T helper cells, also known as CD4⁺ T cells, are a subset of T lymphocytes that serve as key orchestrators of the adaptive immune response, providing essential help to other immune cells such as B cells, cytotoxic T cells, and macrophages to mount effective defenses against pathogens.[1][2] These cells are considered crucial for nearly all adaptive immune functions, as they recognize antigens presented by major histocompatibility complex (MHC) class II molecules on antigen-presenting cells, leading to their activation and proliferation.[3] Upon activation, naive T helper cells differentiate into specialized effector subsets, enabling tailored responses to diverse threats like intracellular pathogens, extracellular bacteria, and parasites.[4]The differentiation of T helper cells is driven by signals from the microenvironment, including cytokines and antigen-presenting cell interactions, resulting in distinct subsets such as Th1, Th2, Th17, and T follicular helper (Tfh) cells.[5] Th1 cells primarily produce interferon-gamma (IFN-γ) to activate macrophages and promote cell-mediated immunity against intracellular microbes, while Th2 cells secrete interleukins like IL-4, IL-5, and IL-13 to support B cell antibody production and combat helminth infections.[6] Th17 cells, characterized by IL-17 production, are vital for defense against extracellular bacteria and fungi at mucosal surfaces but can contribute to autoimmune diseases when dysregulated.[7] Additionally, regulatory T cells (Tregs), a suppressive subset, maintain immune homeostasis by inhibiting excessive responses and preventing autoimmunity; their discovery was recognized by the 2025 Nobel Prize in Physiology or Medicine awarded to Mary E. Brunkow, Fred Ramsdell, and Shimon Sakaguchi.[8][9]Beyond infection control, T helper cells play pivotal roles in immune memory formation, vaccine responses, and pathological conditions; for instance, they are indispensable for generating long-lived memory CD8⁺ T cells and high-affinity antibodies.[10] Their dysfunction, as seen in HIV infection where CD4⁺ counts decline dramatically, underscores their importance, leading to broad immunodeficiency.[11] Ongoing research highlights the plasticity of these subsets, allowing adaptation to complex immune challenges.[12]
Definition and overview
Discovery and nomenclature
The discovery of T helper cells traces back to the early 1960s, when Jacques Miller identified the thymus 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 humoral immunity.[13] 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 antibodies against antigens like sheep red blood cells in irradiated hosts.[14]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 antibody responses, leading to the initial recognition of "helper" T cells separate from B cells and cytotoxic T cells.[15] This helper function was further characterized in adoptive transfer experiments, confirming T cells' indispensable role in humoral immunity without direct antibody production.[16]The nomenclature evolved alongside these findings. In 1979, the monoclonal antibody OKT4 was developed, specifically recognizing a subset of human T cells that exhibited helper activity in vitro, marking the first surface distinction of these cells and leading to their designation as OKT4+ or helper T cells.[17] By the mid-1980s, this marker was identified as the CD4 glycoprotein, solidifying CD4+ T cells as the standard term for helper T cells, which express CD4 to interact with MHC class II molecules on antigen-presenting cells.[17] The subset nomenclature advanced in 1986, when Tim Mosmann and Robert Coffman isolated murine CD4+ T cell clones and classified them into Th1 (producing interferon-γ and interleukin-2, promoting cell-mediated immunity) and Th2 (producing interleukins-4, -5, and -6, supporting humoral responses), based on distinct cytokine profiles.[18] This Th1/Th2 paradigm became foundational for understanding T helper cell diversity.[18]
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.[19][1] 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.[20]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 transcription factor T-bet and surface markers such as CXCR3, Th2 cells by GATA3 and CCR4, Th17 cells by RORγt and CCR6, regulatory T cells (Tregs) by Foxp3 and CD25, and follicular helper T cells (Tfh) by Bcl6 along with CXCR5 expression.[21][22] These markers enable precise identification through flow cytometry and immunohistochemistry, allowing researchers to track subset-specific behaviors in vivo.[23]Naive T helper cells exhibit a long lifespan, often spanning years in humans through homeostatic proliferation driven by cytokines like IL-7, and they continuously recirculate between the blood and secondary lymphoid organs such as lymph nodes via interactions with adhesion molecules like L-selectin (CD62L) and chemokine receptors CCR7.[24][25] 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.[26][27]While monocytes and other myeloid cells can express low levels of CD4, 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 CD14 and CD16, confirming their identity as adaptive immune effectors rather than innate phagocytes.[28]
