Fact-checked by Grok 2 weeks ago

Peripheral tolerance

Peripheral tolerance is a critical immunological mechanism that establishes and maintains unresponsiveness to self-antigens in peripheral tissues, preventing the activation of autoreactive T and B lymphocytes that escape central tolerance in the and . The importance of peripheral tolerance was recognized by the 2025 Nobel Prize in or awarded to Mary E. Brunkow, Fred Ramsdell, and Shimon Sakaguchi for their discoveries concerning regulatory T cells and peripheral . This process occurs primarily in secondary lymphoid organs and peripheral sites, ensuring by inhibiting unwanted immune responses that could lead to , allergies, or . Unlike central tolerance, which eliminates many self-reactive cells during lymphocyte development, peripheral tolerance acts as a secondary safeguard, employing diverse strategies to control potentially harmful lymphocytes without compromising effective immunity against pathogens. The primary mechanisms of peripheral tolerance include anergy, deletion, ignorance, and suppression. Anergy induces a state of functional hyporesponsiveness in self-reactive T cells upon recognition without sufficient costimulatory signals, such as those provided by CD28-B7 interactions, rendering the cells unable to proliferate or produce cytokines. Deletion involves the apoptotic elimination of autoreactive s, often triggered by strong TCR signaling in the absence of survival signals or through pro-apoptotic pathways like Bim and in T cells. arises from physical or molecular barriers that limit autoreactive T cell access to self-s in peripheral tissues, such as in immune-privileged sites or low-affinity peptide-MHC interactions below activation thresholds. Suppression is mediated by regulatory T cells (Tregs) and regulatory B cells (Bregs), which secrete immunosuppressive cytokines like IL-10, TGF-β, and IL-35 to inhibit effector activity and promote tolerance. These mechanisms collectively prevent autoimmune diseases by balancing immune vigilance with self-tolerance, and their dysregulation is implicated in conditions such as , , and allergic disorders. For instance, induced Tregs generated in the upon exposure can expand to dampen ongoing responses, highlighting the adaptive nature of peripheral tolerance. Ongoing research emphasizes the role of tissue-specific factors, such as maturation states and milieus, in fine-tuning these processes to adapt to environmental challenges while preserving self-tolerance.

Introduction

Definition and overview

Peripheral tolerance refers to the collection of immunological mechanisms that establish and maintain unresponsiveness to self-antigens in mature lymphocytes after they have exited the primary lymphoid organs, such as the for T cells and the for B cells. These processes ensure that potentially autoreactive lymphocytes, which may have escaped initial negative selection, do not initiate harmful immune responses against the body's own tissues. Unlike central tolerance, which primarily occurs during lymphocyte development in primary lymphoid organs, peripheral tolerance operates in secondary lymphoid tissues and peripheral sites to reinforce immune self-nonself discrimination. The mechanisms of peripheral tolerance broadly encompass active suppression mediated by regulatory cells, intrinsic hyporesponsiveness in lymphocytes through processes like anergy and deletion, and sequestration of antigens in immunologically privileged environments to limit exposure. These pathways collectively prevent the activation of self-reactive T and B cells upon encounter in the , thereby preserving immune . For instance, self-s presented in the absence of appropriate co-stimulatory signals can render lymphocytes tolerant rather than responsive. The concept of peripheral tolerance emerged from foundational experiments in the 1950s, notably through studies on allograft rejection and acquired tolerance conducted by Peter Medawar and colleagues, who demonstrated that neonatal exposure to foreign tissues could induce long-term unresponsiveness to those antigens in mice. This work, building on earlier observations of transplantation immunity, earned Medawar and Frank Macfarlane Burnet the 1960 Nobel Prize in Physiology or Medicine for discovering immunological tolerance. The critical role of regulatory T cells in peripheral tolerance was further highlighted by the 2025 Nobel Prize in Physiology or Medicine, awarded to Mary E. Brunkow, Fred Ramsdell, and Shimon Sakaguchi for their discoveries concerning these immune regulatory cells. Peripheral tolerance is crucial for preventing autoimmunity while permitting effective responses to pathogens and tumors; its disruption has been implicated in diseases such as type 1 diabetes, where failure to suppress self-reactive lymphocytes leads to beta-cell destruction.

Relation to central tolerance

Central tolerance forms the primary barrier against by eliminating or inactivating highly self-reactive lymphocytes during their development in primary lymphoid organs. In T cells, this occurs through negative selection in the , where double-positive thymocytes recognizing self-peptides presented on (MHC) molecules with high undergo , primarily mediated by pro-apoptotic proteins such as Bim and Nur77 in medullary thymic epithelial cells and dendritic cells. For B cells, central tolerance takes place in the , involving clonal deletion via or receptor editing—rearrangement of immunoglobulin light chains to alter specificity—of immature B cells that bind self-antigens with strong , reducing autoreactive clones from an estimated 50-80% in early stages to about 10% in mature naive B cells. However, central tolerance is imperfect and allows a significant fraction of low-affinity self-reactive T cells to escape into the circulation, with studies indicating that 25-40% of T cells reactive to certain self-peptides persist despite thymic deletion mechanisms. This escape arises because negative selection thresholds spare cells with weaker self-reactivity, and not all tissue-specific or peripheral self-antigens are adequately expressed in the or due to factors like the (AIRE) protein's limited scope. Consequently, peripheral tolerance serves as a complementary safeguard, providing ongoing to suppress or inactivate these escaped autoreactive cells in response to antigens encountered in secondary lymphoid organs and tissues. The evolutionary rationale for this dual-layered system lies in its enhancement of self-nonself discrimination, where central mechanisms efficiently cull developmental threats to minimize risk, while peripheral layers adaptively handle diverse, post-developmental exposures to maintain immune without compromising pathogen defense. In contrast to the primarily deletional and fixed processes of central tolerance during maturation, peripheral tolerance employs a broader repertoire of adaptive strategies, including functional inactivation (anergy), localized deletion, and active suppression, specifically tuned to the dynamic presentation of peripheral antigens.

