response. We identified several genes, including FN1, COL1A2, THBS2, COL3A1, COL5A1, and BGN, which appear to be associated with poorer outcomes for stomach cancer patients. These genes also demonstrate connections to specific immune cells within cancerous tissue. Understanding the role of these genes in the immune response to cancer could facilitate the development of novel treatments and enhance prognostic capabilities for individuals with stomach cancer. Abstract Objective: Gastric carcinoma (GC) is the fifth most commonly diagnosed cancer and the third leading cause of cancer-related deaths globally. The tumor microenvironment plays a significant role in the pathogenesis, prognosis, and response to immunotherapy. However, the immune-related molecular mechanisms underlying GC remain elusive. Bioinformatics analysis of the gene expression of GC and paracancerous healthy tissues from the same patient was performed to identify the key genes and signaling pathways, as well as their correlation to the infiltration of the tumor microenvironment (TME) by various immune cells related to GC development. Methods: We employed GSE19826, a gene expression profile from the Gene Expression Omnibus (GEO), for our analysis. Functional enrichment analysis of Differentially Expressed Genes (DEGs) was conducted using the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes database. Results: Cytoscape software facilitated the identification of nine hub DEGs, namely, FN1, COL1A1, COL1A2, THBS2, COL3A1, COL5A1, APOE, SPP1, and BGN. Various network analysis algorithms were applied to determine their high connectivity. Among these hub genes, FN1, COL1A2, THBS2, COL3A1, COL5A1, and BGN were found to be associated with a poor prognosis for GC patients. Subsequent analysis using the TIMER database revealed the infiltration status of the TME concerning the overexpression of these six genes. Specifically, the abovementioned genes demonstrated direct correlations with cancer-associated fibroblasts, M1 and M2 macrophages, myeloid-derived suppressor cells, and activated dendritic cells. Conclusion: Our findings suggest that the identified hub genes, particularly BGN, FN1, COL1A2, THBS2, COL3A1, and COL5A1, play crucial roles in GC prognosis and TME cell infiltration. This comprehensive analysis enhances our understanding of the molecular mechanisms underlying GC development and may contribute to the identification of potential therapeutic targets and prognostic markers for GC patients.
... We showed that the modified tumor lysate-pulsed DCs had a higher expression of surface markers such as CD83 and CD1a, and a better ability to stimulate lymphocyte proliferation and IFN-γ secretion, which indicates a more efficacious adaptive immune response. The cross-presentation of foreign antigens by DCs to CD8 + T lymphocytes underpins antitumor immunity [23,27] . Chen et al proposed that DCs present foreign antigens to naive CD8 + T cells through cross-activation in the form of MHC-I-antigen complexes and stimulate them into CTLs, which exert a strong and specific anti-tumor immune response [28] . ...
Article
Full-text available
Objective This study aimed to improve the antitumor immunocompetence of a tumor lysate-pulsed dendritic cell (DC) vaccine through differential centrifugation and provide a theoretical basis for its clinical application in glioblastoma. Methods Peripheral blood mononuclear cells were extracted using Ficoll-Paque PLUS and induced into mature DCs in vitro with a cytokine cocktail. The modified tumor lysate was generated by differential centrifugation. The maturity markers of DCs in each group, namely the modified tumor lysate, tumor lysate, and negative and positive control groups, were assessed using flow cytometry. Furthermore, their ability to stimulate lymphocyte proliferation and in vitro antitumor effects were assessed using Cell Trace TM CFSE. IFN-γ secretion levels were measured with ELISA. Intracellular reactive oxygen species were measured using 2',7'-dichlorofluorescein diacetate (DCFDA) staining. The results were statistically analyzed using an unpaired Student's t-test and were considered significant at P < 0.05. Results Compared with tumor lysate-pulsed DCs, modified tumor lysate-pulsed DCs had a higher expression of maturity markers: CD1a (7.38 ± 0.53% vs. 4.47 ± 0.75%) and CD83 (19.81 ± 4.09% vs. 9.64 ± 1.50%), were better