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 nucleus occupying most of the cell volume and a thin rim of cytoplasm, resulting in a high nucleus-to-cytoplasmratio.[29][30] Upon differentiation into effector cells, T helper cells increase in size, often reaching 10-12 μm or more, and develop a more abundant cytoplasm that may contain granules, particularly in subsets like Th1 cells, which aids in cytokine secretion.[31]The T cell receptor (TCR) is the primary antigen-recognition structure on T helper cells, composed of a disulfide-linked αβ heterodimer with variable and constant domains that confer specificity for peptideantigens presented in the groove of major histocompatibility complex class II (MHC II) molecules.[32] Each α and β chain features a single immunoglobulin-like variable domain for antigen binding, a constant 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.[33]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.[33] The intracellular portion of CD4, a 38-amino-acid tail, interacts non-covalently with the Src family kinase Lck via two conserved cysteine residues, positioning Lck in proximity to the TCR-CD3 complex to initiate phosphorylation events upon ligand engagement.[34] This association is stabilized by zinc coordination in some contexts, underscoring CD4's role in signal amplification.[35]Accessory molecules CD28 and CTLA-4, both members of the immunoglobulin superfamily, regulate T helper cell activation by competing for the same ligands, CD80 (B7-1) and CD86 (B7-2), on antigen-presenting cells. CD28 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 Grb2 upon ligation, delivering essential costimulatory signals for cytokine production and proliferation.[36] CTLA-4, structurally similar to CD28 as a homodimer with a homologous extracellular domain sharing the conserved MYPPPY motif, binds CD80 and CD86 with 20- to 50-fold higher affinity and is rapidly internalized via clathrin-mediated endocytosis, thereby sequestering ligands and inhibiting T cell responses to prevent autoimmunity.[37]
Thymic development and selection
T cell development originates from hematopoietic stem cells in the bone marrow, which differentiate into common lymphoid progenitors (CLPs) that seed the thymus via the bloodstream.[38] These early thymic progenitors (ETPs), upon entering the thymic cortex, lack expression of CD4 and CD8 coreceptors, marking them as double-negative (DN) thymocytes.[38] The DN stage is subdivided into four phases (DN1–DN4) based on surface marker expression, such as CD44 and CD25, during which commitment to the T cell lineage occurs under the influence of Notch signaling from thymic stromal cells.[39]TCR gene rearrangement begins in the DN stages, with the TCRβ locus rearranging first at the DN2–DN3 transition.[39] 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 double-positive (DP) stage where cells co-express CD4 and CD8.[39] At the DP stage, TCRα rearrangement occurs, assembling the complete αβ TCR, which is displayed on the cell surface for subsequent selection processes.[39]Positive selection ensures the survival of thymocytes capable of recognizing self-major histocompatibility complex (MHC) molecules.[40] For future T helper cells, DP 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 apoptosis, promoting their maturation.[40] This avidity-dependent process, occurring primarily in the thymic cortex, shapes a repertoire restricted to self-MHC while allowing diversity for foreign antigen recognition.[40]Negative selection eliminates thymocytes with high-affinity recognition of self-antigens to establish central tolerance and prevent autoimmunity.[40] In the thymic medulla, strong TCR signals triggered by self-peptide/MHC complexes on medullary thymic epithelial cells (mTECs) or dendritic cells induce apoptosis in self-reactive clones, with mTECs expressing tissue-restricted antigens via the autoimmune regulator (AIRE) to broaden self-tolerance.[40] This process complements positive selection, ensuring only non-self-reactive T cells proceed to maturity.[40]Commitment to the CD4+ T helper lineage occurs post-positive selection in MHC II-restricted DP thymocytes transitioning to single-positive (SP) cells.[41] Interaction with MHC class II induces expression of the transcription factor ThPOK (encoded by Zbtb7b), which is essential and sufficient for CD4+ lineage specification by repressing CD8+ lineage genes and promoting CD4 maintenance.[41] ThPOK expression at the CD4+CD8lo intermediate stage locks in helper cell fate, distinguishing it from CD8+ cytotoxic lineage commitment.[41]