Cellular mediators

Regulatory T cells

Regulatory T cells (Tregs), particularly +Foxp3+ Tregs, play a central role in maintaining peripheral tolerance by actively suppressing autoreactive immune responses and preventing . These cells constitute approximately 5-10% of peripheral + T cells and are essential for immune in non-lymphoid tissues. There are two primary types of Tregs: natural Tregs (nTregs), which develop in the from high-affinity self-reactive T cell precursors and express constitutively, and induced Tregs (iTregs), which are generated in the periphery from naive + T cells under tolerogenic conditions, such as exposure to TGF-β and IL-2 in the presence of suboptimal stimulation. Tregs exert their suppressive effects through multiple mechanisms that target effector T cells, antigen-presenting cells (APCs), and the inflammatory microenvironment. Secretion of immunosuppressive cytokines like IL-10 and TGF-β inhibits effector T cell and while promoting tissue repair. CTLA-4 expression on Tregs competes with on effector T cells for binding to / on APCs, downregulating costimulatory signals and inducing (IDO) production for further suppression. Additionally, Tregs can mediate direct cytotoxicity against target cells via and perforin, and disrupt metabolism by generating through CD39/CD73 ectoenzymes, which activates receptors to inhibit T cell activation. The development and maintenance of Tregs are tightly regulated by key signaling pathways. , a essential for Treg identity and function, is induced during nTreg thymic selection and iTreg peripheral ; its expression is stabilized by IL-2 signaling via STAT5, which is critical for Treg and proliferation. IL-2 deprivation leads to reduced Treg numbers and impaired suppressive capacity. Defects in , often due to mutations, result in immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, characterized by severe autoimmunity including , eczema, and from birth. Experimental evidence underscores the protective role of Tregs in peripheral tolerance. In non-obese diabetic (NOD) mice, a model of spontaneous type 1 diabetes, adoptive transfer of polyclonal or antigen-specific Tregs prevents disease onset by inhibiting diabetogenic T cell responses and preserving beta cell integrity, with protection rates exceeding 80% in some studies. Similarly, Treg transfer into immunodeficient NOD.scid recipients blocks autoimmune diabetes transfer, highlighting their dominant suppressive function in vivo.

Tolerogenic antigen-presenting cells

Tolerogenic antigen-presenting cells (APCs), including dendritic cells (DCs) and macrophages, actively promote peripheral tolerance by presenting self-antigens and innocuous environmental antigens in ways that dampen rather than drive immune responses. These cells differ from immunogenic APCs by exhibiting reduced expression of costimulatory molecules such as and , thereby delivering antigen-specific signals without sufficient second signals for full T cell activation. This low-costimulation environment favors outcomes like T cell anergy or (Treg) differentiation over effector responses.00039-7) Additionally, tolerogenic APCs often engage inhibitory pathways, including of self-antigens to + T cells in the absence of , which induces deletion or dysfunction of autoreactive clones. Among DCs, specialized subsets such as CD103+ conventional DCs in the intestinal are key mediators of tolerance to commensal and dietary antigens in (GALT). These cells express (IDO), which catabolizes to generate immunosuppressive metabolites, and programmed death-ligand 1 (), which engages PD-1 on T cells to inhibit proliferation and production.00819-2)00144-9) Tolerogenic DCs are conditioned by the local microenvironment, particularly anti-inflammatory s like interleukin-10 (IL-10) and transforming growth factor-β (TGF-β), which downregulate proinflammatory maturation and enhance tolerogenic . In the steady state, CD103+ DCs sample antigens from the gut and migrate to mesenteric nodes, where they imprint T cells with gut-homing properties while promoting Treg to prevent aberrant responses against harmless microbes.00345-6) Tolerogenic macrophages, often exhibiting an M2-like in peripheral tissues, contribute to by producing , a metabolite that supports + Treg differentiation and enhances gut-homing receptor expression on T cells.00270-6) These macrophages present antigens with minimal and secrete factors, reinforcing a suppressive milieu that limits effector T cell expansion. In the GALT, M2-like macrophages collaborate with DCs to maintain against commensals. The critical role of tolerogenic APCs is underscored by studies using DC-specific genetic knockouts, such as those disrupting JAK1 signaling in DCs, which result in impaired expression, reduced Treg generation, and spontaneous in models of and other inflammatory diseases.00144-9) Similarly, of specific DC subsets in the gut leads to breakdown of commensal tolerance and heightened susceptibility to autoimmune pathology, highlighting their non-redundant function in peripheral immune regulation.30145-X)

Tissue-specific mechanisms

Lymph node stromal cells

Lymph node stromal cells (LNSCs) are non-hematopoietic cells that form the structural scaffold of secondary lymphoid organs and play a critical role in maintaining peripheral tolerance by presenting self-antigens to T cells. These cells include fibroblastic reticular cells (FRCs), which support the T cell zone; lymphatic endothelial cells (LECs), lining the lymphatic sinuses; and blood endothelial cells (BECs), associated with high endothelial venules. Unlike professional antigen-presenting cells, LNSCs constitutively express major histocompatibility complex class II (MHC II) molecules and present peripheral tissue-restricted antigens (PTAs) in an Aire-independent manner, promoting T cell deletion, anergy, or regulatory T cell (Treg) induction to prevent autoimmunity. FRCs, LECs, and BECs acquire and display self-antigens through direct transcription or transfer from dendritic cells, enabling continuous surveillance of self-reactivity in lymph nodes. LECs, in particular, express high levels of PTAs and induce + T cell deletion via programmed death-ligand 1 () expression, coupled with a lack of costimulatory signals, leading to rapid PD-1 upregulation and in self-reactive T cells. FRCs contribute by producing such as CCL21, which guides CCR7-expressing Tregs to the T cell zone, enhancing suppressive functions and supporting tolerance in steady-state conditions. BECs similarly present antigens but to a lesser extent, aiding in the compartmentalization of immune responses. In draining lymph nodes, LNSCs maintain systemic tolerance by ensuring self-antigens from peripheral tissues are encountered under non-inflammatory conditions; for instance, LEC-specific expression can result in T ignorance for low-avidity clones or deletion for high-avidity ones, preventing effector responses. This process interacts briefly with Tregs, as LNSC-presented antigens promote Treg and recruitment to suppress potential autoreactivity. Genetic models demonstrate the essentiality of this role: conditional of MHC in LNSCs leads to dysregulated T responses, reduced Treg maintenance, and increased , such as spontaneous in aged mice.

