ArticlePDF Available

Autoimmunity: Antigen-specific immunotherapy

Authors:

Abstract and Figures

Nanoparticles coated with fragments of the body's own proteins are shown to induce T cells of the immune system to adopt regulatory functions that suppress autoimmune reactions involving these self-antigens. See Article p.434
Content may be subject to copyright.
DAVID WRAITH
A
utoimmune diseases arise when our
immune system attacks our own
tissues. The immune cells of affected
individuals are insufficiently tolerant towards
certain ‘self’ proteins and attack them as if
they were foreign. Helper Tcells (THcells)
play a central part in autoimmune diseases
because they orchestrate the function of other
cells in the immune system, including Bcells,
cytotoxic Tcells and macrophages. Current
treatments for autoimmune diseases tend to
suppress the whole immune system or, at best,
inhibit the movement or function of Tcells;
such approaches inevitably increase the risk
of infection and cancer. The ideal treatment
for autoimmune diseases would convert the
function of THcells from disease-causing
to disease-regulating without affecting the
rest of the immune system. In this issue,
Clemente-Casares etal.
1
(page 434) describe
coated nano particles that mediate this con-
version by binding to receptors on potentially
self-reactive Tcells.
The authors’ approach can be considered
as a type of antigen-specific immunotherapy.
Antigens are the molecular structures that
induce the activation of T or Bcells; for Tcells
these are generally small fragments of proteins
(peptides). Each Tcell can express a different
surface receptor, thereby allowing our immune
system to respond to countless different anti-
gens, including self-antigens. Antigen-specific
immunotherapy is designed to dampen the
immune response to a particular antigen or
set of closely associated antigens. This con-
cept has been used to treat allergies for more
than a century
2
, but specific immunotherapy
for autoimmune diseases lagged behind until
the discovery that T
H
cells are activated by pep-
tides bound to MHC classII proteins. This led
to the design of peptides that selectively target
T
H
cells without risking the activation of self-
reactive cytotoxic Tcells or Bcells3.
How can exposure to a peptide known to
stimulate self-reactive T
H
cells switch off the
disease they cause? This is best explained by
the ‘two-signal’ rule of T-cell activation4,5.
All antigens, whether self or foreign, must be
broken down into peptides, which must then
bind MHC classII proteins and be displayed at
the surface of antigen-presenting cells (APCs)
to activate THcells. This is referred to as
signal1. The APC must also upregulate co-
stimulatory molecules, such as CD80 and
CD86, to provide the second signal required
for TH-cell survival and proliferation.
What happens when THcells receive signal1,
but not signal2? Historically, this was thought
to induce a state of unresponsiveness known
as anergy6. Now, Clemente-Casares etal.
show that treating T
H
cells with nano particles
coated with a peptide bound to MHCclassII
proteins (pMHC-NP treatment) triggers
signal1 alone. But rather than simply inducing
anergy, the treatment drives the THcells to
differentiate into cells that have characteris-
tics of regulatory Tcells; these act to dampen
immune responses.
The resulting regulatory cells exert their
function by secreting the anti-inflammatory
proteins IL-10 and TGF-β. Furthermore, the
cells express the transcription factor T-bet and
make the cytokine signalling molecule IFN-γ
during their differentiation. These character-
istics imply that they derive from cells of the
TH1 subset of THcells (Fig.1). The differen-
tiation of IL-10-secreting Tcells— referred
to here as TR1-like cells — from TH1 cells is
an immunoregulatory mechanism known
to prevent excessive immune responses to a
range of infections7–9. These cells mediate a
negative feedback mechanism involving sup
-
pression of co-stimulatory molecules on APCs
and a reduction in the inflammatory proteins
secreted by APCs10.