capable of stimulating lymphocyte proliferation [proliferation index (PI): 8.54 ± 0.16 vs. 7.35 ± 0.05], secreting IFN-γ, and inducing stronger in-vitro cytotoxic T lymphocyte (CTL) cytotoxicity against glioblastoma cells. In addition, we found that the level of ROS in modified tumor lysate-pulsed DCs was lower than that in tumor lysate-pulsed DCs. Conclusion Differential centrifugation of tumor lysates can improve the antitumor immunocompetence of DC vaccines, and reactive oxygen species may be the key to affecting DC function in the whole tumor lysate. Abstract Glioblastoma is the most common primary malignant tumor of the central nervous system with an annual incidence rate of 3-5/100 000 and a poor prognosis of 14.6 months, accounting for about 50% of all gliomas [1-2]. Although there are numerous studies on glioblastoma, the anatomical location of the central nervous system limits the application of locoregional treatment. In addition, the tumor is characterized by endogenous radio-and chemoresistance, heterogeneity, and an immunosuppressive microenvironment; these factors limit the progress of glioblastoma treatment. The standard treatment is to safely maximize surgical resection with adjuvant radiotherapy and chemotherapy [3-4]. Other adjuvant treatments include tumor treating fields, immunotherapy, gene therapy, and molecular targeted therapy, among which immunotherapy has great potential in the treatment of glioblastoma. As the most effective antigen-presenting cells, dendritic cells (DCs), bridge the innate and adaptive immune system [5-7]. DCs are cultured in vitro for maturation, sensitized with tumor-associated antigens or tumor-specific antigens, and then injected back into Downloaded from
... While CD8+T cells are typically favored for targeting tumors, evidence suggests that after DCs phagocytose apoptotic tumor cells, the antigens can still be presented on MHC class II molecules, thereby activating tumorspecific CD4+ T cell responses (101). CD4+ T cells are also essential for anti-tumor responses, as they lead to DC "licensing" and initiation, thereby enhancing cross-presentation to CD8+ T cells (252,253). Thus, these factors collectively amplify the effect of the anti-tumor immune response. Additionally, some researchers believe that the direct contact between DCs and apoptotic cells might mimic a more natural antigen presentation environment, with DC surfaces loaded with an appropriate density of tumor antigen peptides, which helps to activate CTLs with higher affinity and more specificity (244). ...
Article
Full-text available
Efferocytosis, the process of engulfing and removing apoptotic cells, plays an essential role in preserving tissue health and averting undue inflammation. While macrophages are primarily known for this task, dendritic cells (DCs) also play a significant role. This review delves into the unique contributions of various DC subsets to efferocytosis, highlighting the distinctions in how DCs and macrophages recognize and handle apoptotic cells. It further explores how efferocytosis influences DC maturation, thereby affecting immune tolerance. This underscores the pivotal role of DCs in orchestrating immune responses and sustaining immune equilibrium, providing new insights into their function in immune regulation.
Article
In addition to cross-presentation, cross-dressing plays an important role in the induction of CD8⁺ T cell immunity. In the process of cross-dressing, conventional dendritic cells (DCs) acquire major histocompatibility complex class I (MHCI) from other cells and subsequently prime CD8⁺ T cells via the pre-formed antigen-MHCI complexes without antigen processing. However, the mechanisms underlying the cross-dressing pathway, as well as the relative contributions of cross-presentation and cross-dressing to CD8⁺ T cell priming are not fully understood. Here, we demonstrate that DCs rapidly acquire MHCI-containing membrane fragments from dead cells via the phosphatidylserine recognition-dependent mechanism for cross-dressing. The MHCI dressing is enhanced by a TLR3 ligand polyinosinic-polycytidylic acid (polyI:C). Further, polyI:C promotes not only cross-presentation but also cross-dressing in vivo. Taken together, these results suggest that cross-dressing as well as cross-presentation is involved in inflammatory diseases associated with cell death and type I IFN production.