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.[42] 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.[42] 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.[42] 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.[42] Mature dendritic cells upregulate MHC II surface density upon maturation, enhancing presentation efficiency to naive CD4+ T cells in secondary lymphoid organs.[42]The T cell receptor (TCR) on naive T helper cells, composed of αβ chains non-covalently associated with the CD3 complex, specifically recognizes the peptide-MHC II complex presented on the dendritic cell surface.[43] This recognition is highly specific, with the TCR complementarity-determining regions interacting with both the peptide and MHC II α-helices to discriminate foreign from self-peptides.[43] Activation requires a minimal affinitythreshold, typically corresponding to dissociation constants in the micromolar range (1-100 μM) or TCR-pMHC II dwell times of 1-3 seconds, below which naive CD4+ T cells fail to engage productively.[44] Low-affinity interactions, prevalent among antigen-specific clones (as low-affinity cells can outnumber high-affinity ones by up to 10-fold), still contribute to the T cell repertoire but demand serial engagements or higher antigen doses for effective signaling.[45] The coreceptor CD4 stabilizes this interaction by binding invariant regions of MHC II, increasing overall avidity without altering specificity.[46]Upon TCR engagement with peptide-MHC II, signal 1 initiates through conformational changes in the TCR-CD3 complex, recruiting and activating the Src family kinase Lck associated with CD4.[43] Lck phosphorylates immunoreceptor tyrosine-based activation motifs (ITAMs) within the cytoplasmic tails of the CD3 γ, δ, ε chains, and ζ homodimer, creating docking sites for downstream effectors.[47] Dual phosphorylation of ITAM tyrosines is critical for full signaling potency, as mono-phosphorylated ITAMs recruit but fail to fully activate effectors.[48] The tandem SH2 domains of ζ-chain-associated protein kinase 70 (ZAP-70) bind these phosphotyrosines, leading to ZAP-70 phosphorylation by Lck on its activation loop tyrosines (Y493 and Y492 in humans), relieving autoinhibition and enabling kinase activity.[49] Activated ZAP-70 then phosphorylates adaptor proteins like LAT and SLP-76, assembling a signalosome that propagates the activation cascade.[43]Immediate downstream events include phospholipase Cγ1 (PLCγ1) activation by ZAP-70, which hydrolyzes PIP2 into IP3 and DAG, triggering calcium release from endoplasmic reticulum stores via IP3 receptors.[50] This calcium flux, peaking within seconds to minutes, activates calmodulin-dependent phosphatasecalcineurin, which dephosphorylates nuclear factor of activated T cells (NFAT) proteins.[51] Dephosphorylated NFAT translocates to the nucleus, where it binds DNA consensus sites to drive transcription of activation genes like IL-2.[51] Sustained calcium oscillations, facilitated by store-operated calcium entry through CRAC channels (Orai1/STIM1), ensure prolonged NFAT activation essential for T helper cell commitment.[50]
Costimulation and signal 2
The activation of T helper cells requires not only antigen recognition via the T cell receptor (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 CD28 on T helper cells and B7 ligands (CD80 and CD86) expressed on antigen-presenting cells (APCs). This CD28-B7 engagement provides essential second signals that promote T cell proliferation, survival, and cytokine production, particularly in naive CD4+ T cells responding to antigens.Upon ligation, CD28 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 apoptosis. Additionally, this pathway supports metabolic reprogramming, including increased glucose uptake and glycolysis, which are necessary for the biosynthetic demands of proliferating T cells.[52]To balance activation, inhibitory signals counteract CD28 costimulation through CTLA-4, a homolog of CD28 expressed on activated T cells. CTLA-4 competes with CD28 for binding to CD80 and CD86 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.[53]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 peripheral tolerance. This mechanism ensures that only T helper cells encountering antigens in the context of appropriate APC activation proceed to effector functions, avoiding autoimmunity.