What are the downstream effects of the
AUTOIMMUNITY
Antigen-specific immunotherapy
Nanoparticles coated with fragments of the body’s own proteins are shown to induce T cells of the immune system to adopt
regulatory functions that suppress autoimmune reactions involving these self-antigens. S A .434
T
H
1 cell
Nanoparticle
MHC class II
Peptide
T
R
1-like cell
CD8
+
T cell
B cell
Regulatory B cell
IL-10
IL-10
IL-10
Antigen-presenting cell
Inhibition by
T
R
1-like cell
CD4
+
T cell
Figure 1 | Coated nanoparticles induce differentiation of regulatory Tcells. Clemente-Casares etal.1
produced nanoparticles coated with peptide fragments of the body’s own proteins that are associated
with autoimmune disease, bound to MHC classII proteins. They show that treating mice with these
nanoparticles modifies the function of TH1 cells that have receptors specific for that particular peptide:
instead of inducing an immune response against the self-protein, the TH1 cells differentiate into regulatory
(TR1-like) Tcells that secrete the anti-inflammatory protein IL-10. The IL-10 promotes the differentiation
of Bcells into IL-10-secreting regulatory Bcells, and also modifies the ability of antigen-presenting cells
(APCs) to present the specific peptide to immune cells. Furthermore, the TR1-like cells can inhibit the
activation of helper (CD4+) and cytotoxic (CD8+) Tcells that are specific for other peptides presented by the
same APC, and thus mediate bystander suppression. In this way, TR1-like cells can target APCs in tissues
affected by autoimmune reactions and thereby suppress the inflammation associated with the disease.
422 | NATURE | VOL 530 | 25 FEBRUARY 2016
NEWS & VIEWS For News & Views online, go to
nature.com/newsandviews
© 2016 Macmillan Publishers Limited. All rights reserved
ANNA M. KIETRYS & ERIC T. KOOL
T
he fact that chemical modifications to
DNA bases can alter gene expression
without changing the nucleic-acid
sequence has been known for more than a
decade. But the key regulatory function of
many such epigenetic modifications to mes-
senger RNA molecules has been recognized
only recently1–3. A simple mark — the methyl
group — is widely observed in DNA and its
associated histone proteins, and has been
studied in mRNA in the forms of 5-methyl-
cytosine and N6-methyladenosine. On
page441 of this issue, Dominissini etal.4
present a new member of the mRNA methyl-
marked family, N1-methyl adenosine, and
propose that its presence in mRNAs has an
influence on biological processes. What is
surprising about this modification is that it has
previously been described as a form of cellular
damage5.
N1-methyladenosine (m1A) is unusual in
having a positive charge at physiological pH
(other bases are uncharged) and a methyl
group that blocks the Watson–Crick base-
pairing edge of adenine. The modification
was previously documented in transfer RNA
molecules, where it plays a crucial part in
the formation of tertiary RNA structure.
The methyl group forces the m1A base to
pair with a non-Watson–Crick configura-
tion6, and the positive charge has also been
hypothesized to exert an electrostatic influ-
ence on protein interactions7. Dominissini
etal. propose that this base modification may
have similar biophysical effects in mRNAs: it
could affect base-pairing inter actions close
to the site at which protein translation starts,
and might alter RNA folding and electrostatic
interactions.
m1A was previously known to be formed
by the exposure of RNA molecules to alkylat-
ing agents, and under alkaline conditions it
is converted to N6-methyladenosine (m6A)
through the Dimroth rearrangement8 (Fig.1).
This is potentially a big problem in analysis,
TR1-like cells induced by pMHC-NP treat-
ment? Clemente-Casares etal. show that
the cells suppress the function of APCs and
reinforce immune regulation by promot-
ing IL-10 production by Bcells (Fig.1). The
authors verify the specificity of their approach
by using different experimental models of
autoimmune disease. pMHC-NPs carrying
peptides from collagen, an antigen derived
from joints, suppressed disease in a mouse
model of rheumatoid arthritis, but not in mice
with experimental autoimmune encephalitis
(EAE), a model of multiple sclerosis. Con-
versely, pMHC-NPs carrying peptides of
antigens from the central nervous system con-
trolled EAE but not collagen-induced arthri-
tis. This confirms that the immune regulation
induced by pMHC-NP treatment is specific to
the antigen and tissue, and so to the disease.
