Fact-checked by Grok 2 weeks ago

Memory T cell

Memory T cells are a of antigen-experienced T lymphocytes that persist long-term after an initial or , providing rapid and enhanced protective immune responses upon re-exposure to the same . They originate from naive T cells that, upon by antigen-presenting cells, undergo clonal expansion and differentiate into effector T cells, a portion of which survive to form the memory pool. This process marks the cornerstone of adaptive immunity, enabling the to "remember" pathogens and mount secondary responses that are faster, stronger, and more efficient than primary ones, often clearing before symptoms arise. Memory T cells exhibit remarkable heterogeneity, classified into several subtypes based on their location, migratory patterns, and functional properties. Central memory T cells (T_CM) circulate through secondary lymphoid organs, express markers like CCR7 and CD62L, and possess high proliferative capacity to generate large numbers of effectors upon recall. In contrast, effector memory T cells (T_EM) patrol non-lymphoid peripheral tissues, lack CCR7, and rapidly produce effector cytokines such as IFN-γ and TNF-α for immediate activity. A specialized subset, tissue-resident memory T cells (T_RM), remains permanently stationed in barrier tissues like the skin, lungs, and mucosa, expressing retention markers such as and CD103 to provide localized, frontline defense without recirculation. These subtypes collectively ensure comprehensive surveillance and response across the body. The longevity of memory T cells is a defining feature, with human studies demonstrating persistence for decades without ongoing stimulation, maintained through slow homeostatic driven by cytokines like IL-7 and IL-15. Functionally, they not only accelerate clearance but also contribute to efficacy and antitumor immunity by sustaining robust recall responses. Recent insights highlight epigenetic modifications, such as stable patterns, as molecular hallmarks that underpin their differentiation and durability beyond surface markers.

Introduction and Fundamentals

Definition and Historical Discovery

Memory T cells are a subset of long-lived T lymphocytes that arise following the clearance of a primary or and persist in the host to confer long-term protective immunity. Unlike effector T cells, which are short-lived and mediate immediate responses, memory T cells enable accelerated and amplified secondary immune responses upon re-encounter with the same , characterized by rapid proliferation, production, and effector functions. This immunological memory is central to adaptive immunity, allowing for more effective control of pathogens without the need to restart the full primary response. Key characteristics of memory T cells include their capacity for self-renewal through homeostatic proliferation, which maintains their numbers over extended periods, and a reduced threshold compared to naïve T cells, facilitating quicker responses to lower doses due to altered signaling pathways such as enhanced TCR sensitivity and costimulatory requirements. They also express distinct surface markers, including the CD45RO isoform, which replaces CD45RA upon and correlates with antigen-experienced states, and IL-7Rα (CD127), which promotes and is upregulated on memory subsets to support their longevity. These features distinguish memory T cells from other T cell populations and underpin their role in sustained immunity. The historical discovery of memory T cells traces back to the , when foundational experiments on adoptive immunity in mice revealed the thymus's critical role in generating cells capable of transferring protective responses. In 1961, Jacques Miller demonstrated that neonatal in mice led to profound , identifying thymus-derived lymphocytes (T cells) as essential mediators of cellular immunity, including aspects of . Building on this, in the early 1970s, Miller and Jonathan Sprent conducted adoptive transfer studies in irradiated or thymectomized mice, showing that primed T cells from immune donors could confer secondary antibody responses and resistance to pathogens, providing direct evidence that T cells harbor immunological independent of B cells. A major milestone in the 1980s came with the development of and monoclonal antibodies, enabling the phenotypic identification of memory T cells. In 1988, Arne Akbar and colleagues reported that primed human T cells lose expression of the CD45RA isoform and acquire CD45RO (recognized by the UCHL1 antibody), marking a shift from naïve to memory states based on functional assays of antigen recall responses. These tools allowed researchers to isolate and characterize memory populations, solidifying their distinct identity and paving the way for deeper insights into their .