Immunoprivileged sites

Immunoprivileged sites represent specialized anatomical locations where peripheral tolerance is actively maintained through a combination of physical barriers and local immunosuppressive mechanisms, safeguarding essential tissues from autoimmune damage. These sites, including the anterior chamber of the eye, the brain, the testes, and the pregnant , evolved to protect non-regenerative or developmentally critical structures by limiting immune infiltration and modulating inflammatory responses. In the eye, immune privilege is enforced by the blood-ocular barrier, formed by tight junctions in retinal pigment epithelium and endothelial cells, which restricts access of immune effectors to intraocular antigens. Additional mechanisms include high expression of Fas ligand (FasL) on ocular tissues, which induces apoptosis in infiltrating Fas-expressing T cells, and local production of transforming growth factor-β (TGF-β) that promotes regulatory T cell (Treg) differentiation and suppresses pro-inflammatory cytokines. Indoleamine 2,3-dioxygenase (IDO) activity in ocular antigen-presenting cells further depletes tryptophan, inhibiting T cell proliferation. The relative paucity of lymphatic vessels in the eye limits antigen drainage to draining lymph nodes, reducing systemic priming of autoreactive lymphocytes. A key example is anterior chamber-associated immune deviation (ACAID), where antigens introduced into the anterior chamber are processed by specialized F4/80+ macrophages that migrate to the spleen and induce antigen-specific Tregs and NKT cells, deviating immune responses toward tolerance rather than delayed-type hypersensitivity. In contrast, breaching this privilege, as in sympathetic ophthalmia following penetrating eye injury, exposes sequestered uveal antigens to the systemic immune system via conjunctival lymphatics, triggering bilateral granulomatous uveitis through autoreactive CD4+ T cell activation. The brain maintains peripheral tolerance via the (BBB), a tight junction-sealed endothelial layer that prevents parenchymal entry of immune cells and antibodies while allowing selective transport. TGF-β secreted by and dampens , and FasL expression on neuronal surfaces promotes deletion of infiltrates. Reduced lymphatic drainage traditionally contributed to isolation, but discoveries since 2015 reveal meningeal lymphatic vessels (mLVs) in the that drain (CSF) and antigens to , facilitating peripheral tolerance by enabling Treg-mediated suppression of autoreactive responses without compromising surveillance. In experimental autoimmune models, mLV ablation exacerbates , underscoring their role in clearing self-antigens to prevent . Testicular immune privilege protects post-meiotic germ cells, which express novel antigens, through the blood-testis barrier (BTB) established by tight junctions, physically segregating the adluminal compartment from immune surveillance. FasL on s and germ cells triggers of invading lymphocytes, while TGF-β isoforms from Leydig and s inhibit T cell activation and promote Treg expansion. expression in s and macrophages catabolizes tryptophan to starve effector T cells, and although lymphatics exist, their limited density minimizes . These mechanisms collectively prevent antisperm , with evolutionary conservation evident in their preservation across mammals to ensure reproductive fitness. During , the becomes immunoprivileged to tolerate the semi-allogeneic , with the placental layer acting as a barrier that lacks classical /II expression, evading T cell recognition. TGF-β and IL-10 produced by cells and regulatory B cells suppress NK and T cell cytotoxicity, while IDO in trophoblasts and uterine macrophages induces local T cell anergy and Treg accumulation. Reduced lymphatic vessels in the decidua limit immune trafficking, fostering peripheral tolerance essential for fetal survival; disruptions, such as IDO inhibition, lead to inflammatory cascades and . Tregs play a supportive role in amplifying these site-specific suppressions. The critical role of Tregs in peripheral tolerance, including in immunoprivileged sites, was recognized by the 2025 in or awarded to Mary Brunkow, Fred Ramsdell, and Shimon Sakaguchi for their discoveries on regulatory T cells and immune self-tolerance.

Intrinsic mechanisms in T cells

Ignorance and quiescence

In peripheral tolerance, refers to the state in which self-reactive T cells fail to encounter or respond to self-, remaining naive and functionally competent despite their presence in the body. This mechanism primarily arises from the physical sequestration of antigens in immunoprivileged sites, such as the , eye, and testes, where anatomical barriers like the blood- barrier or lack of lymphatic drainage limit antigen access to the and prevent effective presentation to T cells. Additionally, self- expressed at low levels or in a tissue-specific manner, such as insulin confined to pancreatic beta cells, contribute to ignorance by evading sufficient by antigen-presenting cells in secondary lymphoid organs. Evidence for ignorance is demonstrated in models using OT-I TCR transgenic mice, which recognize ovalbumin (OVA) presented in the context of H-2K^b. In mice expressing low levels of OVA in cells (RIP-OVA^lo), OT-I ^+ T cells recirculate without , proliferation, or functional impairment, indicating complete due to subthreshold ; in contrast, higher antigen doses lead to active mechanisms. This subimmunogenic presentation ensures that self-reactive T cells do not receive the signals necessary for , preserving their potential responsiveness to foreign antigens while avoiding . Quiescence complements by enforcing a metabolically dormant state in naive self-reactive T cells, keeping them arrested in the of the with minimal proliferation, low biosynthetic activity, and reliance on for energy needs. This dormancy is maintained by low-level tonic (TCR) signaling from interactions with self-peptide-MHC complexes, which sustains without triggering effector functions and sets a high threshold. Transcriptional regulators such as FOXO1, KLF2, and TGFβ1 further reinforce quiescence by repressing metabolic and pathways, ensuring T cells remain small, long-lived, and tolerant in the periphery. Unlike anergy, which involves hyporesponsiveness following partial TCR engagement, both ignorance and quiescence prevent any initial activation of self-reactive T cells, maintaining their naive phenotype without altering intrinsic signaling pathways.