Furthermore, the pMHC-NPs did not need
to target Tcells specific for all peptides in the
affected organ. Even peptides from sub-domi-
nant antigens (weaker antigens that do not trig-
ger disease in the first place) were able to induce
TR1-like cells that suppressed helper and cyto-
toxic Tcells with activity against other antigens
(Fig.1). Thus, although this treatment is highly
antigen-specific at the induction phase, it can
influence other arms of the immune response
locally, through induction of regulatory B-cell
activity and suppression of helper and cyto-
toxic Tcells specific for different antigens.
This requires that the peptide fragment from
the inducing antigen and the other antigens are
presented by the same APC.
Is it possible that such bystander suppression
could lead to systemic immune suppres -
sion by switching off cells not involved in the
autoimmune response, thereby increasing
the risk of infection or cancer? No: bystander
suppression will be limited to lymph nodes
associated with the affected organ and will
influence only those APCs presenting the
relevant self-antigen. Such specificity is clearly
demonstrated by Clemente-Casares and col-
leagues— mice treated with pMHC-NPs are
protected against the relevant autoimmune
disease, yet show undiminished responses to
infections and foreign antigens.
The experimental treatments in this study
use well-character-
ized models of auto-
immune disease.
But is this work just
another therapeu-
tic approach that
works in mice but
will never work in
humans? It seems
not: the authors
show that pMHC-
NP treatment leads
to differentiation
and proliferation of human TR1-like cells in
immunodeficient mice transplanted with
human T and Bcells, demonstrating that
pMHC-NP treatment works on human cells.
The teams work also suggests that treatment
with pMHC-NPs is more effective than with
monomers of MHC-bound peptides at an
equivalent dose. Furthermore, pMHC-NP
treatment seems to be more suppressive than
the application of peptide alone; however, the
doses and routes of administration in these tests
were not comparable.
There is overwhelming evidence that
peptide antigens can induce TR1-like cells11
and suppress autoimmune diseases in both
mice and humans
9
. The fact that pMHC-NP
treatment induces TR1-like cells similar to
those seen after the administration of peptide
alone suggests that pMHC-NPs mimic the
APC to which therapeutic peptides bind
invivo. The challenge with each of these
approaches will be to find the optimal dose and
route of administration for treating people. As
these options progress towards clinical trials, it
is vital that their mechanism of action is inves-
tigated in detail so that patients can benefit
fully from antigen-specific immunotherapy for
auto immune disease.
David Wraith is at the School of Cellular and
Molecular Medicine, University of Bristol,
BristolBS8 1TD, UK.
e-mail: d.c.wraith@bristol.ac.uk
1. Clemente-Casares, X. et al. Nature 530, 434–440
(2016).
2. Noon, L. Lancet i, 1572–1573 (1911).
3. Larché, M. & Wraith, D. C. Nature Med. 11, S69–S76
(2005).
4. Lafferty, K. J. & Cunningham, A. J. Aust. J. Exp. Biol.
Med. Sci. 53, 27–42 (1975).
5. Baxter, A. G. & Hodgkin, P. D. Nature Rev. Immunol.
2, 439–446 (2002).
6. Schwartz, R. H. Annu. Rev. Immunol. 21, 305–334
(2003).
7. O’Garra, A., Vieira, P. L., Vieira, P. & Goldfeld, A. E.
J.Clin. Invest. 114, 1372–1378 (2004).
8. Trinchieri, G. J. Exp. Med. 204, 239–243 (2007).
9. Sabatos-Peyton, C. A., Verhagen, J. & Wraith, D. C.
Curr. Opin. Immunol. 22, 609–615 (2010).
10. Gabryšová, L. et al. J. Exp. Med. 206, 1755–1767
(2009).
11. Burton, B. R. et al. Nature Commun. 5, 4741
(2014).
The author declares competing financial interests.
See go.nature.com/ukjrkv for details.
This article was published online on 17 February 2016.
EPIGENETICS
A new methyl mark
on messengers
The presence of an N1 methyl group on adenine bases in DNA and RNA was
thought to be a form of damage. Results now show that it also occurs at specific
sites in messenger RNAs, where it affects protein expression. S A .441
“The treatment
drives the TH cell
to differentiate
into cells with
characteristics of
regulatory T cells;
these act to
dampen immune
responses.”
