Fact-checked by Grok 2 weeks ago

Clonal anergy

Clonal anergy is a central mechanism of whereby antigen-specific lymphocytes, including both T cells and B cells, enter a state of functional hyporesponsiveness or inactivation upon encountering their cognate without sufficient co-stimulatory signals, thereby preventing inappropriate immune activation and . In T cells, clonal anergy is induced primarily through (TCR) engagement (signal 1) in the absence of via (signal 2), leading to a long-term blockade of interleukin-2 (IL-2) production, proliferation, and effector functions. This process involves molecular alterations such as inhibition of the / kinase pathway, upregulation of diacylglycerol kinase alpha (DGKα) to dampen signaling, and of E3 ubiquitin ligases like Cbl-b and , which degrade key signaling molecules. Environmental factors, including nutrient deprivation and , further promote anergy by modulating pathways like and AMPK, distinguishing it from full activation. For B cells, clonal anergy arises in the periphery when self-reactive cells evade central deletion in the , resulting in cells that bind self-antigens but fail to produce antibodies or proliferate effectively upon re-stimulation. Key molecular features include elevated IgD/IgM ratios, enhanced activity of inhibitory molecules like Lyn kinase and SHIP-1 phosphatase, and altered BCR signaling that limits calcium flux and activation. Anergic B cells often recirculate through lymphoid tissues but can be recruited to germinal centers under certain conditions, where may edit their autoreactivity—a process termed clonal redemption. Overall, clonal anergy complements central tolerance mechanisms like clonal deletion, ensuring peripheral tolerance to self-antigens and maintaining immune homeostasis; its disruption is implicated in autoimmune diseases such as systemic lupus erythematosus, where anergic B cells may regain responsiveness. Recent studies have confirmed the presence of anergic B cells in human naïve B cell populations, supporting the relevance of these mechanisms in human immunity. Experimental models, including transgenic mice with defined antigen specificities, have been instrumental in elucidating these pathways since the concept's proposal in the early 1980s for B cells and late 1980s for T cells.

Fundamentals

Definition and Historical Context

Clonal anergy refers to a reversible state of functional inactivation in mature lymphocytes, either T cells or B cells, induced by recognition in the absence of adequate co-stimulatory signals, resulting in antigen-specific unresponsiveness that inhibits activation, proliferation, and effector functions while preserving cell viability. This mechanism contributes to by silencing potentially autoreactive clones without their elimination, allowing lymphocytes to persist in a hyporesponsive state. The concept of clonal anergy originated in studies of B-cell tolerance in the late 1970s and early 1980s, with Gustav Nossal and Beverley Pike demonstrating in 1980 that self- exposure in neonatal mice led to persistent, non-responsive B lymphocytes capable of binding but unable to proliferate or secrete antibodies upon restimulation. This finding built on earlier self-tolerance research from the 1970s, which explored how the avoids through mechanisms beyond central deletion, as proposed in Frank Macfarlane Burnet's . For T cells, Ronald Schwartz's laboratory extended the idea in the 1980s, showing through experiments that by chemically fixed splenocytes without induced long-term unresponsiveness in antigen-specific + T cells, preventing IL-2 production and proliferation. A pivotal 1987 study by Marc Jenkins and Schwartz formalized T-cell anergy as a distinct pathway, using adoptive transfer models to demonstrate that T-cell clones exposed to without became unresponsive , highlighting the role of signal strength in determining activation versus inactivation. This was complemented by models developed in 1990, where T-cell clones stimulated with immobilized anti-CD3 antibodies or phorbol ester/ionomycin in the absence of exogenous IL-2 entered a state of clonal anergy, failing to produce IL-2 or proliferate upon rechallenge but remaining viable and reversible by IL-2 addition. By the , the concept evolved through integration with broader self- studies, establishing anergy as a key peripheral mechanism alongside deletion and suppression, with seminal reviews synthesizing its implications for immune regulation.