Anergy

T cell anergy serves as an intrinsic mechanism of peripheral tolerance, rendering self-reactive T cells functionally unresponsive to antigenic stimulation while allowing their persistence in the periphery. This state is induced primarily when naive T cells encounter self-antigens via T cell receptor (TCR) engagement without sufficient costimulatory signals, such as those from CD28-B7 interactions, leading to a hyporesponsive phenotype that inhibits proliferation, cytokine production (particularly interleukin-2, or IL-2), and effector differentiation. The absence of costimulatory support is critical, as it blocks full activation and instead promotes transcriptional and signaling changes that enforce long-term tolerance. At the molecular level, partial TCR signaling (signal 1 without signal 2) results in incomplete activation of pathways like Ras-MAPK and calcineurin-NFAT, with upregulation of inhibitory molecules such as CTLA-4, which dampens further responses. This is accompanied by epigenetic modifications and altered activity, including reduced IL-2 promoter accessibility and increased expression of anergy-associated genes like (gene related to anergy in lymphocytes), which ubiquitinates key signaling proteins to limit ERK and JNK activation. Consequently, anergic T cells exhibit defective IL-2 production and responsiveness, maintaining unresponsiveness without immediate . In peripheral lymphoid tissues, anergic T cells often downregulate CD25 ( α chain) and may adopt a partially activated but non-proliferative state, with shortened lifespans compared to naive cells. This positioning limits their recruitment into immune responses, and while anergy can be partially reversible upon strong restimulation or IL-2 provision, chronic self-antigen exposure sustains the hyporesponsive state and prevents effector function. Seminal evidence for these processes derives from superantigen models, where injection of bacterial superantigens like staphylococcal enterotoxin B (SEB) induces anergy in specific Vβ TCR-expressing CD4+ T cells due to massive initial activation followed by deletion and hyporesponsiveness in survivors, as they fail to proliferate or produce IL-2 upon re-challenge. In TCR-transgenic models, such as DO11.10 mice specific for ovalbumin, presentation of antigen by non-professional antigen-presenting cells without costimulation results in anergic CD4+ T cells with impaired responses, distinguishing anergy from ignorance at higher antigen doses.

Deletion

Deletion represents a critical mechanism of peripheral tolerance wherein self-reactive T cells undergo to prevent . This process eliminates potentially harmful lymphocytes that escape central tolerance in the , ensuring immune homeostasis in secondary lymphoid organs and tissues. Unlike non-apoptotic forms of tolerance, deletion actively removes these cells through , primarily via extrinsic and intrinsic apoptotic pathways triggered by T cell receptor (TCR) engagement with self-antigens. The extrinsic pathway is mediated by the interaction between (CD95) and (FasL), members of the (TNF) family, which initiates caspase-dependent in activated T cells. Upon repeated TCR stimulation, FasL expression is upregulated on T cells, binding to Fas on neighboring cells and forming the death-inducing signaling complex () that recruits Fas-associated death domain () and procaspase-8, leading to caspase activation and . This pathway is particularly prominent in activation-induced cell death (AICD), where chronic exposure to low levels of self-antigen in the drives the deletion of overexpanded autoreactive clones. Complementing the extrinsic route, the intrinsic mitochondrial pathway involves an imbalance in proteins, notably the pro-apoptotic BH3-only protein Bim and the anti-apoptotic , following sustained TCR signaling. Repeated stimulation promotes Bim upregulation and translocation to mitochondria, where it antagonizes , releasing and activating the (caspase-9 and effector caspases), culminating in . This pathway operates in contexts of high-dose peripheral exposure, where strong TCR signals overwhelm survival cues and tip the balance toward . Bim and Fas pathways function concurrently, as evidenced by double-deficient mice exhibiting severe and rapid due to failed T cell . Regulatory signals further modulate deletion efficiency, with TNF family members like FasL providing pro-apoptotic cues, while inhibition of the PI3K/Akt pathway enhances susceptibility to both extrinsic and intrinsic . PI3K/Akt activation promotes T cell survival by phosphorylating pro-apoptotic targets and upregulating anti-apoptotic members; thus, its blockade sensitizes self-reactive T cells to deletion by reducing survival signals during encounter. In high-dose scenarios, this inhibition amplifies mitochondrial outer membrane permeabilization via Bim. Experimental evidence underscores deletion's role in preventing autoimmunity, as seen in the RIP-mOVA model where cross-presentation of islet-expressed ovalbumin deletes autoreactive + T cells in pancreatic lymph nodes, averting onset. Fas deficiency impairs this deletion, allowing autoreactive + T cell persistence and disease progression, while Bim-mediated intrinsic is essential for the process, as Bim-deficient T cells accumulate without causing overt due to impaired effector function. Defects in these pathways cause systemic ; for instance, lpr mice with a mutation exhibit lymphoproliferation and lupus-like disease due to failed peripheral T cell deletion and AICD. Similarly, leads to accumulation of autoreactive lymphocytes and multi-organ , highlighting the pathway's necessity.

Intrinsic mechanisms in B cells

Anergy

B cell anergy serves as an intrinsic mechanism of peripheral tolerance, rendering self-reactive B cells functionally unresponsive to antigenic stimulation while allowing their persistence in the periphery. This state is induced primarily in transitional B cells that encounter self-antigens in the spleen, where chronic engagement of the B cell receptor (BCR) occurs without T cell help, leading to downregulated surface BCR expression and impaired intracellular signaling. The absence of costimulatory signals from CD4+ T cells is critical, as it prevents full activation and instead promotes a hyporesponsive phenotype that silences autoreactive responses. At the molecular level, chronic BCR occupancy drives increased internalization of the immunoglobulin receptor, which sequesters it from lipid rafts and uncouples signaling from pathways like activation, thereby limiting and production. This is accompanied by altered adapter protein recruitment, including elevated involvement of in complexing with and Shc to activate inhibitory phosphatases such as SHIP-1, which further dampens BCR-mediated calcium flux and ERK activation. Consequently, anergic B cells exhibit constitutive low-level signaling that maintains unresponsiveness without triggering . In peripheral lymphoid tissues, anergic B cells translocate from B cell follicles to extrafollicular pools or the T-B border, adopting a distinct An1 phenotype with shortened lifespans of approximately 5 days. This positioning excludes them from germinal center entry, and while anergy is reversible upon antigen withdrawal—recovering responsiveness within 48 hours—prolonged self-antigen exposure sustains follicle exclusion and prevents participation in immune responses. Seminal evidence for these processes derives from the hen egg lysozyme (HEL) transgenic model, in which B cells expressing HEL-specific BCRs encounter soluble self-HEL , resulting in anergic cells with downregulated BCR levels that fail to secrete antibodies or proliferate upon re-challenge, thereby enforcing . In this system, receptor occupancy above 5-45% directly correlates with anergy induction, distinguishing it from at lower levels.