25 FEBRUARY 2016 | VOL 530 | NATURE | 423
NEWS & VIEWS RESEARCH
© 2016 Macmillan Publishers Limited. All rights reserved
... The IS is the body's natural defense against pathogens and foreign materials. Considerable research has been conducted to either manipulate the IS to prevent allergy and autoimmune reactions or harness the IS to cure cancer and infectious illnesses [16,17]. These defensive systems affect a wide variety of organs, as well as cells, tissues, and chemical mediators of IS. ...
Article
Full-text available
Immunotherapy shows a lot of promise for addressing the problems with traditional cancer treatments. Researchers and clinicians are working to create innovative immunological techniques for cancer detection and treatment that are more selective and have lower toxicity. An emerging field in cancer therapy, immunomodulation offers patients an alternate approach to treating cancer. These therapies use the host’s natural defensive systems to identify and remove malignant cells in a targeted manner. Cancer treatment is now undergoing somewhat of a revolution due to recent developments in nanotechnology. Diverse nanomaterials (NMs) have been employed to overcome the limits of conventional anti-cancer treatments such as cytotoxic, surgery, radiation, and chemotherapy. Aside from that, NMs could interact with live cells and influence immune responses. In contrast, unexpected adverse effects such as necrosis, hypersensitivity, and inflammation might result from the immune system (IS)’s interaction with NMs. Therefore, to ensure the efficacy of immunomodulatory nanomaterials, it is essential to have a comprehensive understanding of the intricate interplay that exists between the IS and NMs. This review intends to present an overview of the current achievements, challenges, and improvements in using immunomodulatory nanomaterials (iNMs) for cancer therapy, with an emphasis on elucidating the mechanisms involved in the interaction between NMs and the immune system of the host.
... Another subpopulation of CD4 + T-derived regulatory T cells, CD4 + CD25 -FOXP3 -T-regulatory type 1 (Tr1) cells have the ability to secrete the anti-inflammatory cytokines IL-10 and IL-21, and also express surface markers CD49b, LAG-3, and transcription factor c-Maf (Gagliani et al., 2013). NPs coated with ADs-associated antigenic peptide-MHC (pMHC) complex can attenuate ADs by reprogramming homologous effector T cells into disease-inhibiting Tr1 cells and undergoing massive expansion (Wraith, 2016). On the foundation of the aforementioned concept, Parvus Therapeutics, a biopharmaceutical company dedicated to developing specific immunomodulatory drugs for ADs, developed a Navicims platform technology for the treatment of MS, type 1 diabetes (T 1 D), RA, and other ADs. ...
Article
Full-text available
Rheumatoid arthritis (RA) is a systemic autoimmune disease (AD) caused by the aberrant attack of the immune system on its own joint tissues. Genetic and environmental factors are the main reasons of immune system impairment and high incidence of RA. Although there are medications on the market that lessen disease activity, there is no known cure for RA, and patients are at risk in varying degrees of systemic immunosuppression. By transporting (encapsulating or surface binding) RA-related self-antigens, nucleic acids, immunomodulators, or cytokines, tolerogenic nanoparticles—also known as immunomodulatory nano-preparations—have the potential to gently regulate local immune responses and ultimately induce antigen-specific immune tolerance. We review the recent advances in immunomodulatory nano-preparations for delivering self-antigen or self-antigen plus immunomodulator, simulating apoptotic cell avatars in vivo, acting as artificial antigen-presenting cells, and based on scaffolds and gels, to provide a reference for developing new immunotherapies for RA.
... With the emergence of antigen-specific immunotherapies, the interest in identifying novel autoantigens has intensified alongside their potential as future treatment targets (18). This treatment strategy is hypothesized to be effective while safer than currently available broadly acting immune-modulatory agents associated with risks for side effects such as infections and possibly cancer (19,20). In animal models, where the autoantigens are both limited and defined, this strategy has shown promising results (21,22), whereas in clinical trials in MS treatments targeting myelin antigens, they have been, if safe, of modest efficacy at best (23)(24)(25)(26)(27). ...