Cytokine signaling and signal 3
Upon activation through antigen recognition (signal 1) and costimulation (signal 2), naïve CD4+ T cells require cytokine-mediated signal 3 to direct their differentiation into specific effector subsets, ensuring appropriate immune responses tailored to the pathogen or environmental context.[54] 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 cytokine signals into intracellular transcriptional changes.[55]Key cytokines instruct lineage commitment by activating specific STAT proteins that promote master transcription factors. For Th1 differentiation, IL-12 primarily signals through JAK2/STAT4 to induce T-bet expression, while IFN-γ reinforces this via JAK1/STAT1, enhancing IFN-γ production and macrophage activation.[56] In Th2 polarization, IL-4 activates JAK1/JAK3/STAT6, driving GATA3 upregulation and IL-4/IL-5/IL-13 secretion for humoral immunity and anti-parasitic responses. Th17 cells arise under the influence of TGF-β combined with IL-6, where IL-6 engages JAK1/STAT3 to promote RORγt and IL-17 production, countering extracellular bacteria and fungi.[57] For regulatory T cells (Treg), TGF-β initiates Foxp3 expression, which is stabilized by IL-2 signaling through JAK1/JAK3/STAT5, maintaining immune tolerance.[58]Signal 3 cytokines do not act in isolation but integrate with TCR and costimulatory signals to fine-tune differentiation; for instance, TCR-induced pathways prime STAT responsiveness, allowing cytokines to selectively amplify lineage-specific transcription factors like T-bet or GATA3 without overriding initial activation.[59] This coordination prevents default fates and ensures robust effector function, as evidenced by impaired differentiation when signal 3 precedes or disrupts TCR engagement.[60]Tissue-specific environmental cues further shape signal 3 by varying local cytokine 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.[61]
Differentiation into subsets
Th1 and Th2 polarization
The classical Th1/Th2 dichotomy was first proposed in 1986 based on observations of distinct cytokine 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.[18] This model highlighted how these subsets drive divergent immune responses, with Th1 cells promoting cell-mediated immunity against intracellular pathogens and Th2 cells supporting humoral immunity, including antibody production and eosinophil activation often associated with allergic responses.Th1 polarization occurs when naive CD4+ T cells are exposed to IL-12, a cytokine produced by activated dendritic cells and macrophages during encounters with intracellular pathogens like Listeria monocytogenes.[62] IL-12 signaling activates STAT4, which induces expression of the transcription factor T-bet (encoded by Tbx21), the master regulator that commits cells to the Th1 lineage by directly promoting IFN-γ production and repressing Th2-associated genes.80702-3) T-bet further amplifies Th1 differentiation through positive feedback on IL-12 receptor expression, ensuring robust cell-mediated responses such as macrophage activation and cytotoxic T cell support.In contrast, Th2 polarization is driven by IL-4, often derived from innate sources like basophils or mast cells during parasitic infections, which activates STAT6 to upregulate GATA3, the key transcription factor for Th2 commitment. GATA3 binds to regulatory elements in the Il4, Il5, and Il13 loci, orchestrating cytokine production that fosters B cell class switching to IgE and recruitment of eosinophils, 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 proliferation and differentiation by downregulating IL-4 receptor expression and GATA3 activity, while IL-4 from Th2 cells suppresses Th1 development by reducing IL-12 responsiveness and T-bet induction.[63] 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 transcription factor 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 Candida albicans, by inducing antimicrobial peptides and neutrophil chemoattractants like CXCL1 and CXCL2.