Distinctions from Other Tolerance Mechanisms

Clonal anergy represents a passive form of peripheral , distinct from other mechanisms by its reliance on functional inactivation of mature lymphocytes without active suppression or cell elimination. Unlike clonal deletion, which irreversibly eliminates self-reactive T and B cells through primarily during central in the or , anergy induces a reversible hyporesponsive state in peripheral cells upon recognition without costimulatory signals. This reversibility allows anergic cells to potentially regain responsiveness if the inhibitory conditions are alleviated, such as through provision of interleukin-2, whereas deletion permanently removes the threat of . In contrast to regulatory T cell (Treg)-mediated suppression, which operates as an active, dominant mechanism involving release (e.g., IL-10 and TGF-β) to inhibit effector responses from other lymphocytes, anergy is intrinsically passive and cell-autonomous, requiring no intercellular communication for enforcement. Tregs, derived from both thymic and peripheral sources, exert ongoing suppressive effects that can be context-dependent and reversible but demand the presence of these specialized cells, whereas anergy affects individual clones directly upon suboptimal activation. Anergy also differs from immunological , another passive peripheral mechanism where self-reactive lymphocytes simply fail to encounter their due to or low , leading to no or inactivation. While ignorance preserves potentially reactive clones in a naive state without altering their function, anergy actively silences mature cells that have encountered , marking a checkpoint to prevent in the periphery despite prior . Conceptually, anergy functions as a safeguard within the two-signal model of , where signal 1 (antigen binding to the or ) alone triggers anergy, but full and require signal 2 (, e.g., via CD28-B7 interaction). This model, originally proposed to explain self-nonself , underscores anergy's role in averting inappropriate responses to self-s in mature peripheral cells, distinguishing it from central processes that occur before maturity.

Induction Mechanisms

In T Lymphocytes

Clonal anergy in T lymphocytes arises primarily from the engagement of the T cell receptor (TCR) with peptide antigen presented by major histocompatibility complex (MHC) molecules—class II on antigen-presenting cells for CD4+ T cells and class I for CD8+ T cells—in the absence of requisite co-stimulatory signals such as the CD28-B7 interaction. This "signal 1" without "signal 2" results in a hyporesponsive state, as first demonstrated in vitro using purified Ia molecules to present antigen to normal inducer T cell clones, leading to long-lived proliferative nonresponsiveness. While much of the experimental detail focuses on CD4+ T cells, similar induction principles apply to CD8+ T cells, where lack of co-stimulation during MHC I-peptide recognition enforces tolerance. Suboptimal antigen doses further contribute to this induction by delivering partial TCR stimulation insufficient to trigger full activation, thereby favoring anergy over proliferation or effector differentiation. Chronic exposure to low levels of antigen, as occurs in persistent self-antigen presentation, can similarly enforce anergy by maintaining T cells in a state of incomplete activation without escalating to inflammatory responses. Inhibitory signals amplify this process; for example, engagement of CTLA-4 on T cells with B7 ligands actively promotes anergy by outcompeting for binding and dampening co-stimulatory effects during antigen encounter. In vivo, these induction conditions manifest in peripheral tissues where self-s are encountered without or professional maturation. Anergic T cells thus serve as a safeguard against during incidental recognition of tissue-restricted self-antigens. Prominent examples come from transgenic mouse models, such as those expressing (HA) under the insulin promoter in (), where HA-specific TCR-transgenic CD4+ T cells (e.g., from 6.5 TCR mice) infiltrate the but become anergic, failing to trigger despite persistence. In these contexts, anergy prevents destructive immune responses to islet autoantigens, highlighting its role in maintaining without central deletion. Upon re-challenge with , anergic T cells remain viable and retain normal TCR expression but exhibit profound defects, most notably a failure to produce interleukin-2 (IL-2), which is essential for autocrine and clonal expansion. This IL-2 deficiency stems from impaired transcriptional activation rather than cell death, ensuring long-term suppression of responsiveness. Partial anergy states also occur, particularly under graded stimulation, where T cells display reduced but partially retain of other cytokines like interferon-gamma, allowing nuanced control of immune potential without complete inactivation. These outcomes underscore anergy's adaptability as a mechanism, balancing vigilance against pathogens with restraint toward .