Article
Full-text available
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS), in which pathological T cells, likely autoimmune, play a key role. Despite its central importance, the autoantigen repertoire remains largely uncharacterized. Using a novel in vitro antigen delivery method combined with the Human Protein Atlas library, we screened for T cell autoreactivity against 63 CNS-expressed proteins. We identified four previously unreported autoantigens in MS: fatty acid-binding protein 7, prokineticin-2, reticulon-3, and synaptosomal-associated protein 91, which were verified to induce interferon-γ responses in MS in two cohorts. Autoreactive profiles were heterogeneous, and reactivity to several autoantigens was MS-selective. Autoreactive T cells were predominantly CD4+ and human leukocyte antigen-DR restricted. Mouse immunization induced antigen-specific responses and CNS leukocyte infiltration. This represents one of the largest systematic efforts to date in the search for MS autoantigens, demonstrates the heterogeneity of autoreactive profiles, and highlights promising targets for future diagnostic tools and immunomodulatory therapies in MS.
Article
Full-text available
As a chronic autoinflammatory condition, ulcerative colitis is often managed via systemic immunosuppressants. Here we show, in three mouse models of established ulcerative colitis, that a subcutaneously injected colon-specific immunosuppressive niche consisting of colon epithelial cells, decellularized colon extracellular matrix and nanofibres functionalized with programmed death-ligand 1, CD86, a peptide mimic of transforming growth factor-beta 1, and the immunosuppressive small-molecule leflunomide, induced intestinal immunotolerance and reduced inflammation in the animals’ lower gastrointestinal tract. The bioengineered colon-specific niche triggered autoreactive T cell anergy and polarized pro-inflammatory macrophages via multiple immunosuppressive pathways, and prevented the infiltration of immune cells into the colon’s lamina propria, promoting the recovery of epithelial damage. The bioengineered niche also prevented colitis-associated colorectal cancer and eliminated immune-related colitis triggered by kinase inhibitors and immune checkpoint blockade.
Article
Full-text available
Nonspecific immunotherapies often induce general immune activation or suppression. Conversely, antigen‐specific immunotherapy, which refers to dampening or augmenting adaptive immunity against a disease‐specific antigen, increases T‐cell target specificity to pathological tissues, thereby reducing side effects on the rest of the immune system. Advances in engineering strategies for nanomaterials have enabled the feasible modulation of their physicochemical features to incorporate antigens and inherently interact with innate immune cells, which remarkably amplifies the orchestration of antigen‐specific immune responses against cancer and autoimmune diseases. From this contemporary perspective, the basic principles of antigen‐specific immunotherapy are briefly introduced and we elucidate how the latest nanoengineering paradigms regulate the functions of heterogeneous subsets of immune cells, such as antigen‐presenting cells, B cells, and regulatory or cytotoxic T cells, promoting antigen‐specific immunotherapy to treat autoimmune diseases and cancer. An outlook on prospects and remaining challenges have been discussed for, translating scientific discoveries of powerful nanomaterials into medical advances in antigen‐specific immunotherapy, thus offering new treatment modalities for patients with unmet needs.
