Fact-checked by Grok 2 weeks ago

T helper cell

T helper cells, also known as CD4⁺ T cells, are a subset of T lymphocytes that serve as key orchestrators of the adaptive , providing essential help to other immune cells such as B cells, cytotoxic T cells, and macrophages to mount effective defenses against pathogens. These cells are considered crucial for nearly all adaptive immune functions, as they recognize antigens presented by (MHC) class II molecules on antigen-presenting cells, leading to their and . Upon , naive T helper cells differentiate into specialized effector subsets, enabling tailored responses to diverse threats like intracellular pathogens, extracellular bacteria, and parasites. The differentiation of T helper cells is driven by signals from the microenvironment, including cytokines and interactions, resulting in distinct subsets such as Th1, Th2, Th17, and T follicular helper (Tfh) cells. Th1 cells primarily produce interferon-gamma (IFN-γ) to activate macrophages and promote against intracellular microbes, while Th2 cells secrete like IL-4, IL-5, and IL-13 to support antibody production and combat helminth infections. Th17 cells, characterized by IL-17 production, are vital for defense against extracellular bacteria and fungi at mucosal surfaces but can contribute to diseases when dysregulated. Additionally, regulatory T cells (Tregs), a suppressive subset, maintain immune by inhibiting excessive responses and preventing ; their discovery was recognized by the 2025 Nobel Prize in Physiology or awarded to Mary E. Brunkow, Fred Ramsdell, and Shimon Sakaguchi. Beyond infection control, T helper cells play pivotal roles in immune formation, responses, and pathological conditions; for instance, they are indispensable for generating long-lived CD8⁺ T cells and high-affinity antibodies. Their dysfunction, as seen in infection where CD4⁺ counts decline dramatically, underscores their importance, leading to broad . Ongoing research highlights the plasticity of these subsets, allowing adaptation to complex immune challenges.

Definition and overview

Discovery and nomenclature

The discovery of T helper cells traces back to the early 1960s, when Jacques Miller identified the as a critical organ for immune function through experiments in mice, demonstrating that thymus-derived lymphocytes (later termed T cells) were essential for cellular and . In 1966, Henry Claman and colleagues revealed the cooperative role of thymus-derived cells and bone marrow-derived cells in antibody production, establishing that thymus cells provided "helper" functions to enable B cells to generate against antigens like sheep red blood cells in irradiated hosts. By the early 1970s, T cells were distinguished into functional subsets: while some mediated direct cytotoxicity against infected or abnormal cells—as shown in 1970 by Cerottini, Nordin, and Brunner, who assigned cytotoxic activity to thymus-derived lymphocytes—others supported responses, leading to the initial recognition of "helper" T cells separate from B cells and cytotoxic T cells. This helper function was further characterized in adoptive transfer experiments, confirming T cells' indispensable role in without direct antibody production. The nomenclature evolved alongside these findings. In 1979, the monoclonal antibody OKT4 was developed, specifically recognizing a subset of T cells that exhibited helper activity , marking the first surface distinction of these cells and leading to their designation as OKT4+ or helper T cells. By the mid-1980s, this marker was identified as the glycoprotein, solidifying + T cells as the standard term for helper T cells, which express to interact with molecules on antigen-presenting cells. The subset nomenclature advanced in 1986, when Tim Mosmann and Coffman isolated murine + T cell clones and classified them into Th1 (producing interferon-γ and interleukin-2, promoting ) and Th2 (producing interleukins-4, -5, and -6, supporting humoral responses), based on distinct profiles. This Th1/Th2 paradigm became foundational for understanding T helper cell diversity.

Basic characteristics and markers

T helper cells, also known as CD4+ T cells, are primarily identified by their high expression of the CD4 co-receptor, a glycoprotein that binds to non-polymorphic regions of major histocompatibility complex (MHC) class II molecules on antigen-presenting cells, thereby stabilizing the interaction between the T cell receptor (TCR) and peptide-MHC complexes during antigen recognition. This CD4 expression distinguishes them as a subset of T lymphocytes essential for coordinating adaptive immune responses, with CD4 levels typically much higher on these cells compared to other immune populations. Subtypes of T helper cells are further delineated by unique combinations of intracellular transcription factors and cell surface markers, which reflect their specialized functions. For instance, Th1 cells are characterized by the T-bet and surface markers such as , Th2 cells by GATA3 and , Th17 cells by RORγt and CCR6, regulatory T cells (Tregs) by and CD25, and follicular helper T cells (Tfh) by Bcl6 along with expression. These markers enable precise identification through and , allowing researchers to track subset-specific behaviors . Naive T helper cells exhibit a long lifespan, often spanning years in humans through homeostatic driven by cytokines like IL-7, and they continuously recirculate between the and secondary lymphoid organs such as lymph nodes via interactions with molecules like (CD62L) and chemokine receptors CCR7. In contrast, upon activation, these cells differentiate into short-lived effector T helper cells that downregulate lymphoid homing receptors and migrate to inflamed peripheral tissues to exert their effects. While monocytes and other myeloid cells can express low levels of , T helper cells are clearly distinguished by their lymphoid lineage markers, including CD3 and the TCR complex, in the absence of monocyte-specific markers such as and , confirming their identity as adaptive immune effectors rather than innate .