[64][65]In contrast, regulatory T (Treg) cells arise through pathways that emphasize immune suppression, with FOXP3 serving as the master transcription factor that defines their identity and suppressive function. Differentiation of induced Treg (iTreg) cells occurs in the periphery from naïve CD4+ T cells under the influence of TGF-β and IL-2, which stabilize FOXP3 expression and inhibit pro-inflammatory responses. Natural Treg (nTreg) cells, however, develop in the thymus from CD4+ single-positive thymocytes that encounter self-antigens with intermediate affinity, leading to FOXP3 upregulation and their export to peripheral tissues for ongoing immune tolerance.00199-X) The discovery of these FOXP3+ Treg cells and their role in maintaining peripheral immune tolerance was recognized by the 2025 Nobel Prize in Physiology or Medicine, awarded to Shimon Sakaguchi, Fred Ramsdell, and Mary Brunkow for their foundational contributions.[66]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 FOXP3, whereas shifting to TGF-β with IL-2 enhances FOXP3 expression and Treg differentiation, potentially converting Th17-like cells into suppressive Tregs.[67] 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 subset of CD4+ T helper cells that provide critical assistance to B cells within germinal centers of secondary lymphoid organs, characterized by expression of the chemokine receptorCXCR5 and driven by the transcription factorBcl6.[68]Bcl6 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 B cell proliferation, class switching, and affinity maturation.[69] These cells migrate to B cell follicles via CXCR5-mediated chemotaxis to CXCR5 ligand, ensuring localized support for humoral immunity.[70]Beyond classical subsets, Th9 cells emerge as IL-9-secreting effectors derived from naive CD4+ T cells under the influence of TGF-β and IL-4, contributing to allergic responses, anti-parasitic defense, and antitumor activity.[71] IL-9 production by Th9 cells is transcriptionally regulated by factors like IRF4 and PU.1, amplifying mucosal inflammation and mast cell activation without strong overlap with Th2 cytokine profiles.[72] Similarly, Th22 cells produce IL-22 in response to IL-6 and TNF-α, playing a key role in epithelial barrier protection against extracellular pathogens and maintaining mucosal homeostasis.[73]IL-22 from Th22 cells induces antimicrobial peptides and tight junction proteins in epithelial cells, fortifying defenses at skin and gut interfaces without promoting overt inflammation.[74]T helper cell plasticity refers to the dynamic reprogramming of lineage commitments, often mediated by epigenetic modifications that allow transdifferentiation between subsets in response to environmental cues.[75] For instance, Th17 cells can shift toward a Th1-like phenotype under IL-12 influence, acquiring IFN-γ production through chromatin remodeling at the Ifng locus, which is partially accessible in Th17 cells due to RORγt and T-bet interplay.[76] This plasticity is governed by histone acetylation and DNA methylation changes, enabling adaptive responses but also contributing to chronic inflammation in autoimmunity.[77]Recent studies from 2023 to 2025 have highlighted stem-like properties in CD4+ T cells, revealing intrinsic programs that confer adaptability and long-term potential beyond rigid subset definitions.[78] These stemness features, marked by expression of Tcf7 and self-renewal capacity, allow CD4+ T cells to maintain progenitor states for sustained effector differentiation in tumors and infections, redefining fate decisions through reversible epigenetic landscapes.[79] Such adaptations underscore the continuum of T helper cell responses, where environmental signals can redirect stem-like precursors to optimize immunity.[78]
Effector functions
Cytokine secretion profiles
T helper cells differentiate into distinct subsets, each characterized by unique cytokine secretion profiles that dictate their effector functions in immune responses. These profiles are established during polarization and enable targeted modulation of inflammation, antibody production, and immune suppression. The seminal identification of Th1 and Th2 subsets highlighted how differential cytokine patterns lead to contrasting roles in cell-mediated versus humoral immunity.[80]Th1 cells primarily secrete interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α), which promote macrophage activation, enhance antigen presentation, and drive cytotoxic responses against intracellular pathogens. In contrast, Th2 cells produce interleukin-4 (IL-4), IL-5, and IL-13, cytokines that stimulate B cell class switching to IgE, eosinophil activation, and goblet cell 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.