Article
Full-text available
BACKGROUND Antigen-specific regulation of autoimmune disease is a major goal. In seropositive rheumatoid arthritis (RA), T cell help to autoreactive B cells matures the citrullinated (Cit) antigen-specific immune response, generating RA-specific V domain glycosylated anti-Cit protein antibodies (ACPA VDG) before arthritis onset. Low or escalating antigen administration under "sub-immunogenic" conditions favors tolerance. We explored safety, pharmacokinetics, and immunological and clinical effects of s.c. DEN-181, comprising liposomes encapsulating self-peptide collagen II259-273 (CII) and NF-κB inhibitor 1,25-dihydroxycholecalciferol.METHODSA double-blind, placebo-controlled, exploratory, single-ascending-dose, phase I trial assessed the impact of low, medium, and high DEN-181 doses on peripheral blood CII-specific and bystander Cit64vimentin59-71-specific (Cit-Vim-specific) autoreactive T cell responses, cytokines, and ACPA in 17 HLA-DRB1*04:01+ or *01:01+ ACPA+ RA patients on methotrexate.RESULTSDEN-181 was well tolerated. Relative to placebo and normalized to baseline values, Cit-Vim-specific T cells decreased in patients administered medium and high doses of DEN-181. Relative to placebo, percentage of CII-specific programmed cell death 1+ T cells increased within 28 days of DEN-181. Exploratory analysis in DEN-181-treated patients suggested improved RA disease activity was associated with expansion of CII-specific and Cit-Vim-specific T cells; reduction in ACPA VDG, memory B cells, and inflammatory myeloid populations; and enrichment in CCR7+ and naive T cells. Single-cell sequencing identified T cell transcripts associated with tolerogenic TCR signaling and exhaustion after low or medium doses of DEN-181.CONCLUSION The safety and immunomodulatory activity of low/medium DEN-181 doses provide rationale to further assess antigen-specific immunomodulatory therapy in ACPA+ RA.TRIAL REGISTRATIONAnzctr.org.au identifier ACTRN12617001482358, updated September 8, 2022.FUNDINGInnovative Medicines Initiative 2 Joint Undertaking (grant agreement 777357), supported by European Union's Horizon 2020 research and innovation programme and European Federation of Pharmaceutical Industries and Associations; Arthritis Queensland; National Health and Medical Research Council (NHMRC) Senior Research Fellowship; and NHMRC grant 2008287.
Article
Free energy perturbation (FEP) calculations can predict relative binding affinities of an antigen and its point mutants to the same human leukocyte antigen (HLA) with high accuracy (e.g., within 1.0 kcal/mol to experiment); however, a more challenging task is to compare binding affinities of wholly different antigens binding to completely different HLAs using FEP. Researchers have used a variety of different FEP schemes to compute and compare absolute binding affinities, with varied success. Here, we propose and assess a unifying scheme to compute the relative binding affinities of different antigens binding to completely different HLAs using absolute binding affinity FEP calculations. We apply our affinity calculation technique to HLA-antigen-T-cell receptor (TCR) systems relevant to celiac disease (CeD) by investigating binding affinity differences between HLA-DQ2.5 (enhanced CeD risk) and HLA-DQ7.5 (CeD protective) in the binary (HLA-gliadin) and ternary (HLA-gliadin-TCR) binding complexes for three gliadin derived epitopes: glia-α1, glia-α2, and glia-ω1. Based on FEP calculations with our carefully designed thermodynamic cycles, we demonstrate that HLA-DQ2.5 has higher binding affinity than HLA-DQ7.5 for gliadin and enhanced binding affinity with a common TCR, agreeing with known results that the HLA-DQ2.5 serotype exhibits increased risk for CeD. Our findings reveal that our proposed absolute binding affinity FEP method is appropriate for predicting HLA binding for disparate antigens with different genotypes. We also discuss atomic-level details of HLA genotypes interacting with gluten peptides and TCRs in regard to the pathogenesis of CeD.
Article
Full-text available
Autoimmune diseases, especially among young people in the US, are one of the leading causes of morbidity and death. The immune responses are the fundamental pathogenicity of autoimmune disorders. The equilibrium between stimulatory and inhibitory signals is critical for the stimulation, migration, survival, and T cell-related immune responses. The B7 family can substantially regulate T cell-mediated immune responses. Nevertheless, recent breakthroughs in immune checkpoint blockade in cancer immunotherapy have facilitated autoimmune diseases, especially among the prone populations. In the current study, we tried to concisely review the role of the B7 family in regulating immune reactions and the influence of immune checkpoint inhibitors on autoimmunity development.