Article
The inevitability of pandemics creates an urgent requirement for emergency action to develop effective technologies to reduce harm to the human population in the period between the onset of an epidemic and the development and production of a vaccine. In this review we discuss the possibility of engineering universal vaccines. Such vaccines would exploit the nonspecific potential of innate immunity, would allow the population to be vaccinated when an unidentified pathogen appears, and would reduce disease severity until pathogen-specific vaccines become available. There are strong evidences that bacterial or viral vaccines such as BCG, measles and polio have heterologous protective effects against unrelated pathogens. This is attributed to the innate immune system’s ability to maintain the memory of past infections and use it to develop immune defenses against new ones. This effect has been called “trained” immunity. The use of trained immunity may also represent an important new approach to improving existing vaccines or to developing new vaccines that combine the induction of classical adaptive immune memory and innate immune memory. Such approaches can be boosted by genetic technology and prove extremely useful against future pandemics.
Preprint
Full-text available
Targeting cancer cell mitochondria holds great promise for cancer treatment, yet current strategies for specifically and effectively destroying cancer mitochondria in vivo are limited. Here, we introduce mLumiOpto, an innovative mitochondrial-targeted luminoptogenetics technology designed to directly disrupt the inner mitochondrial membrane (IMM) potential and induce cancer cell death. We synthesize a blue light-gated channelrhodopsin (CoChR) in the IMM with a mitochondrial leading sequence and co-express a blue bioluminescence-emitting Nanoluciferase (NLuc) in the cytosol of the same cells. The mLumiOpto genes are selectively delivered to cancer cells in vivo by using two strategies: adeno-associated virus (AAV) carrying a cancer-specific promoter (cfos) or cancer-targeted monoclonal antibody-tagged exosome-associated AAV (mAb-Exo-AAV). Induction with ViviRen (an NLuc luciferin) elicits robust endogenous NLuc bioluminescence, which activates mitochondrial CoChR, triggering cancer cell IMM permeability disruption, IMM potential depolarization and subsequent cell death. Importantly, mLumiOpto demonstrates remarkable efficacy in reducing tumor burden and killing tumor cells in glioblastomas (administrated with AAV) or triple-negative breast cancers (administrated with mAb-Exo-AAV) xenografted mouse models. These findings establish mLumiOpto as a novel and promising therapeutic strategy by targeting cancer cell mitochondria in vivo.
Chapter
Lung cancer is an imminent health threat and the predominant cause of cancer-related fatalities worldwide. Significant advancements in identifying important mutational variations in the cells have resulted in the creation of molecularly targeted medicines, the efficacy of which has been hampered by the establishment of resistance mechanisms. The current technological revolution in cancer treatment has resulted in substantial medical therapy and cancer research advances. New cancer treatment techniques, such as immunotherapy, are being hailed as “game changers” and “miracles in the making” due to encouraging results in treating patients with advanced diseases. In the present immunotherapy epoch, a growing number of immunotherapeutic medications have been used in the clinical management of several types of cancer. Adoptive cell therapy (ACT), which uses T cells that have been genetically engineered, such as tumor-infiltrating lymphocytes (TIL therapy) and chimeric antigen receptor-modified T (CAR-T) cells, is a revolutionary immunotherapy technique that offers the most promising advanced cancer therapeutics, with unrivaled results in hematological malignancies. This has made it possible to treat solid tumors like lung cancer with immunotherapy in new ways. Nonetheless, due to the tumor’s immunosuppressive microenvironment, its efficacy remains inadequate. As a result, in the current environment, employing adoptive cell therapies to improve efficacy, survival, persistence, and safety in solid tumors remains an important frontier in cancer immunotherapy. A general discussion of the biological milieu of lung tumors is provided in this chapter along with recent developments in adoptive T-cell therapy (T-CT) research, the molecular basis of effective lung cancer management, as well as challenges and potential future directions for the clinical use of TIL and CAR-T cell therapy against lung cancer.