In B Lymphocytes

Clonal anergy in B lymphocytes is primarily induced when the (BCR) binds to soluble self-antigens in the absence of T cell help or co-stimulatory signals. This encounter occurs in mature B cells that have exited the and entered peripheral circulation, where circulating self-molecules engage the BCR without the multivalent cross-linking provided by membrane-bound antigens on cell surfaces. In contrast, multivalent membrane-bound self-antigens typically drive B cell activation or deletion rather than anergy, highlighting the role of in determining tolerance outcomes. In vivo, anergy silences B cells reactive to self-antigens such as DNA in non-autoimmune mouse models, preventing aberrant responses to ubiquitous circulating self-molecules. For instance, in transgenic mice expressing anti-double-stranded DNA BCRs (VH3H9 × Vλ2), anergic B cells persist in reduced numbers in the periphery without producing detectable anti-DNA antibodies, demonstrating effective silencing of potentially autoreactive clones. Similarly, models with anti-single-stranded DNA specificity (VH3H9 × Vκ8) show normal B cell development but maintain anergy to avoid autoimmunity. These examples underscore anergy's role in peripheral tolerance to soluble autoantigens like nucleic acids, which are normally present in blood without triggering immunity. Upon induction, anergic B cells exhibit distinct cellular outcomes, including downregulation of surface BCR expression, particularly IgM, while maintaining or upregulating IgD. These cells recirculate from follicles to the margins of the or the T-B cell boundary, positioning them away from optimal activation environments. Functionally, they display reduced calcium flux upon BCR stimulation—often with elevated basal levels but no further mobilization—and impaired in response to , rendering them unresponsive to self-challenge. Notably, this anergic state is reversible; upon clearance of the inducing , such as in adoptive experiments to antigen-free hosts, B cells recover responsiveness within approximately 48 hours, allowing potential reactivation under non-self conditions.

Molecular Basis

Signaling Pathways Leading to Anergy

In T lymphocytes, clonal anergy is primarily induced through (TCR) engagement in the absence of costimulatory signals, such as those provided by . This incomplete activation triggers a partial signaling cascade where TCR ligation activates phospholipase Cγ (PLCγ), leading to increased intracellular calcium levels and subsequent activation of . Calcineurin then dephosphorylates nuclear factor of activated T cells (NFAT), promoting its translocation to the . However, without CD28 , the Ras-extracellular signal-regulated kinase (ERK)-activator protein 1 (AP-1) pathway remains inadequately activated, resulting in NFAT functioning without sufficient co-activators like AP-1. This imbalance represses interleukin-2 (IL-2) promoter activity, as NFAT alone cannot effectively drive IL-2 transcription, committing the T cell to an anergic state. In B lymphocytes, anergy arises from B cell receptor (BCR) stimulation by self-antigen without sufficient co-signals from CD40 or Toll-like receptors (TLRs). This elicits a weak PLCγ-mediated response, producing inositol 1,4,5-trisphosphate (IP3) and a subdued calcium release from intracellular stores, insufficient for robust nuclear factor of activated T cells (NFAT) or nuclear factor-κB (NF-κB) activation. Concurrently, the phosphatase and tensin homolog (PTEN) is upregulated, which dephosphorylates phosphatidylinositol (3,4,5)-trisphosphate (PIP3), thereby dampening the phosphoinositide 3-kinase (PI3K)-Akt pathway and preventing pro-survival and proliferative signals. This attenuated signaling maintains the B cell in a hyporesponsive state, with reduced calcium mobilization upon rechallenge. Recent studies indicate that chronic BCR signaling can drive metabolic and epigenetic changes leading to differentiation of anergic B cells into age-associated B cells (ABCs) in aged or autoimmune contexts. Across both T and B lymphocytes, common signaling features contribute to anergy induction, including the upregulation of inhibitory receptors such as (PD-1). PD-1 engagement further suppresses TCR or BCR proximal signaling by recruiting phosphatases like SHP-2, inhibiting downstream effectors. Additionally, diacylglycerol (DAG), a second messenger generated by PLCγ, accumulates but is metabolized by diacylglycerol kinases (DGKs, e.g., DGKα and DGKζ), preventing sustained Ras-MAPK activation and reinforcing the calcium-NFAT bias over proliferative pathways. These elements ensure long-term hyporesponsiveness following initial incomplete activation.