Deletion and ignorance

In peripheral B cell tolerance, deletion serves as a key mechanism to eliminate mature self-reactive that escape central tolerance, primarily through -mediated triggered by encounters with self-antigens. This process is activated when the (BCR) on mature binds self-antigens, leading to signaling that upregulates (CD95) expression and sensitizes cells to (FasL)-induced death, often delivered by surrounding immune cells. Unlike central tolerance in the , where immature are deleted at lower BCR affinities due to heightened sensitivity, peripheral deletion in mature requires higher BCR affinity thresholds and stronger signaling, reflecting adaptations in mature cell survival pathways that prioritize recirculation and responsiveness to foreign antigens. A critical regulator in this deletion is the downregulation of Blimp-1 (), a typically promoting differentiation and survival; in self-reactive mature , BCR-induced PI3K signaling transiently upregulates Blimp-1 to repress proliferation and anti-apoptotic genes like , facilitating without differentiation. Evidence from transgenic models highlights the efficiency of peripheral deletion for certain self-reactive clones. For instance, in anti-DNA models relevant to systemic (SLE), self-reactive B cells specific for double-stranded DNA that mature and enter the periphery undergo Fas-dependent apoptotic deletion, preventing production in non-autoimmune contexts; defects in this process, as seen in SLE-prone strains, allow survival and contribute to disease. Similarly, in rheumatoid factor-specific models, mature peripheral B cells binding self-IgG2a undergo deletion following abortive activation, underscoring the role of this mechanism in maintaining to ubiquitous serum antigens. Ignorance represents another peripheral tolerance strategy, where mature self-reactive B cells persist but fail to activate due to limited or ineffective encounters with self-antigens, particularly soluble forms in that do not provide sufficient BCR crosslinking. These B cells continue to recirculate through lymphoid tissues without productive signaling, maintaining a quiescent state and avoiding both and deletion, as the low-avidity interactions do not surpass thresholds. Compartmentalization contributes to this ignorance by sequestering certain self-antigens in tolerance-prone sites, such as the or splenic red pulp, where transitional and mature B cells encounter them transiently during recirculation but without co-stimulatory cues necessary for response. The transgenic mouse model exemplifies peripheral ignorance, where B cells expressing a BCR specific for hen egg lysozyme (HEL) remain unresponsive when HEL is expressed as a soluble serum neo-self-antigen; these cells recirculate normally but produce no anti-HEL antibodies upon , demonstrating functional ignorance due to inadequate . This mechanism complements anergy by addressing self-reactive B cells exposed to soluble antigens at concentrations too low for unresponsiveness but sufficient to evade lethal outcomes.

Specialized phenomena

Split tolerance

Split tolerance refers to a state of partial immunological in which the exhibits unresponsiveness in one arm—either cellular (T cell-mediated) or humoral (antibody-mediated)—while maintaining responsiveness in the other to the same . This phenomenon is particularly observed in contexts such as chronic infections and , where full immune activation does not occur, allowing antigen persistence without complete clearance or severe . Mechanisms underlying split tolerance often involve selective impairment of T cell function, such as deletion or anergy, which spares B cell activity and permits antibody production without effective cytotoxic responses. In chronic settings, prolonged antigen exposure drives T cell exhaustion, characterized by upregulated inhibitory receptors like PD-1, further contributing to this compartmentalized tolerance. A classic example is chronic infection with (LCMV) in mice, where persistently infected carriers develop T cell exhaustion and fail to mount cytotoxic responses, yet sustain production against the , leading to lifelong persistence without acute . Neonatal tolerance models also demonstrate split tolerance; for instance, injection of allogeneic cells into newborn mice induces T cell unresponsiveness to grafts while allowing humoral responses to certain epitopes. This partial tolerance has significant implications for transplant outcomes, explaining scenarios of long-term graft survival despite incomplete , as seen in orthotopic liver allografts in mice that induce donor-specific T tolerance without eliminating responses.

Therapeutic implications

Understanding peripheral has profound implications for treating autoimmune diseases, where aim to restore self-tolerance by expanding regulatory T cells (Tregs) or inducing T cell anergy. Low-dose interleukin-2 (IL-2) promotes selective Treg expansion, demonstrating efficacy in (RA) clinical trials; for instance, a 2022 randomized controlled trial showed improved clinical symptoms through restored Treg function in refractory RA patients, with sustained benefits observed up to 24 weeks. Similarly, a 2024 study combining low-dose IL-2 with reported safe symptom reduction in active RA, highlighting its role in modulating peripheral tolerance without broad . PD-1 , which enhance inhibitory signaling to induce T cell anergy, have advanced in autoimmunity; peresolimab, a PD-1 agonist , achieved a 45% response rate in a 2023 phase 2 trial for RA patients unresponsive to anti-TNF , supporting its potential to reinstate . In transplantation, blockade targets peripheral T cell deletion to prevent allograft rejection while preserving tolerance. , a CTLA-4-Ig inhibiting CD28-B7 interactions, induces peripheral deletion of alloreactive T cells and has been approved for ; phase 3 trials demonstrated superior long-term graft function compared to cyclosporine, with reduced chronic allograft nephropathy. A 2024 review underscores its role in promoting Treg function and tolerance, though challenges like belatacept-resistant rejection from memory T cells persist, prompting combination strategies. For , reversing peripheral tolerance to tumor antigens via checkpoint inhibitors mobilizes exhausted T cells, but this risks disrupting self-tolerance. Anti-PD-1 antibodies like block PD-1/ interactions, reinvigorating anti-tumor immunity; in and non-small cell , they yield objective response rates of 30-40%, transforming peripheral tolerance mechanisms into therapeutic targets. However, immune-related adverse events, such as autoimmune or in 10-20% of patients, arise from unintended breaking of self-tolerance, emphasizing the need for balanced modulation. Emerging therapies leverage peripheral tolerance for precision interventions. Chimeric antigen receptor (CAR)-Treg cells provide tissue-specific suppression; preclinical 2024 studies in models using glycoprotein-specific CAR-Tregs demonstrated targeted Treg recruitment and reduced , with phase 1 trials for and (T1D) initiated by 2025 showing feasibility and safety. Gene editing enhances tolerogenic dendritic cells (tolDCs) to promote antigen-specific ; CRISPR/Cas9 editing of DC genes like Ndrg2 in 2023 murine models boosted regenerative potential and , with 2024 reviews advocating its application in autoimmune therapies. In T1D prevention, (anti-CD3) trials as of 2025 delayed clinical onset by a median of 24 months in at-risk individuals, achieving 30-40% efficacy in preserving beta-cell function over 2 years. mRNA-based tolerogens induce peripheral via non-inflammatory delivery; a 2025 spleen-targeted mRNA-lipid encoding self-antigens expanded Tregs and suppressed in experimental models, addressing post-2020 gaps in antigen-specific therapies. Microbiome modulation restores gut-associated peripheral in ; short-chain fatty acids from influence Treg differentiation, with 2023 studies showing fecal microbiota transplantation reduced inflammation in by enhancing barrier integrity and .