Structure and development

Cellular morphology and receptors

T helper cells exhibit distinct morphological features that reflect their functional states. Naive T helper cells are small, spherical lymphocytes approximately 7-10 μm in diameter, with a large, centrally located occupying most of the cell volume and a thin rim of , resulting in a high -to- . Upon into effector cells, T helper cells increase in size, often reaching 10-12 μm or more, and develop a more abundant that may contain granules, particularly in subsets like Th1 cells, which aids in secretion. The (TCR) is the primary antigen-recognition structure on T helper cells, composed of a disulfide-linked αβ heterodimer with and domains that confer specificity for presented in the groove of class II (MHC II) molecules. Each α and β features a single immunoglobulin-like domain for antigen binding, a domain, a transmembrane region, and a short cytoplasmic tail; the TCR associates non-covalently with the CD3 complex (comprising γ, δ, ε, and ζ chains) to transduce signals, though the heterodimer itself does not possess intrinsic signaling capability. CD4 functions as the defining coreceptor for T helper cells, enhancing TCR affinity for MHC II through its extracellular region, which consists of two fibronectin type III-like domains followed by two immunoglobulin-like domains that bind to non-polymorphic regions on MHC II. The intracellular portion of CD4, a 38-amino-acid tail, interacts non-covalently with the Lck via two conserved residues, positioning Lck in proximity to the TCR-CD3 complex to initiate events upon engagement. This association is stabilized by zinc coordination in some contexts, underscoring CD4's role in signal amplification. Accessory molecules CD28 and CTLA-4, both members of the , regulate T helper cell activation by competing for the same ligands, (B7-1) and (B7-2), on antigen-presenting cells. exists as a disulfide-linked homodimer with an extracellular V-set immunoglobulin domain, a single transmembrane helix, and a short cytoplasmic tail containing YMNM and PYMKM motifs that recruit PI3K and upon ligation, delivering essential costimulatory signals for production and . CTLA-4, structurally similar to CD28 as a homodimer with a homologous extracellular domain sharing the conserved MYPPPY motif, binds and with 20- to 50-fold higher affinity and is rapidly internalized via clathrin-mediated , thereby sequestering ligands and inhibiting T cell responses to prevent .

Thymic development and selection

T cell development originates from hematopoietic stem cells in the , which differentiate into common lymphoid progenitors (CLPs) that seed the via the bloodstream. These early thymic progenitors (ETPs), upon entering the thymic cortex, lack expression of and coreceptors, marking them as double-negative (DN) thymocytes. The DN stage is subdivided into four phases (DN1–DN4) based on surface marker expression, such as and CD25, during which commitment to the T cell lineage occurs under the influence of signaling from thymic stromal cells. TCR gene rearrangement begins in the DN stages, with the TCRβ locus rearranging first at the DN2–DN3 transition. Successful production of a functional TCRβ chain pairs with pre-Tα to form the pre-TCR, triggering β-selection at the DN3 stage; this process promotes survival, proliferation, and differentiation of these cells into the DN4 stage, followed by progression to the stage where cells co-express and CD8. At the stage, TCRα rearrangement occurs, assembling the complete αβ TCR, which is displayed on the cell surface for subsequent selection processes. Positive selection ensures the survival of thymocytes capable of recognizing self-major histocompatibility complex (MHC) molecules. For future T helper cells, thymocytes interact with peptide-MHC class II complexes presented by cortical thymic epithelial cells (cTECs); weak TCR signals from these interactions rescue MHC II-restricted cells from , promoting their maturation. This avidity-dependent process, occurring primarily in the thymic cortex, shapes a repertoire restricted to self-MHC while allowing diversity for foreign antigen recognition. Negative selection eliminates thymocytes with high-affinity recognition of self-antigens to establish central and prevent . In the thymic medulla, strong TCR signals triggered by self-peptide/MHC complexes on medullary thymic epithelial cells (mTECs) or dendritic cells induce in self-reactive clones, with mTECs expressing tissue-restricted antigens via the (AIRE) to broaden self-. This process complements positive selection, ensuring only non-self-reactive T cells proceed to maturity. Commitment to the + T helper lineage occurs post-positive selection in MHC II-restricted thymocytes transitioning to single-positive () cells. Interaction with induces expression of the ThPOK (encoded by Zbtb7b), which is essential and sufficient for + lineage specification by repressing + lineage genes and promoting maintenance. ThPOK expression at the +lo intermediate stage locks in helper cell fate, distinguishing it from + cytotoxic lineage commitment.