Article
Full-text available
Regulatory T cells hold promise as targets for therapeutic intervention in autoimmunity, but approaches capable of expanding antigen-specific regulatory T cells in vivo are currently not available. Here we show that systemic delivery of nanoparticles coated with autoimmune-disease-relevant peptides bound to major histocompatibility complex class II (pMHCII) molecules triggers the generation and expansion of antigen-specific regulatory CD4(+) T cell type 1 (TR1)-like cells in different mouse models, including mice humanized with lymphocytes from patients, leading to resolution of established autoimmune phenomena. Ten pMHCII-based nanomedicines show similar biological effects, regardless of genetic background, prevalence of the cognate T-cell population or MHC restriction. These nanomedicines promote the differentiation of disease-primed autoreactive T cells into TR1-like cells, which in turn suppress autoantigen-loaded antigen-presenting cells and drive the differentiation of cognate B cells into disease-suppressing regulatory B cells, without compromising systemic immunity. pMHCII-based nanomedicines thus represent a new class of drugs, potentially useful for treating a broad spectrum of autoimmune conditions in a disease-specific manner.
Article
Full-text available
Nearly a century has passed since the first report describing antigen-specific immunotherapy (antigen-SIT) was published. Research into the use of antigen-SIT in the treatment of both allergic and autoimmune disease has increased dramatically since, although its mechanism of action is only slowly being unravelled. It is clear though, from recent studies, that success of antigen-SIT depends on the induction of regulatory T (T reg) cell subsets that recognise potentially disease-inducing epitopes. The major challenge remaining for the widespread use of antigen-SIT is to safely administer high doses of immunodominant and potentially pathogenic epitopes in a manner that induces T cell tolerance rather than activation. This review illustrates that intelligent design of treatment agents and strategies can lead to the development of safe and effective antigen-SIT.
Article
Full-text available
Regulation of the immune response to self- and foreign antigens is vitally important for limiting immune pathology associated with both infections and hypersensitivity conditions. Control of autoimmune conditions can be reinforced by tolerance induction with peptide epitopes, but the mechanism is not currently understood. Repetitive intranasal administration of soluble peptide induces peripheral tolerance in myelin basic protein (MBP)-specific TCR transgenic mice. This is characterized by the presence of anergic, interleukin (IL)-10-secreting CD4(+) T cells with regulatory function (IL-10 T reg cells). The differentiation pathway of peptide-induced IL-10 T reg cells was investigated. CD4(+) T cells became anergic after their second encounter with a high-affinity MBP peptide analogue. Loss of proliferative capacity correlated with a switch from the Th1-associated cytokines IL-2 and interferon (IFN)-gamma to the regulatory cytokine IL-10. Nevertheless, IL-10 T reg cells retained the capacity to produce IFN-gamma and concomitantly expressed T-bet, demonstrating their Th1 origin. IL-10 T reg cells suppressed dendritic cell maturation, prevented Th1 cell differentiation, and thereby created a negative feedback loop for Th1-driven immune pathology. These findings demonstrate that Th1 responses can be self-limiting in the context of peripheral tolerance to a self-antigen.
Article
Full-text available
Two-signal theories of lymphocyte activation have evolved considerably over the past 35 years. In this article, we examine the contemporary experimental observations and theoretical concerns that have helped to forge the most influential variants of the theory. We also propose that more-rigorous quantitative methods are required to sustain theoretical development in the future.
Article
Full-text available
Allergic and autoimmune diseases are forms of immune hypersensitivity that increasingly cause chronic ill health. Most current therapies treat symptoms rather than addressing underlying immunological mechanisms. The ability to modify antigen-specific pathogenic responses by therapeutic vaccination offers the prospect of targeted therapy resulting in long-term clinical improvement without nonspecific immune suppression. Examples of specific immune modulation can be found in nature and in established forms of immune desensitization. Understanding and exploiting common mechanisms such as the ability to induce antigen-specific regulatory cells should allow the development of effective therapeutic strategies for both forms of immunopathology. Targeting pathogenic T cells using vaccines consisting of synthetic peptides representing T cell epitopes is one such strategy that is currently being evaluated with encouraging results. Future challenges in the development of therapeutic vaccines include selection of appropriate antigens and peptides, optimization of peptide dose and route of administration and identifying strategies to induce bystander suppression.