Epigenetic and Gene Expression Changes

In T cell anergy, transcriptional shifts arise from imbalanced signaling that sustains nuclear factor of activated T cells (NFAT) activity while impairing activator protein-1 (AP-1) components such as c-Fos and , leading to the upregulation of inhibitory transcription factors like early growth response 2 and 3 (Egr-2/3) and . These factors repress genes essential for T cell activation, including interleukin-2 (IL-2). Additionally, the E3 ubiquitin ligase , induced by NFAT dimers, targets key activation proteins such as the TCR zeta chain for proteasomal degradation, further enforcing hyporesponsiveness. Epigenetic modifications stabilize these transcriptional changes in anergic T cells. Recruitment of histone deacetylases (HDACs) by to the IL-2 promoter induces hypoacetylation of s H3 and H4, reducing accessibility and silencing IL-2 expression; this hypoacetylation persists even after restimulation and can be reversed by HDAC inhibitors like . also contributes, with hypermethylation at promoters of activation-associated genes such as those encoding cytokines and proliferation factors, as identified in genome-wide analyses of anergized T cells, thereby locking in the refractory state. In anergy, analogous epigenetic and transcriptional alterations occur, though specific mechanisms are less well-characterized. Long-term maintenance of anergy involves microRNAs (miRNAs) that fine-tune inflammatory pathways; for instance, miR-146a is upregulated in anergic T cells, where it targets TRAF6 and IRAK1 to suppress activation, limiting pro-inflammatory and promoting resolution of responses. The anergic state exhibits partial reversibility, particularly upon withdrawal of tolerogenic signals, through metabolic shifts that enhance TET-mediated at key loci like the Foxp3 conserved non-coding sequence 2 (CNS2), allowing potential transition to regulatory phenotypes.

Role in Immune Tolerance

Contribution to Peripheral Tolerance

Clonal anergy serves as a critical mechanism by rendering mature lymphocytes unresponsive upon encountering self-antigens in peripheral tissues, thereby preventing autoimmune responses against tissue-specific antigens that evade central deletion. This process is particularly relevant for antigens sequestered in non-lymphoid organs, where antigen-presenting cells lacking sufficient costimulatory signals induce a state of hyporesponsiveness in self-reactive T cells, inhibiting their and production without eliminating the cells. For instance, in models of peripheral self-antigen exposure, anergic T cells fail to initiate effector functions, safeguarding against tissue damage. Anergy complements central by addressing autoreactive clones that escape thymic deletion, dominating in scenarios where deletion is inefficient or undesirable. In to inhaled antigens, conventional dendritic s in nasal-associated lymphoid tissues enforce T unresponsiveness—characterized by hyporesponsiveness akin to anergy—through contact-independent suppression, preventing overactivation to harmless environmental antigens. Similarly, in the gut mucosa, high-dose exposure to microbiota-derived epitopes from commensals like induces anergy in + T s, promoting to microbial antigens and reducing without relying on clonal deletion. These models illustrate anergy's role in maintaining at mucosal barriers, where it prevails over deletion for persistent challenges. From an evolutionary perspective, anergy provides the with flexibility to respond to novel pathogens while avoiding self-attack, as anergic cells persist in a quiescent rather than being permanently removed. Quantitative models of T cell indicate that 25-40% of potentially self-reactive clones escape central selection and are subsequently managed peripherally, often through anergy thresholds that balance reactivity and . This mechanism ensures adaptive immunity's adaptability without compromising self-. Molecular maintenance of anergy, via sustained signaling alterations, enables long-term peripheral persistence of these silenced clones.

Relation to Dominant Tolerance

Dominant tolerance refers to active suppression of immune responses mediated by regulatory T cells (Tregs), particularly + + Tregs, which inhibit effector T cell activation through mechanisms such as secretion (e.g., IL-10, TGF-β) and direct cell-cell contact, thereby preventing and allograft rejection in a non-cell-intrinsic manner. In contrast, clonal anergy represents a cell-intrinsic form of where T cells become hyporesponsive due to incomplete activation signals, such as TCR engagement without sufficient , leading to impaired and production without active suppression of other cells. This distinction positions anergy as a passive, non-dominant mechanism, while Tregs enforce dominant control by broadly dampening immune reactivity. The interplay between anergy and dominant tolerance often involves sequential or concurrent actions, where anergy can precede Treg induction by limiting initial effector responses, or both mechanisms may coexist to establish robust peripheral tolerance. Shared molecular regulators, such as E3 ubiquitin ligases like Cbl-b and Itch, link the two processes; for instance, Cbl-b promotes anergy by inhibiting TCR signaling and also supports iTreg suppressive function, while Itch enhances Foxp3 expression in Tregs. In transplantation settings, this interplay is evident in models where anti-CD3 antibody therapy induces tolerance to skin allografts in mice by initially expanding Foxp3+ Tregs to suppress acute rejection, followed by the establishment of CD4+ T cell anergy marked by PD-1 and CD73hiFR4hi expression. Notably, anergy sustains long-term graft acceptance independently of Tregs, as tolerance persists even after Treg depletion via PC61 antibody or diphtheria toxin at day 100 post-transplant, demonstrating anergy's standalone role in preventing rejection without requiring ongoing dominant suppression. Recent studies from the 2010s onward have revealed that anergic T cells can convert into Foxp3+ Tregs under specific conditions, such as exposure to IL-2, which reverses anergy by activating mTOR and promoting aerobic glycolysis, leading to epigenetic remodeling at the Foxp3 conserved non-coding sequence 2 (CNS2) via TET-mediated demethylation. In Treg-deficient hosts, up to 20% of anergic CD4+ T cells (identified as Foxp3– FR4+ CD73+ Nrp1+) differentiate into functional Tregs, acquiring a fully demethylated CNS2 signature akin to natural Tregs and preventing autoimmunity. This convertibility challenges the 1990s perspective of strict separation between anergy and Treg-mediated tolerance, highlighting anergy as a potential precursor state that integrates into dominant mechanisms upon environmental cues like IL-2 signaling.