[80][80]Th17 cells are defined by their production of IL-17A, IL-17F, and IL-22, which induce chemokine expression to recruit neutrophils, promote epithelial barrier integrity, and amplify inflammation at mucosal sites, particularly against extracellular bacteria 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 cytokine release, and maintain peripheral tolerance, preventing autoimmunity. These profiles distinguish Th17 from other subsets by their pro-inflammatory yet tissue-protective effects, while Treg cytokines enforce immunosuppression through direct receptor engagement on target cells.[81][82]The specificity of these cytokine secretion profiles is regulated by lineage-determining transcription factors that activate subset-specific promoters and enhancers in cytokine 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 STAT1, creating a positive feedback 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 IL-22 transcription, with IL-21 providing autocrine reinforcement, whereas Foxp3 in Treg cells represses pro-inflammatory loci while promoting IL-10 and TGF-β via Smad complexes. These mechanisms ensure stable, heritable cytokine patterns while allowing plasticity under chronic stimulation.[83][83]
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 B cells, which is essential for B cell activation and class-switch recombination in germinal centers. This ligation triggers B cell proliferation, survival, and differentiation into plasma cells capable of producing high-affinity antibodies of specific isotypes, such as IgG or IgA, thereby amplifying humoral immunity.[84]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 CD8+ T cells and DCs, Th cells upregulate costimulatory molecules on DCs, such as CD80 and CD86, which in turn bolster CD8+ T cell priming and effector function. Additionally, Th cells supply IL-2 during these encounters to sustain CD8+ T cell expansion and memory formation, ensuring robust cellular immunity against intracellular pathogens.[85][86]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.[87][88]These interactions are spatially orchestrated within secondary lymphoid tissues, such as lymph nodes and spleen, where Th cells localize to specific niches like the T-B border and germinal centers to optimize coordination. T follicular helper (Tfh) cells, a specialized Th subset, migrate into B cell follicles guided by chemokines like CXCL13, positioning them to interact sequentially with multiple B cells in a choreographed manner that sustains germinal center reactions. This architectural organization ensures efficient amplification of immune responses while minimizing off-target effects.[89]
Memory formation and maintenance
Generation of memory T helper cells
Upon activation, naïve T helper cells undergo asymmetric cell division, a process that unequally partitions cellular components between daughter cells, thereby generating one progenitor cell biased toward memory fate and another toward effector differentiation. 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 infection where polarity proteins like PKCζ and Numb are asymmetrically inherited to regulate fate decisions.[90][91]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 memory phenotype, 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 cytokine release upon re-encounter with antigen.[92][93]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 antigen. 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.[94][95]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.[96]
Long-term regulation and exhaustion
Memory CD4+ T cells maintain long-term homeostasis through interleukin-7 receptor (IL-7R) signaling, which is essential for their survival and slow homeostatic proliferation in the absence of antigen.[97] This cytokine is primarily produced by stromal cells in secondary lymphoid organs such as lymph nodes and spleen, as well as in the bone marrow, where mesenchymal stem cells serve as a key source supporting memory CD4+ T cell persistence.[98] 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 sepsis, bone marrow niches preferentially support IL-7-dependent proliferation of memory CD4+ T cells over naïve ones.[99] 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.[100]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.[101] 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 lymphocytic choriomeningitis virus (LCMV). PD-1 engagement with its ligand PD-L1 dampens T cell receptor signaling, reducing cytokine production and proliferation, while TIM-3 further exacerbates this by promoting apoptosis and metabolic impairments, leading to a hierarchical loss of functions in exhausted CD4+ T cells.