Activation process

Antigen recognition and signal 1

Antigen presentation to T helper cells begins with professional antigen-presenting cells, particularly dendritic cells, which capture exogenous antigens through endocytosis or phagocytosis. These cells process the antigens in endosomal compartments, where lysosomal proteases degrade them into peptides of 13-25 amino acids suitable for binding to major histocompatibility complex class II (MHC II) molecules. The invariant chain (Ii) initially occupies the peptide-binding groove of nascent MHC II in the endoplasmic reticulum, preventing premature peptide loading and directing MHC II to endosomal compartments via the CLIP peptide segment. In these acidic, protease-rich compartments, Ii is degraded, and HLA-DM facilitates the exchange of CLIP for antigenic peptides, stabilizing the peptide-MHC II complex for surface expression. Mature dendritic cells upregulate MHC II surface density upon maturation, enhancing presentation efficiency to naive CD4+ T cells in secondary lymphoid organs. The (TCR) on naive T helper cells, composed of αβ chains non-covalently associated with the CD3 complex, specifically recognizes the -MHC complex presented on the surface. This recognition is highly specific, with the TCR complementarity-determining regions interacting with both the and MHC α-helices to discriminate foreign from self-. Activation requires a minimal , typically corresponding to constants in the micromolar range (1-100 μM) or TCR-pMHC dwell times of 1-3 seconds, below which naive + T cells fail to engage productively. Low- interactions, prevalent among antigen-specific clones (as low- cells can outnumber high- ones by up to 10-fold), still contribute to the T cell repertoire but demand serial engagements or higher antigen doses for effective signaling. The coreceptor stabilizes this interaction by binding invariant regions of MHC , increasing overall without altering specificity. Upon TCR engagement with peptide-MHC II, signal 1 initiates through conformational changes in the TCR-CD3 complex, recruiting and activating the Lck associated with CD4. Lck phosphorylates immunoreceptor tyrosine-based motifs (ITAMs) within the cytoplasmic tails of the CD3 γ, δ, ε chains, and ζ homodimer, creating docking sites for downstream effectors. Dual phosphorylation of ITAM tyrosines is critical for full signaling potency, as mono-phosphorylated ITAMs recruit but fail to fully activate effectors. The tandem SH2 domains of ζ-chain-associated 70 (ZAP-70) bind these phosphotyrosines, leading to ZAP-70 by Lck on its loop tyrosines (Y493 and Y492 in humans), relieving autoinhibition and enabling activity. Activated ZAP-70 then phosphorylates adaptor proteins like LAT and SLP-76, assembling a signalosome that propagates the activation cascade. Immediate downstream events include phospholipase Cγ1 (PLCγ1) by ZAP-70, which hydrolyzes PIP2 into IP3 and DAG, triggering calcium release from stores via IP3 receptors. This calcium flux, peaking within seconds to minutes, activates calmodulin-dependent , which dephosphorylates nuclear factor of activated T cells (NFAT) proteins. Dephosphorylated NFAT translocates to the nucleus, where it binds DNA consensus sites to drive transcription of genes like IL-2. Sustained calcium oscillations, facilitated by store-operated calcium entry through CRAC channels (Orai1/STIM1), ensure prolonged NFAT essential for T helper cell .

Costimulation and signal 2

The activation of T helper cells requires not only antigen recognition via the (TCR) but also a costimulatory signal, known as signal 2, to ensure full responsiveness and prevent tolerance. The primary costimulatory pathway involves the interaction between on T helper cells and B7 ligands ( and ) expressed on antigen-presenting cells (APCs). This CD28-B7 engagement provides essential second signals that promote , survival, and production, particularly in naive CD4+ T cells responding to antigens. Upon ligation, recruits intracellular signaling molecules, activating the phosphatidylinositol 3-kinase (PI3K)-Akt pathway, which is critical for downstream effects in T helper cell activation. The PI3K-Akt signaling cascade enhances the production of interleukin-2 (IL-2) by stabilizing IL-2 mRNA and upregulating its transcription, thereby driving T cell clonal expansion and preventing . Additionally, this pathway supports metabolic reprogramming, including increased glucose uptake and , which are necessary for the biosynthetic demands of proliferating T cells. To balance activation, inhibitory signals counteract costimulation through CTLA-4, a homolog of expressed on activated T cells. CTLA-4 competes with for binding to and ligands due to its higher affinity, thereby limiting costimulatory access and dampening T cell responses. Upon engagement, CTLA-4 recruits phosphatases such as PP2A and SHP-2, which dephosphorylate key signaling intermediates, inhibiting PI3K activation and reducing IL-2 production. The absence of CD28-B7 costimulation during TCR engagement leads to T cell anergy, a state of functional unresponsiveness, or in some cases, deletion of self-reactive clones, thereby maintaining . This mechanism ensures that only T helper cells encountering antigens in the context of appropriate activation proceed to effector functions, avoiding .