Differentiation from Naïve T Cells

The differentiation of naïve T cells into memory T cells begins with their activation, which requires recognition of antigenic peptides presented by (MHC) molecules on antigen-presenting cells (APCs), alongside co-stimulatory signals such as the interaction between on T cells and B7 ligands (/) on APCs, and support from the inflammatory milieu. This multi-signal integration triggers clonal expansion, where activated naïve T cells proliferate rapidly to generate a large pool of antigen-specific effectors. Without co-stimulation, activation can lead to anergy or , underscoring its essential role in committing naïve T cells to . Following initial activation, plays a critical role in generating heterogeneity among progeny, with early divisions producing daughter cells destined for effector or lineages. In this process, one daughter inherits factors promoting effector differentiation, while the other retains characteristics closer to the naïve state, facilitating precursor formation. precursor effector cells (MPECs) are distinguished by higher expression of the alpha chain (IL-7Rα, or CD127) and lower levels of killer cell lectin-like receptor G1 (KLRG1), markers that correlate with their potential to survive and form long-lived . In contrast, short-lived effector cells (SLECs) express low IL-7Rα and high KLRG1, biasing them toward terminal differentiation and eventual . Post-expansion, survival signals are pivotal for memory formation during the contraction phase, where the majority of effectors undergo . Upregulation of the anti-apoptotic protein in MPECs enhances their resistance to , allowing selective survival. Dependence on homeostatic cytokines IL-7 and IL-15 further supports MPEC persistence; IL-7 promotes survival and slow , while IL-15 drives self-renewal without . These signals operate after peak , ensuring that only a subset of cells transitions to stable . The differentiation process unfolds in distinct temporal stages following . The effector peaks around days 7-10, marked by maximal clonal expansion and production to control the . This is followed by the contraction , during which 90-95% of effectors die via , reshaping the response. Memory establishment then occurs over weeks to months, as surviving precursors mature into long-lived memory T cells capable of self-maintenance. Epigenetic modifications accompany these stages to lock in the memory .

Core Functions

Role in Secondary Immune Responses

Memory T cells play a pivotal role in secondary immune responses by enabling a swift and robust defense against previously encountered pathogens, contrasting sharply with the slower primary response mounted by naïve T cells. Upon re-exposure to antigen, memory T cells exhibit a lower activation threshold, requiring fewer antigen signals for initiation compared to naïve T cells, which demand higher doses and prolonged stimulation. This reduced threshold facilitates activation within hours—typically 3-6 hours for cytokine production—versus the days required for naïve T cell proliferation and differentiation. Consequently, memory T cells rapidly proliferate and differentiate into effectors, producing key molecules such as interferon-gamma (IFN-γ) and granzymes to mediate cytotoxicity and antiviral activity. The secondary response orchestrated by memory T cells surpasses the primary in both magnitude and quality, delivering amplified cytokine output and enhanced cytotoxic potential. For instance, memory CD8+ T cells secrete higher levels of IFN-γ and tumor necrosis factor-alpha (TNF-α), while also expressing perforin and more efficiently, leading to faster clearance. This heightened efficacy stems from pre-programmed transcriptional states that allow immediate effector function without the need for extensive reprogramming, resulting in a substantially larger response than the initial encounter. Recent studies have also highlighted the role of the integrated stress response pathway in regulating rapid cytokine production while preventing chronic activation in memory T cells. Memory T cells are strategically positioned to intercept reinfection through targeted to peripheral tissues, guided by receptors such as , which respond to ligands like CXCL9 and at infection sites. This anatomical distribution enables rapid infiltration and localized control of pathogens. Central memory T cells primarily recirculate through lymphoid organs for amplification, while effector memory T cells patrol non-lymphoid tissues for immediate action. Collectively, these cells provide long-term protection against viruses such as (CMV) and , sustaining immunity that reduces reinfection severity and duration in exposed individuals.

Homeostatic Proliferation and Maintenance

Memory T cells maintain their population size through antigen-independent homeostatic , primarily driven by the cytokines interleukin-7 (IL-7) and interleukin-15 (IL-15). These γ-chain cytokines promote slow and survival without triggering full activation, ensuring long-term persistence in the absence of reinfection. IL-7 signaling via the IL-7 receptor α (IL-7Rα, also known as CD127) is particularly essential for the survival of both central and effector memory T cells, while IL-15 supports , especially in CD8+ memory subsets, often through trans-presentation by accessory cells.00506-2) This process involves basal homeostatic turnover, with memory T cells undergoing intermittent division at a rate of approximately 1-2% per day in lymphoid tissues such as lymph nodes and . Unlike naïve T cells, this turnover is largely independent of (MHC) class I interactions with hematopoietic cells but can be influenced by MHC class I expression on non-hematopoietic stromal cells, which provide supportive signals in certain contexts. The slow proliferation rate balances cell loss due to , maintaining stable pool sizes over time.30888-7) Memory T cells occupy specific survival niches in the and secondary lymphoid organs, where they compete with naïve T cells for limited IL-7 and other growth factors produced by stromal cells. This competition limits the overall size of the memory compartment and ensures selective retention of antigen-experienced cells, with memory CD8+ T cells showing a in seeding due to enhanced expression. In aging individuals, homeostatic of memory T cells declines, contributing to impaired immune . This reduction stems from , which diminishes IL-7 production, alongside dysregulation that impairs responsiveness to IL-7 and IL-15. Consequently, memory T cell pools accumulate oligoclonally but with reduced proliferative capacity, exacerbating .