References

  1. [1]
    Immunological mechanisms of tolerance: Central, peripheral ... - PMC
    Dec 11, 2023 · Peripheral tolerance mechanisms, such as exhaustion, deletion, anergy, and regulation are mainly relied upon to suppress the activity of self- ...
  2. [2]
    Mechanisms maintaining peripheral tolerance | Nature Immunology
    Dec 17, 2009 · In this review, I discuss data that reveal barriers preventing peripheral T cell recognition of self-peptide–MHC complexes, as well as the physiological ...
  3. [3]
    Peripheral tolerance - Latest research and news - Nature
    Peripheral tolerance describes the mechanisms that take place outside of primary lymphoid tissues to prevent lymphocytes from initiating potentially dangerous ...
  4. [4]
    T-Cell Tolerance: Central and Peripheral - PMC
    Peripheral tolerance processes exist wherein self-reactive T cells become functionally unresponsive (anergy) or are deleted after encountering self-antigens ...Missing: review | Show results with:review
  5. [5]
    Tolerance and autoimmunity - PMC - NIH
    A faulty central tolerance sows the seeds for autoimmune disease, and faulty peripheral tolerance leads to its eruption.
  6. [6]
    The curious case of the 1960 Nobel Prize to Burnet and Medawar
    Jan 20, 2016 · The 1960 Nobel Prize was awarded to Macfarlane Burnet and Peter Medawar for immunological tolerance. The Nobel Archives reveal that the two were never ...
  7. [7]
    Breakdown in Peripheral Tolerance in Type 1 Diabetes in Mice and ...
    Type 1 Diabetes (T1D) is an example of one such autoimmune disease wherein the breakdown in tolerance leads to the initiation and progressive destruction of the ...
  8. [8]
    Mechanisms of central tolerance for B cells - PMC - NIH
    Central tolerance refers to the regulatory mechanisms that occur at the early stages of B cell development in the bone marrow, when B cells carry a surface ...
  9. [9]
    Impact of Negative Selection on the T Cell Repertoire Reactive to a ...
    We found that as many as 25%–40% of T cells reactive to a self-peptide escaped clonal deletion in the thymus and in the periphery.
  10. [10]
    Cellular and molecular mechanisms breaking immune tolerance in ...
    Apr 1, 2021 · Incomplete induction of peripheral tolerance and autoimmunity in DOCK8 deficiency reflects the involvement of DOCK8 in Treg homeostasis and ...
  11. [11]
    Immune Tolerance vs. Immune Resistance: The Interaction Between ...
    Mar 29, 2022 · Immune tolerance is of two types, i.e., Central immune tolerance and Peripheral immune tolerance. These mechanisms involve the T regulatory ...
  12. [12]
    The development and function of regulatory T cells - PMC - NIH
    There are two types of Tregs: natural Tregs, which develop in the thymus, and induced Tregs, which are derived from naive CD4+ T cells in the periphery. Tregs ...Tcr Ligation, Strength Of... · Maintenance Of Ntreg... · Mechanisms Of Treg...
  13. [13]
    Generation and Function of Induced Regulatory T Cells - PMC
    There are two major subsets of Treg cells, “natural” Treg (nTreg) cells that develop in the thymus, and “induced” Treg (iTreg) cells that arise in the periphery ...
  14. [14]
    How regulatory T cells work - PMC
    Regulatory T (Treg) cells are essential for maintaining peripheral tolerance, preventing autoimmune diseases and limiting chronic inflammatory diseases.
  15. [15]
    Mechanisms of regulatory T-cell suppression - PMC - NIH
    Here we review recent advances in the identification of distinct mechanisms of Treg action and of signals that enable cellular targets to escape regulation.
  16. [16]
    IPEX Syndrome - GeneReviews® - NCBI Bookshelf
    Oct 19, 2004 · FOXP3 missense variants can result in FOXP3 expression resulting in normal regulatory T cell (Treg) enumeration by flow cytometry but abnormal ...
  17. [17]
    Regulatory T cells prevent transfer of type 1 diabetes in NOD mice ...
    Oct 1, 2008 · Regulatory T cells (Tregs) can potentially be used as tools to suppress pathogenic T cells in autoimmune diseases such as type 1 diabetes.
  18. [18]
    In Vitro–expanded Antigen-specific Regulatory T Cells Suppress ...
    Previous studies have shown that the number and function of Tregs in NOD mice decrease over time correlating with clinical disease onset between 16 and 24 wk of ...
  19. [19]
    Metabolic programming in dendritic cells tailors immune responses ...
    Aug 19, 2021 · DCs also bestow peripheral tolerance by continuously presenting self-antigens as immature DCs in the absence of pro-inflammatory factors, ...
  20. [20]
    Macrophages in immunoregulation and therapeutics - Nature
    May 22, 2023 · Macrophages are crucial in innate immunity by regulating several homeostatic and evolutionary host defense immune responses.
  21. [21]
    Lymph node–resident lymphatic endothelial cells mediate ...
    Mar 22, 2010 · Peripheral tolerance prevents self-reactive T cells that escape thymic negative selection from causing autoimmunity.
  22. [22]
    Lymph node stromal cells constrain immunity via MHC class II self ...
    Nov 19, 2014 · Altogether, our results reveal a novel mechanism by which lymph node stromal cells regulate peripheral immunity. https://doi.org/10.7554/eLife.
  23. [23]
    Lymph node stroma broaden the peripheral tolerance paradigm - PMC
    We now know that lymph node stromal cells (LNSC) are important mediators of deletional tolerance to peripheral tissue-restricted antigens (PTAs).
  24. [24]
    Role of lymph node stroma and microenvironment in T cell tolerance
    Sep 19, 2019 · This review will present our current understanding of LN SC subsets roles in regulating T cell tolerance. Three major types of LN SC subsets, ...<|control11|><|separator|>
  25. [25]