Cytokine signaling and signal 3

Upon activation through antigen recognition (signal 1) and (signal 2), naïve CD4+ T cells require -mediated signal 3 to direct their into specific effector subsets, ensuring appropriate immune responses tailored to the or environmental context. This third signal integrates with prior activation cues to modulate gene expression, particularly through the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway, which transduces extracellular signals into intracellular transcriptional changes. Key cytokines instruct lineage commitment by activating specific proteins that promote master transcription factors. For Th1 differentiation, IL-12 primarily signals through JAK2/ to induce T-bet expression, while IFN-γ reinforces this via JAK1/STAT1, enhancing IFN-γ production and activation. In Th2 polarization, IL-4 activates JAK1/JAK3/STAT6, driving GATA3 upregulation and IL-4/IL-5/IL-13 secretion for and anti-parasitic responses. Th17 cells arise under the influence of TGF-β combined with IL-6, where IL-6 engages JAK1/ to promote RORγt and IL-17 production, countering extracellular bacteria and fungi. For regulatory T cells (Treg), TGF-β initiates expression, which is stabilized by IL-2 signaling through JAK1/JAK3/STAT5, maintaining . Signal 3 cytokines do not act in isolation but integrate with TCR and costimulatory signals to fine-tune ; for instance, TCR-induced pathways prime responsiveness, allowing cytokines to selectively amplify lineage-specific transcription factors like T-bet or GATA3 without overriding initial . This coordination prevents default fates and ensures robust effector function, as evidenced by impaired when signal 3 precedes or disrupts TCR engagement. Tissue-specific environmental cues further shape signal 3 by varying local availability; for example, high TGF-β in mucosal sites favors Treg or Th17 fates, while inflammatory milieus rich in IL-12 promote Th1 responses in infected tissues, adapting helper cell profiles to localized threats.

Differentiation into subsets

Th1 and Th2 polarization

The classical Th1/Th2 dichotomy was first proposed in based on observations of distinct secretion profiles among murine CD4+ T cell clones, where Th1 cells primarily produce interferon-gamma (IFN-γ) and interleukin-2 (IL-2), while Th2 cells secrete IL-4, IL-5, and IL-10. This model highlighted how these subsets drive divergent immune responses, with Th1 cells promoting against intracellular pathogens and Th2 cells supporting , including production and activation often associated with allergic responses. Th1 polarization occurs when naive CD4+ T cells are exposed to IL-12, a produced by activated dendritic cells and s during encounters with intracellular pathogens like . IL-12 signaling activates STAT4, which induces expression of the T-bet (encoded by ), the master regulator that commits cells to the Th1 by directly promoting IFN-γ production and repressing Th2-associated genes.80702-3) T-bet further amplifies Th1 through on IL-12 receptor expression, ensuring robust cell-mediated responses such as activation and support. In contrast, Th2 polarization is driven by IL-4, often derived from innate sources like or mast cells during parasitic infections, which activates STAT6 to upregulate GATA3, the key for Th2 commitment. GATA3 binds to regulatory elements in the Il4, Il5, and Il13 loci, orchestrating production that fosters class switching to IgE and recruitment of , thereby enhancing humoral defenses but also contributing to allergic inflammation.80240-8) Mutual cross-regulation maintains the Th1/Th2 balance, with IFN-γ from Th1 cells inhibiting Th2 and by downregulating IL-4 receptor expression and GATA3 activity, while IL-4 from Th2 cells suppresses Th1 by reducing IL-12 responsiveness and T-bet . This antagonistic interplay, evident in early studies of T cell clones, prevents simultaneous dominance of both subsets and fine-tunes adaptive immunity to pathogen type.

Th17 and Treg differentiation

Th17 cells differentiate from naïve CD4+ T cells in response to transforming growth factor-β (TGF-β) combined with pro-inflammatory cytokines such as interleukin-6 (IL-6) or IL-23, which drive the expression of the retinoic acid receptor-related orphan receptor γt (RORγt).01105-6) RORγt orchestrates the Th17 differentiation program by promoting genes associated with interleukin-17 (IL-17) production, including Il17a and Il17f, while suppressing alternative lineages.01105-6) These cells produce IL-17 family cytokines that recruit and activate neutrophils, playing a critical role in host defense against extracellular bacteria and fungi, such as , by inducing and neutrophil chemoattractants like and CXCL2. In contrast, regulatory T (Treg) cells arise through pathways that emphasize immune suppression, with serving as the master that defines their identity and suppressive function. Differentiation of induced Treg (iTreg) cells occurs in the periphery from naïve + T cells under the influence of TGF-β and IL-2, which stabilize expression and inhibit pro-inflammatory responses. Natural Treg (nTreg) cells, however, develop in the from + single-positive thymocytes that encounter self-antigens with intermediate affinity, leading to upregulation and their export to peripheral tissues for ongoing .00199-X) The discovery of these + Treg cells and their role in maintaining peripheral was recognized by the 2025 in Physiology or Medicine, awarded to Shimon Sakaguchi, Fred Ramsdell, and Mary Brunkow for their foundational contributions. A key feature of Th17 and Treg lineages is their developmental plasticity, influenced by cytokine environments that can drive interconversion between these subsets. High levels of TGF-β with IL-6 favor Th17 commitment by promoting RORγt and inhibiting , whereas shifting to TGF-β with IL-2 enhances expression and Treg differentiation, potentially converting Th17-like cells into suppressive Tregs. This bidirectional plasticity allows adaptive responses to inflammatory cues but can contribute to immune dysregulation if unbalanced.