Article
Full-text available
Conventional cancer treatments rely on radiotherapy and chemotherapy. Such treatments supposedly mediate their effects via the direct elimination of tumor cells. Here we show that the success of some protocols for anticancer therapy depends on innate and adaptive antitumor immune responses. We describe in both mice and humans a previously unrecognized pathway for the activation of tumor antigen–specific T-cell immunity that involves secretion of the high-mobility-group box 1 (HMGB1) alarmin protein by dying tumor cells and the action of HMGB1 on Toll-like receptor 4 (TLR4) expressed by dendritic cells (DCs). During chemotherapy or radiotherapy, DCs require signaling through TLR4 and its adaptor MyD88 for efficient processing and cross-presentation of antigen from dying tumor cells. Patients with breast cancer who carry a TLR4 loss-of-function allele relapse more quickly after radiotherapy and chemotherapy than those carrying the normal TLR4 allele. These results delineate a clinically relevant immunoadjuvant pathway triggered by tumor cell death.
Article
Transplantation experiments have demonstrated that most mouse tumors express antigens that can constitute targets for rejection responses mediated by syngeneic T lymphocytes. For human tumors, autologous cultures mixing tumor cells and blood lymphocytes or tumor-infiltrating lymphocytes have produced CD8(+) and CD4(+) cytolytic T cell (CTL) clones that recognize tumor cells specifically. Attempts to identify the target antigens by biochemical fractionation of tumor cells up to now have failed, with the important exception of the identification of underglycosylated mucins present on breast and pancreatic carcinomas. Gene transfection approaches have proved more successful. A gene family named MAGE codes for antigens recognized by autologous CTL on a melanoma tumor. These genes are not expressed in normal tissues except for testis. They are expressed in many tumors of several histological types. Differentiation antigens coded by genes such as tyrosinase are also recognized on human melanoma by autologous CTL. The identification of human tumor rejection antigens opens new possibilities for systematic approaches to the specific immune therapy of cancer.
Article
Using a tumor model of spontaneously arising insulinomas expressing a defined tumor-associated antigen, we investigated whether tumor growth promotes cross-presentation and tolerance of tumor-specific T cells. We found that an advanced tumor burden enhanced cross-presentation of tumor-associated antigens to high avidity tumor-specific T cells, inducing T cell proliferation and limited effector function in vivo. However, contrary to other models, tumor-specific T cells were not tolerized despite a high tumor burden. In fact, in tumor-bearing mice, persistence and responsiveness of adoptively transferred tumor-specific T cells were enhanced. Accordingly, a potent T cell–mediated antitumor response could be elicited by intravenous administration of tumor-derived peptide and agonistic anti-CD40 antibody or viral immunization and reimmunization. Thus, in this model, tumor growth promotes activation of high avidity tumor-specific T cells instead of tolerance. Therefore, the host remains responsive to T cell immunotherapy.
Article
Unlabelled: Peripheral CD8 T-cell tolerance can be generated outside lymphatic tissue in the liver, but the course of events leading to tolerogenic interaction of hepatic cell populations with circulating T-cells remain largely undefined. Here we demonstrate that preferential uptake of systemically circulating antigen by murine liver sinusoidal endothelial cells (LSECs), and not by other antigen-presenting cells in the liver or spleen, leads to cross-presentation on major histocompatibility complex (MHC) I molecules, which causes rapid antigen-specific naïve CD8 T-cell retention in the liver but not in other organs. Using bone-marrow chimeras and a novel transgenic mouse model (Tie2-H-2K(b) mice) with endothelial cell-specific MHC I expression, we provide evidence that cross-presentation by organ-resident and radiation-resistant LSECs in vivo was both essential and sufficient to cause antigen-specific retention of naïve CD8 T-cells under noninflammatory conditions. This was followed by sustained CD8 T-cell proliferation and expansion in vivo, but ultimately led to the development of T-cell tolerance. Conclusion: Our results show that cross-presentation of circulating antigens by LSECs caused antigen-specific retention of naïve CD8 T-cells and identify antigen-specific T-cell adhesion as the first step in the induction of T-cell tolerance.
Article
MHC class I-restricted presentation is usually associated with the processing of endogenous antigens. However, this restriction element can present antigens that do not originate within the presenting cells. Here, Francis Carbone and colleagues describe how such cross-presentation is critical for cytotoxic T-cell surveillance of peripheral antigens, both self and foreign, located outside the secondary lymphoid compartment.