    Absence of MHC-II expression by lymph node stromal cells results in ...
    Dec 17, 2018 · Altogether, we prove that MHCII expression by LNSCs have a manifest impact in peripheral tolerance. Notably, LECs support self-Ag–specific T ...
  26. [26]
    Regulation of T-cell Tolerance by Lymphatic Endothelial Cells - PMC
    Here, we describe how lymphatic endothelial cells induce peripheral T-cell tolerance and how this relates to tolerance induced by other types of antigen ...
  27. [27]
    Cellular and Molecular Mechanisms of Anterior Chamber ...
    The ACAID mechanism is an immunomodulatory phenomenon induced following an injection of an antigen into the anterior chamber of an eye. The antigen (depicted ...
  28. [28]
  29. [29]
    Sympathetic Ophthalmia - StatPearls - NCBI Bookshelf - NIH
    The exact pathogenesis of sympathetic ophthalmia is unclear. The immune-privileged status of the eye occurs after a penetrating injury and exposure of ...
  30. [30]
    Meningeal Lymphatics: An Immune Gateway for the Central Nervous ...
    Dec 1, 2021 · The blood–brain barrier (BBB) restricts the access of cells or molecules coming from the periphery into the CNS parenchyma [16]. However ...
  31. [31]
  32. [32]
    Meningeal lymphatic drainage: novel insights into central nervous ...
    May 5, 2025 · In recent years, increasing evidence has suggested that meningeal lymphatic drainage plays a significant role in central nervous system (CNS) diseases.
  33. [33]
    Testicular defense systems: immune privilege and innate ... - PMC
    Jun 23, 2014 · Testicular immune privilege protects immunogenic germ cells from systemic immune attack, and local innate immunity is important in preventing testicular ...
  34. [34]
    Role of indoleamine 2,3-dioxygenase in testicular immune-privilege
    Nov 4, 2019 · ... immune rejection. Multiple mechanisms are involved in testis immune privilege: a blood testis barrier modulates antigen and antibody ...Ido/tdo Activity Assay · Results · Ido/tdo Activity In Testis
  35. [35]
    Tolerance of the fetus by the maternal immune system
    This review will cover recent aspects of immune privilege and the innate immune system at the feto-maternal interface, citing examples of the role played by ...
  36. [36]
  37. [37]
    Manifestations of immune tolerance in the human female ... - Frontiers
    Feb 12, 2013 · These observations indicate that the female reproductive tract is immune privileged during pregnancy. However, a sexually active woman's immune ...
  38. [38]
    Tolerance and Exhaustion: Defining Mechanisms of T cell Dysfunction
    We discuss distinct states of CD8 T cell dysfunction, with emphasis on (i) T cell tolerance to self-antigens (self-tolerance), (ii) T cell exhaustion during ...Missing: percentage | Show results with:percentage
  39. [39]
    CD8 T cell ignorance or tolerance to islet antigens depends on ...
    Abstract. There are two major mechanisms reported to prevent the autoreactivity of islet-specific CD8+ T cells: ignorance and tolerance.
  40. [40]
    Guardians of silence: transcriptional networks in T cell quiescence
    Aug 4, 2025 · In the quiescent state, cells exit from normal cell cycle and remain in a nonproliferative state in G0 phase. In addition to being devoid of ...
  41. [41]
    Rethinking peripheral T cell tolerance: checkpoints across a T cell's ...
    Oct 19, 2020 · ... T cell stage, where quiescence and ignorance enforce T cell tolerance. ... Similarly, ignorant self-reactive T cells exist in human peripheral ...
  42. [42]
  43. [43]
  44. [44]
  45. [45]
  46. [46]
    Dual Role of Fas/FasL-Mediated Signal in Peripheral Immune ... - PMC
    Thus, the impairment of Fas/FasL system in the peripheral immune tolerance considerably contributes to the onset or development of a lot of autoimmune diseases.
  47. [47]
    None
    Nothing is retrieved...<|separator|>
  48. [48]
    Life and Death of Activated T Cells: How Are They Different from ...
    Activated T cells are remarkably different in their regulation of apoptosis by pro- and antiapoptotic members of the BCL-2 family.
  49. [49]
    Apoptosis Regulators Bim and Fas Function Concurrently to Control ...
    Bim is a proapoptotic member of the Bcl-2 superfamily that functions in the intrinsic pathway of cell death (O'Connor et al., 1998). Fas plays a role in the ...
  50. [50]
    Loss of the Pro-Apoptotic BH3-only Bcl-2 Family Member Bim ...
    Here we report that B lymphocytes lacking the pro-apoptotic Bcl-2 family member Bim are refractory to apoptosis induced by BCR ligation in vitro. The loss of ...
  51. [51]
    The Role of the PI3K Signaling Pathway in CD4+ T Cell ...
    Tolerance is controlled by two major mechanisms: central tolerance in the thymus that results in deletion of the majority of self-reactive T cells; and ...
  52. [52]
    The role of the PI3K signaling pathway in CD4 + T cell differentiation ...
    Aug 12, 2012 · In this review, we discuss the role of the PI3K signaling pathway in CD4 + T cell differentiation and function, and focus on how modulation of this pathway in ...
  53. [53]
    Peripheral Deletion of Autoreactive CD8 T Cells by Cross ...
    Our model involves the adoptive transfer of OVA-specific CD8 T cells from the OT-I transgenic line (OT-I cells [21]) into RIP-mOVA mice, which express mOVA ...
  54. [54]
    The Peripheral Deletion of Autoreactive CD8 + T Cells Induced by ...
    In conclusion, we have demonstrated that CD95 plays an important role in the deletion of autoreactive CD8+ T cells induced by cross-presentation of self- ...
  55. [55]
    Requirement of Fas expression in B cells for tolerance induction
    Jan 2, 2002 · These results indicate that both mature B cells and T cells must undergo Fas-mediated apoptosis to prevent the development of autoimmune ...2 Results · 2.3 Reduced B Cell Expansion... · 3 Discussion