Emerging subsets and plasticity

T follicular helper (Tfh) cells represent a specialized of + T helper cells that provide critical assistance to s within germinal centers of secondary lymphoid organs, characterized by expression of the and driven by the . acts as the master regulator of Tfh differentiation, repressing alternative T helper fates and enabling the production of cytokines such as IL-21 to promote proliferation, class switching, and affinity maturation. These cells migrate to follicles via -mediated to ligand, ensuring localized support for . Beyond classical subsets, Th9 cells emerge as IL-9-secreting effectors derived from naive + T cells under the influence of TGF-β and IL-4, contributing to allergic responses, anti-parasitic defense, and antitumor activity. IL-9 production by Th9 cells is transcriptionally regulated by factors like and PU.1, amplifying mucosal and activation without strong overlap with Th2 cytokine profiles. Similarly, Th22 cells produce in response to IL-6 and TNF-α, playing a key role in epithelial barrier protection against extracellular pathogens and maintaining mucosal homeostasis. from Th22 cells induces and proteins in epithelial cells, fortifying defenses at and gut interfaces without promoting overt . T helper cell plasticity refers to the dynamic reprogramming of lineage commitments, often mediated by epigenetic modifications that allow between subsets in response to environmental cues. For instance, Th17 cells can shift toward a Th1-like under IL-12 influence, acquiring IFN-γ production through at the Ifng locus, which is partially accessible in Th17 cells due to RORγt and T-bet interplay. This plasticity is governed by histone acetylation and changes, enabling adaptive responses but also contributing to chronic inflammation in . Recent studies from 2023 to 2025 have highlighted stem-like properties in + T cells, revealing intrinsic programs that confer adaptability and long-term potential beyond rigid subset definitions. These stemness features, marked by expression of Tcf7 and self-renewal capacity, allow + T cells to maintain progenitor states for sustained effector in tumors and , redefining fate decisions through reversible epigenetic landscapes. Such adaptations underscore the of T helper cell responses, where environmental signals can redirect stem-like precursors to optimize immunity.

Effector functions

Cytokine secretion profiles

T helper cells differentiate into distinct subsets, each characterized by unique secretion profiles that dictate their effector functions in immune responses. These profiles are established during and enable targeted modulation of , production, and immune suppression. The seminal identification of Th1 and Th2 subsets highlighted how differential patterns lead to contrasting roles in cell-mediated versus . Th1 cells primarily secrete interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α), which promote macrophage activation, enhance , and drive cytotoxic responses against intracellular pathogens. In contrast, Th2 cells produce interleukin-4 (IL-4), IL-5, and IL-13, cytokines that stimulate class switching to IgE, activation, and hyperplasia, thereby supporting defenses against extracellular parasites and contributing to allergic responses. These mutually exclusive profiles were first delineated in murine T cell clones, where Th1 cells showed high IFN-γ but negligible IL-4, while Th2 cells exhibited the opposite pattern. Th17 cells are defined by their production of IL-17A, IL-17F, and , which induce expression to recruit neutrophils, promote epithelial barrier integrity, and amplify at mucosal sites, particularly against extracellular and fungi. Regulatory T (Treg) cells, on the other hand, secrete IL-10 and transforming growth factor-beta (TGF-β) to dampen effector T cell proliferation, inhibit pro-inflammatory release, and maintain , preventing . These profiles distinguish Th17 from other subsets by their pro-inflammatory yet tissue-protective effects, while Treg cytokines enforce through direct receptor engagement on target cells. The specificity of these cytokine secretion profiles is regulated by lineage-determining transcription factors that activate subset-specific promoters and enhancers in genes, coupled with feedback loops that reinforce polarization. For instance, T-bet in Th1 cells binds the IFN-γ promoter to boost its expression, while IFN-γ signaling further upregulates T-bet via , creating a circuit; similarly, GATA3 drives IL-4, IL-5, and IL-13 in Th2 cells through autoregulatory loops. In Th17 cells, RORγt directs IL-17 and transcription, with IL-21 providing autocrine reinforcement, whereas in Treg cells represses pro-inflammatory loci while promoting IL-10 and TGF-β via Smad complexes. These mechanisms ensure stable, heritable patterns while allowing plasticity under chronic stimulation.