[102] In human chronic infections, such as HIV or hepatitis C, co-expression of PD-1 and TIM-3 on CD4+ T cells correlates with impaired helper activity and increased viral persistence.[103]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 cytokine secretion.[104] In tumor microenvironments, anti-PD-1 antibodies have been shown to enhance the helper functions of exhausted PD-1high CD39+ CD4+ T cells, promoting cross-talk with CD8+ effectors and improving antitumor responses.[105] 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.[101]Recent studies have identified stem-like memory CD4+ T cells 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 antigen exposure and maintain a progenitor-like state that sustains long-term immunity without succumbing to inhibitory receptor upregulation.[78] For example, neoantigen-specific stem cell memory-like CD4+ T cells have demonstrated enhanced persistence and mediation of CD8+ T cell-dependent tumor control in solid tumors, resisting exhaustion through preserved metabolic fitness.[106] In 2024 research on tumor-infiltrating lymphocytes, stem-like CD4+ populations offered insights into bolstering durable responses via targeting these progenitors.[107] As of 2025, emerging evidence highlights metabolic factors, such as early methionine availability, in attenuating CD4+ T cell exhaustion during chronic stimulation, potentially enhancing memory maintenance and therapeutic outcomes in cancer and persistent infections.[108]
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 reactive oxygen species and nitric oxide, which restrict pathogen replication within phagosomes.30474-6)00118-7) For instance, in infections like tuberculosis or Listeria monocytogenes, Th1-derived IFN-γ is critical for macrophage-mediated clearance of intracellular bacteria.[109]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 eosinophils, which release cytotoxic granules containing major basic protein and peroxidase to damage helminth cuticles and facilitate parasite expulsion.[110][111] This mechanism is evident in infections with nematodes like Nippostrongylus brasiliensis, where Th2-driven eosinophilia 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 chemokine expression to recruit neutrophils. Neutrophils then release antimicrobial peptides and form extracellular traps to contain and eliminate pathogens like Candida albicans or Staphylococcus aureus.[65][112] 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 B cell activation and antibody production. Tfh cells provide CD40 ligand and cytokines like IL-21 to germinal center B cells, driving class-switch recombination, affinity maturation, and generation of high-affinity neutralizing antibodies.[113] This process underpins the efficacy of vaccines such as those against tetanus or influenza, where Th-mediated help ensures long-lasting humoral immunity.[114]
Involvement in autoimmunity and allergies
Dysregulated T helper cells play a central role in the pathogenesis of autoimmune diseases and allergies, where imbalances in their subsets lead to excessive inflammation and tissue damage. In autoimmunity, overactivation of pro-inflammatory subsets like Th17 cells promotes chronic inflammation, 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 eosinophilia and mucus hypersecretion. These dysregulations highlight the delicate balance required for immune homeostasis, with therapeutic strategies increasingly targeting specific cytokines or cell functions to mitigate disease progression.[115]Th17 cells contribute significantly to autoimmune diseases such as rheumatoid arthritis (RA) and psoriasis through the production of interleukin-17 (IL-17), which induces pro-inflammatory cascades. In RA, elevated Th17 cells and IL-17 levels in synovial fluid promote joint inflammation by recruiting neutrophils and enhancing matrix metalloproteinase expression, leading to cartilage destruction.[116] Similarly, in psoriasis, Th17-derived IL-17 stimulates keratinocyte proliferation and antimicrobial peptide production, perpetuating skin plaques and epidermal hyperplasia.[117] These effects are amplified by IL-23, which stabilizes Th17 differentiation and cytokine secretion, underscoring the IL-23/IL-17 axis as a key therapeutic target in these conditions.