Applications and Study

Clinical Significance

Defects in clonal anergy mechanisms contribute significantly to the pathogenesis of autoimmune diseases, where failure to induce or maintain anergic states in self-reactive lymphocytes leads to unchecked immune responses against host tissues. In , loss of anergic B cells has been observed in prediabetic and new-onset patients, suggesting that disruption of B-cell anergy by environmental factors like predisposes individuals to . Similarly, in (RA), synovial T cells exhibit altered associated with anergy, contributing to hyporesponsiveness and paradoxical proliferation that drives joint inflammation. The non-obese diabetic (NOD) mouse model exemplifies these defects, as age-related loss of T-cell anergy correlates with spontaneous development of autoimmune , highlighting anergy's role in breakdown. In transplantation medicine, inducing clonal anergy offers a strategy to prevent graft rejection by blocking T-cell costimulatory signals, thereby promoting tolerance without broad immunosuppression. Belatacept, a CTLA-4 Ig fusion protein, inhibits CD28-mediated costimulation to induce T-cell anergy and has been evaluated in clinical trials for solid organ transplantation. Post-2015 trials include an ongoing phase 2 study in heart transplant recipients (NCT06478017, recruiting as of 2025), which assesses belatacept's potential to reduce immunosuppression needs. A pilot randomized controlled trial in lung transplantation (NCT03388008, completed 2022; published 2024) found no significant difference in acute rejection rates but reported increased mortality in the belatacept arm (5 vs. 0 deaths), highlighting safety challenges including early rejection and infectious risks. Clonal anergy also plays a dual role in , where tumor-induced anergy suppresses anti-tumor T-cell responses, but targeted reversal can enhance efficacy. Blocking the pathway reinvigorates anergic or exhausted tumor-infiltrating T cells, improving production and tumor control, as evidenced by anti-PD-1 therapies like nivolumab in and non-small cell . In chronic infections such as , PD-1 blockade similarly breaks T-cell anergy, restoring HIV-specific + T-cell proliferation and effector functions to better control viral loads. Emerging therapeutic approaches leverage gene editing to modulate anergy for immune desensitization in allergies. Advances from 2023-2025 using / have targeted genes in immune cells, including dendritic cells, to promote allergen-specific tolerance by reducing Th2 responses, as shown in preclinical models of airway allergies like and food allergies such as .