Longevity and Survival Mechanisms

Memory T cells achieve long-term persistence through the upregulation of anti-apoptotic proteins that protect against pathways, particularly Fas-mediated . Central and effector memory T cells express elevated levels of , which inhibits the pro-apoptotic protein Bim and enables these cells to tolerate higher expression of death signals during quiescence. Similarly, Mcl-1, another member, is essential for the survival and differentiation of memory CD8+ T cells, preventing during contraction phases post-infection. The inhibitor of apoptosis protein Birc3 (also known as cIAP2) further contributes by suppressing extrinsic death receptor signaling, collectively allowing memory T cells to resist Fas-induced elimination and maintain pool size over time. A key survival strategy involves metabolic reprogramming toward energy-efficient pathways that support quiescence and . Upon , memory T cells shift from glycolysis-dominant to reliance on oxidation (FAO) and (OXPHOS), which generate ATP with minimal production. This adaptation, facilitated by transcription factors like PPARβ/δ, enhances mitochondrial function and lipid utilization, enabling tissue-resident memory T cells to persist in nutrient-limited environments for extended periods. Telomere maintenance is critical in stem-like memory T cell subsets to avert replicative during repeated divisions. These cells exhibit sustained activity, which elongates and preserves proliferative capacity, distinguishing them from more differentiated populations with progressive telomere shortening. Mechanisms such as intercellular telomere transfer from antigen-presenting cells further rejuvenate telomere length in naïve and central T cells, promoting indefinite self-renewal without exhaustion. In humans, (CMV)-specific memory T cells demonstrate remarkable longevity, remaining detectable for over 50 years post-infection due to these integrated survival mechanisms. In models, antiviral memory T cells exhibit lifelong persistence, with subsets maintaining functionality throughout the animal's lifespan without re-exposure.

Developmental Pathways

Lineage Origins and Debate

The origins of the memory T cell lineage have been a subject of intense debate, primarily revolving around two competing models: the effector model, which proposes that memory T cells emerge from a subset of short-lived effector T cells that survive the contraction phase of the immune response, and the stem cell (or branched) model, which suggests that memory cells develop as a distinct lineage directly from activated naïve T cells, independent of full effector differentiation. The effector model gained early prominence through observations that memory cells share phenotypic and functional traits with effectors, implying a linear progression where only a fraction of effectors (approximately 5-10%) persist to form memory. In contrast, the stem cell model emphasized the existence of stem-like precursors that branch off early, avoiding terminal effector commitment, and was bolstered by evidence of transcriptionally distinct populations capable of self-renewal. This dichotomy persisted into the early 2000s, with studies highlighting the teleological appeal of the effector model but noting inconsistencies, such as the inability of terminally differentiated effectors to generate robust secondary responses. By the , advances in single-cell technologies and lineage tracing resolved the debate toward a hybrid model, integrating elements of both paradigms, where arise early during the initial activation and priming phase but can exhibit effector-like properties before fully committing to fate. Fate-mapping studies in mice demonstrated that , marked by high expression of the alpha chain (IL-7Rα^hi), emerge as early as the first few divisions of antigen-specific ^+ T cells, representing a small but proliferative subset (about 10-20%) that preferentially survives to seed the . These co-express stem-like factors while acquiring limited effector functions, supporting a branched yet flexible rather than strict linearity. Complementary human single-cell sequencing (scRNA-seq) data from the 2020s further corroborated this early , revealing transcriptional divergence between - and effector-destined ^+ T cells as soon as 3-4 days post-activation and models of viral stimulation. Central to lineage commitment are opposing transcription factors that tip the balance probabilistically: TCF-1 (encoded by Tcf7) promotes fate by sustaining stem-like self-renewal and central precursor formation, enabling robust secondary (up to 9-fold greater than in its absence), while Blimp-1 (encoded by ) drives terminal effector by repressing -associated genes and enhancing cytolytic programs. TCF-1 expression is maintained in early precursors to preserve multipotency, whereas Blimp-1 upregulation in response to strong signals enforces effector terminality, limiting potential. The current consensus views memory T cell differentiation as a , probabilistic process without a singular deterministic , where individual cells' fates are influenced by environmental cues such as dose and intensity during priming. Low doses and moderate favor memory precursor generation by sustaining TCF-1^hi populations, whereas high doses or strong inflammatory signals (e.g., via IL-12) promote Blimp-1 expression and effector , though states allow some even in chronic settings. This model underscores the heterogeneity in T cell responses, with implications for design aiming to skew toward memory-favoring conditions.