Cell-cell interactions and orchestration

T helper cells (Th cells) engage in direct cell-cell interactions to coordinate adaptive immune responses, primarily through membrane-bound molecules that facilitate signaling without relying on soluble cytokines. A key example is the interaction between CD40 ligand (CD40L) on activated Th cells and CD40 on , which is essential for activation and class-switch recombination in germinal centers. This ligation triggers proliferation, survival, and differentiation into plasma cells capable of producing high-affinity antibodies of specific isotypes, such as IgG or IgA, thereby amplifying . Th cells also provide contact-dependent support to CD8+ T cells by licensing dendritic cells (DCs) and promoting their maturation, enhancing cross-presentation of antigens to cytotoxic T cells. Through brief interactions at the interface of + T cells and DCs, Th cells upregulate costimulatory molecules on DCs, such as and , which in turn bolster + T cell priming and effector function. Additionally, Th cells supply IL-2 during these encounters to sustain + T cell expansion and memory formation, ensuring robust cellular immunity against intracellular pathogens. Regulatory T cells (Tregs), a subset of Th cells, exert suppression through contact-dependent mechanisms to maintain immune homeostasis and prevent excessive responses. Tregs express CTLA-4, which binds to CD80 and CD86 on antigen-presenting cells with higher affinity than CD28 on effector T cells, thereby depriving the latter of costimulation and downregulating APC activation via trans-endocytosis of these ligands. Another contact-dependent pathway involves latency-associated peptide (LAP), a surface-bound form of TGF-β on Tregs, which directly inhibits effector T cell proliferation and cytokine production upon cell-cell contact, independent of soluble TGF-β release. These interactions are spatially orchestrated within secondary lymphoid tissues, such as lymph nodes and , where Th cells localize to specific niches like the T-B border and to optimize coordination. T follicular helper (Tfh) cells, a specialized Th subset, migrate into B cell follicles guided by like , positioning them to interact sequentially with multiple in a choreographed manner that sustains germinal center reactions. This architectural organization ensures efficient amplification of immune responses while minimizing off-target effects.

Memory formation and maintenance

Generation of memory T helper cells

Upon activation, naïve T helper cells undergo , a process that unequally partitions cellular components between daughter cells, thereby generating one biased toward fate and another toward effector . This mechanism ensures self-renewal and the production of long-lived memory precursors during clonal expansion, as demonstrated in studies of CD4+ T cell responses to where proteins like PKCζ and Numb are asymmetrically inherited to regulate fate decisions. Memory T helper cells are phenotypically distinguished from effectors and naïve cells by surface markers such as CD45RO expression, which replaces CD45RA and indicates a , while CCR7 expression further subdivides them into central memory (CD45RO+ CCR7+) cells that home to lymph nodes and effector memory (CD45RO+ CCR7-) cells that patrol peripheral tissues. These markers reflect migratory and functional adaptations, with central memory cells retaining proliferative potential and effector memory cells poised for rapid release upon re-encounter with . The survival and homeostatic proliferation of memory T helper cells depend on cytokines IL-7 and IL-15, which provide essential signals for maintenance in the absence of . IL-7 acts as the primary regulator, promoting the transition of effectors to memory cells by enhancing survival without inducing proliferation, while IL-15 serves as an accessory cytokine supporting basal turnover and longevity of antiviral CD4+ memory populations. Blockade of IL-7 leads to significant loss of memory CD4+ T cells, underscoring its non-redundant role. Epigenetic modifications establish stable programming in memory T helper cells, particularly at cytokine loci, enabling rapid and subset-specific recall responses. During differentiation, chromatin remodeling poises genes like IFNG, IL4, and IL13 through histone variants such as H2A.Z and accessible enhancers, maintaining an epigenetically primed state that persists into memory and distinguishes it from transient effector profiles. This poising facilitates quicker transcriptional activation upon rechallenge, contributing to immunological memory.