[118]In allergic diseases like asthma and atopic dermatitis, Th2 cells exacerbate pathology via IL-4 and IL-13 secretion, fostering IgE production and tissue remodeling. In asthma, Th2-mediated IL-4 promotes B-cell class switching to IgE, while IL-13 drives goblet cellmetaplasia and airway mucus hypersecretion, contributing to bronchial hyperresponsiveness.[119] In atopic dermatitis, these cytokines impair epidermal barrier function by downregulating filaggrin expression and inducing chemokine production that recruits Th2 cells and eosinophils, resulting in chronic skin inflammation.[120] Dual inhibition of IL-4 and IL-13 signaling has shown efficacy in reducing these Th2-driven responses in clinical settings.[121]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 type 1 diabetes (T1D) and inflammatory bowel disease (IBD). In T1D, impaired Treg suppressive function, particularly in pancreatic islets, allows unchecked Th1 and Th17 responses against beta cells, accelerating islet destruction.[122] Functional studies reveal that Tregs from T1D patients exhibit impaired proliferation and cytokine production, contributing to disease onset.[123] In IBD, particularly Crohn's disease, 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.[124] Restoring Treg function through ex vivo expansion has demonstrated potential to ameliorate colitis in preclinical models.[125]A striking example of Treg dysfunction is the immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, caused by mutations in the FOXP3 gene, which encodes the master regulator of Treg development and function. These loss-of-function mutations result in absent or non-functional Tregs, leading to multi-organ autoimmunity including severe enteropathy, endocrinopathies, and dermatitis from early infancy.[126] Patients exhibit elevated autoreactive antibodies and uncontrolled Th1/Th2/Th17 responses due to the lack of FOXP3-mediated suppression.[127]Hematopoietic stem cell transplantation remains the primary curative approach, highlighting the critical role of FOXP3 in preventing systemic autoimmunity.[128]
Immunodeficiency and viral infections
T helper cells are central to immune defense, and their depletion underlies severe immunodeficiencies, most notably in HIV infection. HIV primarily targets CD4+ 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 Pneumocystis pneumonia and tuberculosis, and certain cancers. As of 2025, antiretroviral therapy (ART) effectively restores CD4+ counts and immune function in most patients, preventing progression to AIDS, though chronic immune activation and exhaustion persist in some cases.[11]
Cancer and therapeutic targeting
T helper 1 (Th1) cells play a critical role in antitumor immunity by coordinating with cytotoxic CD8+ T cells to enhance tumor cell killing, primarily through the secretion of interferon-gamma (IFN-γ). Th1 cells produce IFN-γ, which activates CD8+ T cells, promotes their infiltration into the tumor microenvironment, and boosts their cytotoxic functions, including the release of perforin and granzyme B to directly lyse tumor cells.[78] This coordination is essential for effective antitumor responses, as IFN-γ also upregulates major histocompatibility complex class I expression on tumor cells, making them more susceptible to CD8+ T cell recognition and elimination.[129]In contrast, regulatory T cells (Tregs), a subset of CD4+ T helper cells, suppress antitumor immunity within the tumor microenvironment, 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 CD8+ T cell activation and proliferation.00042-9.pdf) High Treg infiltration in tumors correlates with poor prognosis across various cancers, as they create an immunosuppressive niche that hinders Th1 and CD8+ T cell-mediated killing.[130]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 cytokine 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.[131] Additionally, immune checkpoint 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 melanoma and lung carcinoma.[132]Recent advances in 2024-2025 clinical trials focus on Treg depletion to enhance immunotherapy, 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.[133] CCR8 antagonists, such as those in ongoing trials, further target Treg-specific markers to reduce suppression without broad immunosuppression, showing enhanced efficacy when combined with PD-1 inhibitors.[134] These approaches aim to shift the tumor microenvironment toward a Th1-dominant state, amplifying overall antitumor immunity.[135]