Precursors and Differentiation Stages

Memory precursor cells for CD8+ T cells emerge during the effector phase of the , characterized by low expression of the killer cell lectin-like receptor G1 (KLRG1^low) and high expression of the alpha chain (IL-7R^high), distinguishing them from short-lived effector cells (SLECs) that are KLRG1^high IL-7R^low. These memory precursor effector cells (MPECs) are poised for long-term survival and rapid reactivation upon secondary antigen encounter. For CD4+ T cells, memory precursors are identified under conditions that drive differentiation into T helper subsets such as Th1, Th2, or Th17, though their markers are less uniform than in + cells and often overlap with effector phenotypes during the response. The differentiation of memory T cells proceeds through distinct stages following activation. Stage 1 involves early precursors that arise shortly after stimulation and initial , marked by the onset of effector functions while retaining potential for memory commitment. In Stage 2, during the contraction phase after peak expansion, the majority of effectors undergo , but survivors transition toward memory phenotypes, enriched for MPEC markers in + cells. Stage 3 represents mature memory cells that achieve quiescence, characterized by long-term persistence, homeostatic , and enhanced functionality without ongoing stimulation. Recent advances from 2023 to 2025, leveraging single-cell sequencing, have refined + precursor identification, revealing transcriptional gradients and subset-specific markers like TCF7 and that predict memory potential across Th lineages. Certain memory T cell progenitors exhibit stem-like properties, particularly TCF-1+ (TCF7-encoded) cells that demonstrate self-renewal capacity and multipotency, allowing them to generate diverse effector and memory progeny while sustaining responses in chronic settings. These progenitors maintain a quiescent state with high proliferative potential, enabling replenishment of the memory pool over time. In the context of ongoing origin debates, such stem-like cells highlight a hierarchical model where early progenitors branch into committed memory subsets. Aging impairs memory precursor formation, with older individuals showing reduced generation of IL-7R^high due to dysregulated Wnt signaling, which disrupts TCF-1 expression and stem-like . This leads to diminished pool size and functionality, contributing to .

Regulatory Mechanisms

Epigenetic Modifications

Epigenetic modifications, including and alterations, are essential for locking in the memory T cell phenotype, enabling long-term survival, quiescence, and rapid reactivation upon re-encounter. During differentiation, patterns shift to demethylate loci associated with memory functions, such as the Sell encoding CD62L, which becomes significantly demethylated in central memory to facilitate lymphoid tissue homing and recirculation. Conversely, while effector like Ifng exhibit hypomethylation in resting memory T cells compared to naive cells—poising them for swift transcription—certain environmental factors can induce hyper at the Ifng promoter in memory T cells, impairing IFN-γ production and highlighting context-dependent regulation. These methylation dynamics ensure that memory T cells maintain a balanced epigenome, repressing unnecessary effector programs in the absence of while preserving accessibility for secondary responses. Histone modifications further refine this epigenetic landscape, with trimethylation of at 27 (H3K27me3) playing a key role in repressing proliferation-associated genes in memory T cells, thereby promoting quiescence and longevity over unchecked expansion. In parallel, acetylation at cytokine loci, such as those for IFN-γ and other effectors, creates a poised state in memory T cells, facilitating faster and more robust upon restimulation compared to naive cells. These active marks, including H3K9 acetylation, correlate with increased accessibility and differential profiles that distinguish memory from effector states. Key enzymes orchestrate these changes during T cell . TET proteins, particularly TET2, drive active at effector loci in response to proinflammatory signals like IL-12, enabling the transition to a memory-competent state with accessible promoters. Similarly, the EZH2, a core component of the Polycomb repressive complex 2, facilitates the effector-to-memory transition by depositing at specific sites to restrain terminal and promote memory precursor formation, with its activity modulated by to sustain T cell persistence. Dysregulation of EZH2 impairs secondary responses, underscoring its role in epigenetic fidelity. Recent studies have linked these epigenetic mechanisms to memory T cell longevity through the lens of epigenetic clocks, which track patterns as proxies for cellular age. In 2024 research, memory T cell epigenetic clocks were shown to advance independently of host chronological age, accumulating over multiple infection cycles and persisting through multiple adoptive transfers in a multilifetime experimental model, suggesting that immune history imprints durable epigenetic aging signatures that influence long-term functionality. These clocks provide insights into how epigenetic modifications not only establish memory but also govern its durability .