Long-term regulation and exhaustion

Memory CD4+ T cells maintain long-term through (IL-7R) signaling, which is essential for their survival and slow homeostatic in the absence of . This is primarily produced by stromal cells in secondary lymphoid organs such as lymph nodes and , as well as in the , where mesenchymal stem cells serve as a key source supporting CD4+ T cell persistence. In these niches, memory cells compete for limited IL-7 and space, ensuring a balanced pool size that prevents overexpansion while sustaining protective immunity; for instance, during recovery from , niches preferentially support IL-7-dependent of CD4+ T cells over naïve ones. Disruption of IL-7R signaling leads to progressive loss of these cells, underscoring its role in niche competition and long-term maintenance across lymphoid and non-lymphoid tissues. In contrast, chronic antigen exposure during persistent infections or cancer can drive memory CD4+ T cells into a state of exhaustion, characterized by progressive hyporesponsiveness and loss of effector functions. This dysfunction involves upregulation of inhibitory receptors such as programmed death-1 (PD-1) and T-cell immunoglobulin and mucin domain-containing-3 (TIM-3), which are expressed on antigen-specific CD4+ T cells in models of chronic viral infections like virus (LCMV). PD-1 engagement with its ligand dampens T cell receptor signaling, reducing cytokine production and proliferation, while TIM-3 further exacerbates this by promoting and metabolic impairments, leading to a hierarchical loss of functions in exhausted CD4+ T cells. In human chronic infections, such as or hepatitis C, co-expression of PD-1 and TIM-3 on CD4+ T cells correlates with impaired helper activity and increased viral persistence. Exhaustion in memory CD4+ T cells can be partially reversed through checkpoint blockade therapies targeting PD-1/PD-L1 interactions, which reinvigorate hyporesponsive cells by restoring proliferative capacity and secretion. In tumor microenvironments, anti-PD-1 antibodies have been shown to enhance the helper functions of exhausted PD-1high CD39+ + T cells, promoting cross-talk with + effectors and improving antitumor responses. This reversal is not complete, as epigenetic changes in exhausted cells may limit full recovery, but it highlights PD-1 as a central regulator amenable to therapeutic intervention in chronic settings. Recent studies have identified stem-like memory as a subset resistant to exhaustion, capable of self-renewal and differentiation into functional effectors even under chronic stimulation. These cells, marked by transcription factors like TCF-1, emerge early during exposure and maintain a progenitor-like state that sustains long-term immunity without succumbing to inhibitory receptor upregulation. For example, neoantigen-specific stem cell memory-like have demonstrated enhanced persistence and mediation of -dependent tumor control in solid tumors, resisting exhaustion through preserved metabolic fitness. In 2024 research on , stem-like populations offered insights into bolstering durable responses via targeting these progenitors. As of 2025, emerging evidence highlights metabolic factors, such as early availability, in attenuating exhaustion during chronic stimulation, potentially enhancing memory maintenance and therapeutic outcomes in cancer and persistent infections.

Role in health and disease

Protective immunity against pathogens

T helper cells play a central role in orchestrating adaptive immune responses against pathogens by differentiating into specialized subsets that coordinate effector mechanisms. Th1 cells, characterized by their production of interferon-gamma (IFN-γ), are essential for controlling intracellular bacterial and viral infections. IFN-γ activates macrophages, enhancing their phagocytic and microbicidal activities, such as increased production of and , which restrict pathogen replication within phagosomes.30474-6)00118-7) For instance, in infections like or , Th1-derived IFN-γ is critical for macrophage-mediated clearance of intracellular bacteria. Th2 cells contribute to protective immunity against extracellular parasites, particularly helminths, by secreting cytokines such as interleukin-5 (IL-5). IL-5 promotes the differentiation, activation, and recruitment of , which release cytotoxic granules containing major basic protein and to damage helminth cuticles and facilitate parasite expulsion. This mechanism is evident in infections with nematodes like Nippostrongylus brasiliensis, where Th2-driven correlates with worm clearance from the gut.30516-2) Th17 cells provide defense at mucosal barriers against extracellular bacteria and fungi through interleukin-17 (IL-17) production, which induces expression to recruit neutrophils. Neutrophils then release and form extracellular traps to contain and eliminate pathogens like or . In fungal infections, IL-17 signaling enhances epithelial barrier integrity and promotes antifungal peptide production, underscoring Th17's role in preventing invasive disease.00552-9) In vaccine responses, T helper cells, especially the follicular helper subset (Tfh), are indispensable for T cell-dependent activation and production. Tfh cells provide and cytokines like IL-21 to B cells, driving class-switch recombination, affinity maturation, and generation of high-affinity neutralizing . This process underpins the efficacy of such as those against or , where Th-mediated help ensures long-lasting .