Article
Full-text available
Interleukin (IL)-10 is a cytokine that modulates both innate and adaptive immunity, primarily by exerting antiinflammatory effects. IL-10 was originally thought to be produced only by T helper (Th)2 cells, but is now known to be made by a variety of cell types. During many infections, CD4(+) T cells produce both interferon (IFN)-gamma, the signature Th1 cytokine, and IL-10. New data now show that the IL-10 produced by effector Th1 cells helps limit the collateral damage caused by exaggerated inflammation. But this control may also limit the effectiveness of the immune response, resulting in a failure to fully eliminate pathogens.
Article
Antigen-specific immunotherapy combats autoimmunity or allergy by reinstating immunological tolerance to target antigens without compromising immune function. Optimization of dosing strategy is critical for effective modulation of pathogenic CD4(+) T-cell activity. Here we report that dose escalation is imperative for safe, subcutaneous delivery of the high self-antigen doses required for effective tolerance induction and elicits anergic, interleukin (IL)-10-secreting regulatory CD4(+) T cells. Analysis of the CD4(+) T-cell transcriptome, at consecutive stages of escalating dose immunotherapy, reveals progressive suppression of transcripts positively regulating inflammatory effector function and repression of cell cycle pathways. We identify transcription factors, c-Maf and NFIL3, and negative co-stimulatory molecules, LAG-3, TIGIT, PD-1 and TIM-3, which characterize this regulatory CD4(+) T-cell population and whose expression correlates with the immunoregulatory cytokine IL-10. These results provide a rationale for dose escalation in T-cell-directed immunotherapy and reveal novel immunological and transcriptional signatures as surrogate markers of successful immunotherapy.
Article
Allogeneic reactions have conventionally been considered as typical immune responses by one population of cells to antigens present on the other. This view is inadequate, since it does not explain many features of these reactions, among which are: (1) reactivity is much higher between different strains within a species than between species, in spite of the much greater antigenic disparity in the second case; (2) a very high proportion of cells may respond to allogeneic stimuli; (3) major histocompatibility differences are not essential for vigorous allogeneic reactions; (4) the responding population need not be immunologically competent to respond to antigens of the stimulating population; (5) the stimulating population must be both metabolically active and immunocompetent.
Article
T cell anergy is a tolerance mechanism in which the lymphocyte is intrinsically functionally inactivated following an antigen encounter, but remains alive for an extended period of time in a hyporesponsive state. Models of T cell anergy affecting both CD4(+) and CD8(+) cells fall into two broad categories. One, clonal anergy, is principally a growth arrest state, whereas the other, adaptive tolerance or in vivo anergy, represents a more generalized inhibition of proliferation and effector functions. The former arises from incomplete T cell activation, is mostly observed in previously activated T cells, is maintained by a block in the Ras/MAP kinase pathway, can be reversed by IL-2 or anti-OX40 signaling, and usually does not result in the inhibition of effector functions. The latter is most often initiated in naïve T cells in vivo by stimulation in an environment deficient in costimulation or high in coinhibition. Adaptive tolerance can be induced in the thymus or in the periphery. The cells proliferate and differentiate to varying degrees and then downregulate both functions in the face of persistent antigen. The state involves an early block in tyrosine kinase activation, which predominantly inhibits calcium mobilization, and an independent mechanism that blocks signaling through the IL-2 receptor. Adaptive tolerance reverses in the absence of antigen. Aspects of both of the anergic states are found in regulatory T cells, possibly preventing them from dominating initial immune responses to foreign antigens and shutting down such responses prematurely.
Article
Effective immune responses against pathogens are sometimes accompanied by strong inflammatory reactions. To minimize damage to self, the activation of the immune system also triggers anti-inflammatory circuits. Both inflammatory and anti-inflammatory reactions are normal components of the same immune response, which coordinately fight infections while preventing immune pathology. IL-10 is an important suppressive cytokine, produced by a large number of immune cells in addition to the antigen-driven IL-10-producing regulatory and the naturally occurring suppressor CD4+ T cells, which is a key player in anti-inflammatory immune responses. However, additional mechanisms have evolved to ensure that pathogen eradication is achieved with minimum damage to the host. Here we discuss those mechanisms that operate to regulate effector immune responses.