Experimental Detection and Models

Experimental detection of clonal anergy in T lymphocytes primarily relies on assays that assess functional unresponsiveness following initial stimulation. A key method involves re-stimulating purified CD4+ T cells with and measuring interleukin-2 (IL-2) production via enzyme-linked immunosorbent assay () or intracellular staining, where anergic cells exhibit markedly reduced IL-2 secretion compared to responsive controls. Proliferation assays, such as those using thymidine incorporation or (CFSE) dilution, further confirm anergy by demonstrating impaired cell division upon re-challenge, even in the presence of costimulatory signals. These assays were pivotal in early characterizations, showing that anergic T cells fail to proliferate or produce IL-2 despite TCR engagement. Flow cytometry provides a complementary approach for identifying anergic T cells through surface marker expression. often display low levels of CD25 ( α chain) and elevated expression of inhibitory receptors like PD-1, alongside other markers such as Egr-2 and , which can be detected via multicolor staining panels. Functional tests, including the delayed-type (DTH) assay, evaluate anergy by injecting intradermally and measuring suppressed skin induration in tolerant animals or adoptively transferred cells, indicating systemic unresponsiveness. The trans-vivo DTH variant enhances specificity by transferring splenocytes into lymphopenic hosts before challenge. Model systems for studying T cell anergy extensively utilize transgenic mice, with the DO11.10 strain being a cornerstone due to its expression of a TCR specific for ovalbumin (OVA) peptide presented on I-A^d. In DO11.10 mice, anergy is induced by administering soluble OVA without costimulation, leading to hyporesponsive KJ1-26+ CD4+ T cells that fail to expand or produce cytokines upon re-stimulation; this model has facilitated mechanistic studies of tolerance induction. In vivo tolerance models employing superantigens, such as staphylococcal enterotoxin B (SEB), target Vβ8+ T cells and induce anergy through repeated exposure, resulting in reduced proliferation and IL-2 production in responsive subsets, mimicking peripheral tolerance scenarios. For B lymphocytes, detection methods mirror T cell approaches but emphasize BCR signaling defects. In vitro assays measure calcium flux or proliferation in response to antigen re-stimulation, with anergic B cells showing blunted responses; flow cytometry identifies them via markers like transitional (IgM^low IgD^+ CD21^low) phenotypes or elevated PD-1 expression. Recent advances in single-cell RNA sequencing (scRNA-seq) have profiled anergic lymphocyte transcriptomes, revealing signatures such as upregulated GRAIL and DGKα in T cells or Itm2a in B cells, enabling high-resolution identification of heterogeneous anergic populations without prior sorting. B cell anergy models have evolved from transgenic systems like the 3-83μ , which expresses anti-MHC class I BCRs, to more physiologically relevant setups. In the 2020s, human immune organoids—3D cultures of s embedded in synthetic hydrogels with stromal support—have emerged to study B cell maturation and responses in germinal center-like environments, including tolerance testing with self-antigens in healthy donors and patients with . These organoids support long-term B cell and bridge gaps in murine models for human-relevant studies.