  56. [56]
    Regulation of Anti-DNA B Cells in Recombination-activating Gene ...
    This model allows us to determine whether anti-DNA B cells are deleted by programmed cell death, whether apoptotic cells present the immunogens that drive DNA- ...
  57. [57]
    Peripheral deletion of rheumatoid factor B cells after abortive ... - PNAS
    The deletion is independent of the Fas/Fas ligand (FasL) pathway of apoptosis and is preceded by a phase of partial activation involving increase in cell size ...
  58. [58]
    Interaction with self antigens selects some lymphocytes for survival ...
    Mature B cells recirculate through lymphoid follicles during their life-span. In normal circumstances, in which B cells binding soluble self antigen are in ...
  59. [59]
    B Cell Development in the Spleen Takes Place in Discrete Steps ...
    Only mature B lymphocytes can enter the lymphoid follicles of spleen and lymph nodes and thus efficiently participate in the immune response.Missing: compartmentalization | Show results with:compartmentalization
  60. [60]
    Immunological Tolerance - an overview | ScienceDirect Topics
    Split tolerance. Split tolerance includes several mechanisms. It is seen either when specific immunological unresponsiveness affects the B cell (antibody) ...
  61. [61]
    Lymphocytic Choriomeningitis Virus - an overview - ScienceDirect.com
    Persistently infected mice were eventually shown to have split-tolerance to LCMV, in that, although they could mount an antibody response to LCMV, they ...
  62. [62]
    Transplant Tolerance, Not Only Clonal Deletion - Frontiers
    Apr 20, 2022 · These animals have a “split tolerance” as peripheral lymphocytes from these animals respond to donor alloantigen in graft versus host assays and ...
  63. [63]
    T and B cell tolerance and responses to viral antigens in transgenic ...
    T and B cell tolerance and responses to viral antigens in transgenic mice: implications for the pathogenesis of autoimmune versus immunopathological disease.
  64. [64]
    studies on the mechanism of split tolerance in mice 1 - Transplantation
    Induction of split tolerance was prevented in neonatal hosts adoptively immunized to either donor strain allogeneic or MS antigens.
  65. [65]
    Split tolerance induced by orthotopic liver transplantation in mice
    Spontaneous orthotopic liver allograft acceptance associated with microchimerism in mice induces tolerance to subsequent skin or heart transplants from the ...
  66. [66]
    Split tolerance permits safe Ad5-GUCY2C-PADRE vaccine-induced ...
    Apr 23, 2019 · Conclusions Split tolerance to GUCY2C in cancer patients can be exploited to safely generate antigen-specific cytotoxic CD8+, but not autoimmune ...
  67. [67]
    Low-dose IL-2 improved clinical symptoms by restoring reduced ...
    Low-dose IL-2 improved clinical symptoms by restoring reduced regulatory T cells in patients with refractory rheumatoid arthritis: A randomized controlled trial.Missing: 2023-2025 | Show results with:2023-2025
  68. [68]
    The efficacy and safety of short-term and low-dose IL-2 combined ...
    Apr 21, 2024 · The results of this study showed that low-dose IL-2 combined with TCZ treatment could safely and effectively reduce arthritis symptoms in RA ...Abstract · Introduction · Results · Discussion
  69. [69]
    A Phase 2 Trial of Peresolimab for Adults with Rheumatoid Arthritis
    May 17, 2023 · Peresolimab showed efficacy in a phase 2a trial in patients with rheumatoid arthritis. These results provide evidence that stimulation of the PD-1 receptor has ...
  70. [70]
    Costimulation Blockade in Kidney Transplantation
    Belatacept has been demonstrated to be as efficient as cyclosporine-based immunosuppression and is associated with significantly better renal function.
  71. [71]
    Exploring Costimulatory Blockade-Based Immunologic Strategies in ...
    Mar 20, 2024 · This review paper aims to summarize current data on the immunologic role of costimulatory blockade in the field of transplantation.
  72. [72]
    Regulatory mechanisms of PD-1/PD-L1 in cancers - Molecular Cancer
    May 18, 2024 · Anti-PD-L1 treatment can reverse this phenotype and induce macrophage-mediated anti-tumor activity [110, 111].
  73. [73]
    Fundamental Mechanisms of Immune Checkpoint Blockade Therapy
    Immune checkpoint blockade removes inhibitory signals of T-cell activation, which enables tumor-reactive T cells to overcome regulatory mechanisms and mount an ...
  74. [74]
    MOG-specific CAR Tregs: a novel approach to treat multiple sclerosis
    Oct 20, 2024 · We developed a myelin oligodendrocyte glycoprotein (MOG)-specific CAR Treg cell therapy for patients with MS. MOG is expressed on the outer ...
  75. [75]
    Cas9-mediated knockout of Ndrg2 enhances the regenerative ...
    Aug 7, 2023 · We developed a CRISPR/Cas9 approach to precisely edit murine dendritic cells to enhance their therapeutic potential for healing chronic wounds.
  76. [76]
    The future of type 1 diabetes therapy - The Lancet
    Sep 19, 2025 · Teplizumab is approved by the US Food and Drug Admistration (FDA) for delaying the clinical onset of type 1 diabetes in individuals aged 8 years ...
  77. [77]
    A Spleen‐Targeted Tolerogenic mRNA‐LNPs Vaccine for the ...
    Feb 8, 2025 · Tolerogenic DCs can promote peripheral tolerance through enhancing the differentiation and expansion of regulatory T cells (Tregs), and/or ...Missing: tolerogens | Show results with:tolerogens
  78. [78]
    Complex regulatory effects of gut microbial short-chain fatty acids on ...
    Mar 1, 2023 · This review focuses on the preventive and immunomodulatory effects of SCFAs on autoimmunity. The tissue- and disease-specific effects of dietary fiber, SCFAs ...