Involvement in autoimmunity and allergies

Dysregulated T helper cells play a central role in the of autoimmune diseases and allergies, where imbalances in their subsets lead to excessive and tissue damage. In , overactivation of pro-inflammatory subsets like Th17 cells promotes chronic , while defects in regulatory T (Treg) cells fail to suppress autoreactive responses. Similarly, in allergic conditions, Th2 cells drive type 2 immune responses characterized by and mucus hypersecretion. These dysregulations highlight the delicate balance required for immune , with therapeutic strategies increasingly targeting specific cytokines or cell functions to mitigate progression. Th17 cells contribute significantly to autoimmune diseases such as (RA) and through the production of interleukin-17 (IL-17), which induces pro-inflammatory cascades. In RA, elevated Th17 cells and IL-17 levels in promote joint inflammation by recruiting neutrophils and enhancing expression, leading to cartilage destruction. Similarly, in , Th17-derived IL-17 stimulates proliferation and antimicrobial peptide production, perpetuating skin plaques and epidermal . These effects are amplified by IL-23, which stabilizes Th17 differentiation and secretion, underscoring the IL-23/IL-17 axis as a key therapeutic target in these conditions. In allergic diseases like and , Th2 cells exacerbate pathology via IL-4 and IL-13 secretion, fostering IgE production and tissue remodeling. In , Th2-mediated IL-4 promotes B-cell class switching to IgE, while IL-13 drives and airway mucus hypersecretion, contributing to . In , these cytokines impair epidermal barrier function by downregulating expression and inducing production that recruits Th2 cells and , resulting in chronic skin inflammation. Dual inhibition of IL-4 and IL-13 signaling has shown efficacy in reducing these Th2-driven responses in clinical settings. Defects in Treg cells, which normally suppress autoreactive T cells through mechanisms like IL-10 secretion and CTLA-4-mediated inhibition, are implicated in autoimmune disorders including (T1D) and (IBD). In T1D, impaired Treg suppressive function, particularly in , allows unchecked Th1 and Th17 responses against beta cells, accelerating islet destruction. Functional studies reveal that Tregs from T1D patients exhibit impaired and production, contributing to disease onset. In IBD, particularly , dysfunctional Treg activity in the gut mucosa, coupled with defective IL-10 signaling, fails to control effector T cell activity, leading to chronic intestinal inflammation. Restoring Treg function through ex vivo expansion has demonstrated potential to ameliorate in preclinical models. A striking example of Treg dysfunction is the immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, caused by mutations in the gene, which encodes the master regulator of Treg and . These loss-of-function mutations result in absent or non-functional Tregs, leading to multi-organ including severe enteropathy, endocrinopathies, and from early infancy. Patients exhibit elevated autoreactive antibodies and uncontrolled Th1/Th2/Th17 responses due to the lack of FOXP3-mediated suppression. remains the primary curative approach, highlighting the critical role of FOXP3 in preventing systemic .

Immunodeficiency and viral infections

T helper cells are central to immune defense, and their depletion underlies severe , most notably in infection. primarily targets + T cells, leading to progressive decline in their numbers and function, which impairs both humoral and cellular immunity. This results in increased susceptibility to opportunistic infections, such as and , and certain cancers. As of 2025, antiretroviral therapy (ART) effectively restores + counts and immune function in most patients, preventing progression to AIDS, though chronic immune activation and exhaustion persist in some cases.

Cancer and therapeutic targeting

T helper 1 (Th1) cells play a critical role in antitumor immunity by coordinating with cytotoxic + T cells to enhance tumor cell killing, primarily through the secretion of interferon-gamma (IFN-γ). Th1 cells produce IFN-γ, which activates + T cells, promotes their infiltration into the , and boosts their cytotoxic functions, including the release of perforin and to directly lyse tumor cells. This coordination is essential for effective antitumor responses, as IFN-γ also upregulates class I expression on tumor cells, making them more susceptible to + T cell recognition and elimination. In contrast, regulatory T cells (Tregs), a of CD4+ T helper cells, suppress antitumor immunity within the , contributing to immune evasion and tumor progression. Tregs inhibit effector T cell functions through mechanisms such as IL-10 and TGF-β secretion, CTLA-4-mediated competition for costimulatory signals, and direct cell-cell contact that dampens + T cell activation and proliferation.00042-9.pdf) High Treg infiltration in tumors correlates with poor across various cancers, as they create an immunosuppressive niche that hinders Th1 and + T cell-mediated killing. Therapeutic strategies targeting T helper cells have emerged to bolster antitumor responses, including chimeric antigen receptor (CAR) T cell therapies that incorporate CD4+ helper cells. CD4+ CAR T cells provide helper functions such as support and enhance the persistence and efficacy of CD8+ CAR T cells, while also demonstrating intrinsic tumor-killing capabilities independent of CD8+ counterparts in certain solid tumors. Additionally, inhibitors like anti-PD-1 and anti-CTLA-4 antibodies reinvigorate Th1 responses by relieving Treg-mediated suppression and promoting IFN-γ production, leading to improved CD8+ T cell activation and tumor regression in cancers such as and lung carcinoma. Recent advances in 2024-2025 clinical trials focus on Treg depletion to enhance , with promising results from CD25-targeted antibodies. Agents like RG6292 (vopikitug) and PF-08046032 selectively deplete tumor-infiltrating Tregs, increasing effector T cell infiltration and antitumor activity in phase 1/2 trials for solid tumors including gastric and colorectal cancers, with manageable safety profiles and early signs of objective responses. CCR8 antagonists, such as those in ongoing trials, further target Treg-specific markers to reduce suppression without broad , showing enhanced efficacy when combined with PD-1 inhibitors. These approaches aim to shift the toward a Th1-dominant state, amplifying overall antitumor immunity.