References

  1. [1]
    Induction of T cell anergy: integration of environmental cues and ...
    Anergy is a state of long term hyporesponsiveness in T cells that is characterized by an active repression of TCR signaling and IL-2 expression.
  2. [2]
    Anergic B Cells: Precarious On-Call Warriors at the Nexus ... - Frontiers
    Anergy is a poorly understood state whereby cells retain the ability to bind to self-antigens but are otherwise rendered insensitive to antigenic stimulation.
  3. [3]
    Clonal redemption and clonal anergy as mechanisms to balance B ...
    Sep 26, 2019 · Here we review this topic and recent advances showing that anergic B cells are recruited into the germinal center to mutate away from self-reactivity.
  4. [4]
    Immunological mechanisms of tolerance: Central, peripheral and the ...
    Dec 11, 2023 · Clonal anergy is a state in which T cells do not proliferate but still produce a limited amount of effector cytokines.
  5. [5]
  6. [6]
  7. [7]
    2. Central & Peripheral Tolerance - Immunopaedia
    Mechanisms of peripehral T-cell tolerance include: A. Clonal deletion; B. Ignorance; C. Anergy; D. Immune regulation. Tolerance mechanisms can also result in ...
  8. [8]
    The history of the two‐signal model of lymphocyte activation
    The two-signal model describes how antigen inactivates and activates lymphocytes, requiring a second signal for activation, and has three phases of development.
  9. [9]
    Stimulation of Normal Inducer T Cell Clones With Antigen ... - PubMed
    1987 Jun 1;138(11):3704-12. Authors. H Quill, R H Schwartz. PMID: 3035012. Abstract. Culture of normal inducer T cell clones with antigen and purified Ek beta ...Missing: anergy | Show results with:anergy
  10. [10]
    T CELL ANERGY ∗ Ronald H. Schwartz - Annual Reviews
    Dec 5, 2002 · Consistent with this argument, clonal anergy has been induced as an additional state in Th-2 cells by stimulating them with altered peptide ...
  11. [11]
    Induction of Peripheral T Cell Tolerance In Vivo Requires CTLA-4 ...
    We show that the induction of T cell anergy in vivo is due to an abortive T cell response that requires recognition of B7 molecules.Results · Tolerogenic Antigen Induces... · In Vitro Cultures And Assays...<|control11|><|separator|>
  12. [12]
    Peripheral tolerance to an islet cell-specific hemagglutinin ... - PubMed
    Peripheral tolerance to an islet cell-specific hemagglutinin transgene affects both CD4+ and CD8+ T cells ... Eur J Immunol. 1992 Apr;22(4):1013-22. doi: 10.1002/ ...Missing: et al self-
  13. [13]
    CD4+ T Cell Tolerance to Parenchymal Self-Antigens Requires ...
    T cell tolerance to parenchymal self-antigens is thought to be induced by encounter of the T cell with its cognate peptide–major histocompatibility complex.
  14. [14]
    Antigen-driven CD4 + T-cell anergy: a pathway to peripheral T ...
    Dec 4, 2020 · T-cell anergy is defined to be a state of long-lasting, partial or total unresponsiveness induced by incomplete CD4+ T-cell activation.
  15. [15]
    Breakdown of self-tolerance in anergic B lymphocytes - Nature
    ### Summary of Key Findings on B Cell Anergy
  16. [16]
    B-cell anergy: from transgenic models to naturally occurring anergic ...
    Anergy, a condition in which cells persist in the periphery but are unresponsive to antigen, is responsible for silencing many self-reactive B cells.Box 1 | Anergy In Humans · Immunoglobulin-Transgenic... · Bcr Signalling In B-Cell...
  17. [17]
    Anergic self-reactive B cells present self antigen and ... - PNAS
    B-cell tolerance to soluble protein self antigens such as hen egg lysozyme (HEL) is mediated by clonal anergy. Anergic B cells fail to mount antibody ...Missing: lymphocytes | Show results with:lymphocytes
  18. [18]
  19. [19]
  20. [20]
    Elevated PTEN expression maintains anergy in human B cells and ...
    Feb 7, 2019 · It has been reported that 2.5%–30% of human peripheral CD27– B cells are autoreactive and anergic based on unresponsiveness to antigen ...Missing: PLCγ IP3 CD40 TLR seminal
  21. [21]
    Regulatory Kinases and Phosphatases Maintaining B Cell Anergy
    Apr 5, 2018 · Below, we drill more deeply into proximal BCR signaling pathways and negative regulatory mechanisms that limit the antigen responsiveness of ...Missing: IP3 TLR
  22. [22]
    Inhibitory Receptors and Pathways of Lymphocytes: The Role of PD ...
    Defects involving PD-1 and Tregs can lead to the development of pathological conditions, including autoimmune disorders or promote cancer progression.
  23. [23]
    Transcriptional complexes formed by NFAT dimers regulate the ...
    Mar 23, 2009 · It has recently been shown that anergic T cells up-regulate transcription of Cblb by inducing the expression of the transcription factors Egr2 ...Disruption Of Nfat Homodimer... · Nfat Dimers Activate The... · Nfat1 Induces Grail...
  24. [24]
    Interleukin 2 gene transcription is regulated by Ikaros-induced ...
    Dec 5, 2006 · We present evidence that anergizing stimuli induce changes in histone acetylation, which mediates transcriptional repression of interleukin 2 expression.Missing: hypoacetylation | Show results with:hypoacetylation
  25. [25]
    Silencing of the Il2 gene transcription is regulated by epigenetic ...
    Jun 11, 2012 · In anergic T cells, the transcription factor Ikaros binds to the Il2 promoter and recruits histone deacetylases (HDAC), inducing changes in the ...
  26. [26]
    Comprehensive analysis of epigenetically regulated genes in ...
    We therefore comprehensively investigated the epigenetic regulation of those genes with altered expression in anergic T cells to fully understand the anergy- ...Missing: modifications seminal
  27. [27]
    miR-146a controls the resolution of T cell responses in mice - PMC
    By suppressing expression of TRAF6 and IRAK1, miR-146a regulates NF-κB activation in T cells through a negative feedback loop and controls the resolution of ...
  28. [28]
    Anergy into T regulatory cells: an integration of... - F1000Research
    Dec 17, 2018 · Anergy into T regulatory cells: an integration of metabolic cues and epigenetic changes at the Foxp3 conserved non-coding sequence 2.
  29. [29]
    The contributions of T-cell anergy to peripheral T-cell tolerance - NIH
    Clonal anergy: persistence in tolerant mice of antigen-binding B lymphocytes incapable of responding to antigen or mitogen. Proc Natl Acad Sci USA. 1980;77 ...
  30. [30]
    Unresponsiveness to inhaled antigen is governed by conventional ...
    DCs with tolerogenic properties have been identified in the intestinal mucosa, where they play a central role in enforcing tolerance to commensal bacteria and ...Missing: microbiota | Show results with:microbiota
  31. [31]
    Self and microbiota-derived epitopes induce CD4 + T cell anergy ...
    Nov 2, 2020 · We show that microbial antigens from Akkermansia muciniphila commensal bacteria can induce anergy and drive conversion of naive CD4 + CD44 - Foxp3 − T (Tn) ...Missing: quantitative | Show results with:quantitative
  32. [32]
    Intestinal mucosal tolerance and impact of gut microbiota to mucosal ...
    In exposure to a single high dose of antigen, clonal deletion or anergy are preferential tolerogenic mechanisms whereas numerous low antigen doses induce T cell ...Missing: inhaled | Show results with:inhaled
  33. [33]
    Impact of Negative Selection on the T Cell Repertoire Reactive to a ...
    As many as 25%–40% of reactive T cells escaped clonal deletion. A correlation was found between T cell avidity, TCRα structures, and susceptibility to negative ...<|separator|>
  34. [34]
    Mechanisms That Regulate Peripheral Immune Responses to ...
    It was originally thought that clonal anergy and regulatory T cells were entirely distinct mechanisms that are used to control peripheral immune responses.
  35. [35]
    Anergy into T regulatory cells: an integration of metabolic cues and ...
    Dec 17, 2018 · Anergy in the normal polyclonal CD4 T-cell repertoire leads to a state of functional unresponsiveness characterized by a block in autocrine ...
  36. [36]
    The Induction and Maintenance of Transplant Tolerance Engages ...
    Mar 6, 2017 · We show that both Foxp3+ Tregs and CD4+ T cell anergy play a role in the induction of tolerance and its maintenance. Foxp3+ Tregs resisted CD3 ...
  37. [37]
  38. [38]
    Ndrg1 is a T-cell clonal anergy factor negatively regulated by CD28 ...
    Oct 28, 2015 · T cell clonal anergy is defined as a hyporesponsive state of T cells following T-cell receptor (TCR) engagement in the absence of costimulation.
  39. [39]
    Immature dendritic cells convert anergic nonregulatory T cells into ...
    Nov 8, 2014 · Although a variety of experimental approaches have been used to induce T-cell anergy in vitro, the most consistent hallmarks are defective IL-2 ...Ctla-4 Blockade Allows... · Ctla-4 Interaction Is... · Cd28 Stimulation Controls...
  40. [40]
    Conversion of Anergic T Cells Into Foxp3- IL-10+ ... - Frontiers
    We describe the induction of anergic CD4 + T cells in vivo by intravenous application of high doses of antigen and their subsequent conversion into suppressive ...Abstract · Introduction · Results · DiscussionMissing: seminal papers
  41. [41]
    Trans-vivo delayed type hypersensitivity assay for antigen ... - PubMed
    May 2, 2013 · The lack of a delayed-type hypersensitivity response to a recall Ag demonstrated by skin testing is often regarded as an evidence of anergy.
  42. [42]
    Induction of antigen-specific CD4+ T-cell anergy and deletion by in ...
    DO11.10 transgenic mice represent one of the best-characterized immunologic model systems for studies on CD4+ T-cell differentiation and tolerance induction.
  43. [43]
    T cells with anergic and suppressive genetic signatures
    In this study, we sought to characterize the suppressive qualities of SEB-exposed CD4 + T cells and correlate this with genetic signatures of anergy and ...
  44. [44]
    Human naïve B cells show evidence of anergy and clonal ... - Nature
    May 14, 2025 · Clonal redemption and clonal anergy as mechanisms to balance B cell tolerance and immunity. ... The Quarterly Review of Biology 64, (1989).
  45. [45]
    Comprehensive single-cell analysis reveals novel anergic antigen ...
    Sep 14, 2023 · A few studies have emerged utilizing single-cell RNA sequencing (scRNA-seq) to analyze gene expression at the single-cell resolution in ...
  46. [46]
    Identification of an anergic B ND cell–derived activated B cell ...
    May 15, 2023 · We combined single-cell RNA sequencing (scRNA-seq) with CITE-seq ... Normalizing and denoising protein expression data from droplet-based single ...
  47. [47]
    Human immune organoids to decode B cell response in healthy ...
    Nov 6, 2024 · Current ex vivo models fail to sustain long-term GC reactions and effectively test B cell responses. Here we developed synthetic hydrogels ...