Transcriptional and Metabolic Regulation

Memory T cells are characterized by distinct transcriptional programs that maintain their identity and functionality post-infection. The TCF-1 (T-cell factor 1) plays a pivotal role in promoting memory ⁺ T cell differentiation and self-renewal by repressing effector genes and sustaining stem-like properties, enabling long-term persistence and rapid recall responses. Similarly, Eomesodermin (Eomes) is essential for memory maintenance, as it regulates prosurvival genes like and supports effector functions in ⁺ T cells through NFκB-Pim-1 signaling pathways. Runx3 contributes to residency by driving the expression of molecules and cytotoxic programs in memory ⁺ T cells, ensuring localized immune surveillance without recirculation. Recent advances highlight Lef1 (lymphoid enhancer-binding factor 1) as a key stemness factor, where its expression correlates with enhanced proliferative capacity and metabolic flexibility in memory precursors, fostering durable immunity. Metabolic shifts underpin the transition from effector to memory T cells, with effector cells relying heavily on for rapid energy production and biomass synthesis, whereas memory T cells favor (OXPHOS) and for sustained survival and quiescence. PGC-1α (peroxisome proliferator-activated receptor gamma coactivator 1-alpha) is a central regulator of this biogenesis, as its enforced expression enhances mitochondrial function, improves metabolic fitness, and boosts memory formation and antitumor efficacy in CD8⁺ T cells. A 2025 study further demonstrates that mitochondrial respiration, particularly complex III-derived , is crucial for supporting during expansion and optimal into long-lived memory cells, highlighting mitochondria's role beyond energy provision in memory establishment. Transcriptional and metabolic regulation are tightly integrated through crosstalk mechanisms that fine-tune memory T cell fate. For instance, inhibition of IDH2 (isocitrate dehydrogenase 2) enhances memory formation by elevating cytosolic citrate levels, which supports epigenetic modifications such as increased H3K27 . Feedback loops involving microRNAs further refine these processes; miR-146a negatively regulates inflammatory responses in memory T cells by targeting IRAK1 and TRAF6, thereby limiting excessive activation while preserving the memory pool and preventing exhaustion. These regulatory networks, which may involve epigenetic priming of metabolic genes, ensure adaptive responses to persistent antigens.

Subpopulations and Heterogeneity

Central and Effector Memory T Cells

Memory T cells are heterogeneous and can be broadly classified into central memory T cells (T_CM) and effector memory T cells (T_EM), which represent distinct circulating subpopulations with specialized roles in secondary immune responses. Both subsets express CD45RO, a marker of antigen-experienced T cells, but differ in their expression of homing receptors. T_CM cells are characterized by the CD45RO^+ CCR7^+ CD62L^+, enabling their preferential migration to secondary lymphoid organs such as lymph nodes. In contrast, T_EM cells exhibit a CD45RO^+ CCR7^- CD62L^- , which promotes their patrolling of peripheral non-lymphoid tissues. These distinctions were first delineated in CD4^+ and CD8^+ T cells based on chemokine receptor expression and functional assays. T_CM cells primarily reside in lymphoid tissues and possess high proliferative potential upon antigen re-encounter, allowing for rapid expansion into effector populations to sustain long-term immunity. They exhibit lower immediate effector functions but contribute to the generation of secondary effectors through robust and . T_EM cells, conversely, are poised for immediate effector responses, including rapid production (e.g., IFN-γ and TNF-α) and , providing frontline defense at sites of without needing to traffic through lymphoid organs. These functional differences position T_CM for orchestrating sustained responses and T_EM for acute, localized protection. The ratio of T_EM to T_CM varies depending on the type of , with such as (CMV) often favoring a T_EM-dominant pool (e.g., T_EM/T_CM ratio ≈1.2 in healthy donors). This skew may reflect ongoing antigenic pressure that promotes toward effector-like states. suggests between these subsets, with T_CM capable of converting to T_EM upon to inflamed peripheral tissues via signals like inflammation-induced gradients. Such interconversions highlight the dynamic nature of T cell , though T_EM cells show limited reversion to T_CM. Unlike tissue-resident T cells, which are anchored in specific organs, T_CM and T_EM circulate systemically to survey the body.

Tissue-Resident and Virtual Memory T Cells

Tissue-resident memory T cells (T_RM) represent a distinct of memory T cells that permanently reside in non-lymphoid tissues, such as the skin and mucosal barriers, without recirculating into the bloodstream or lymphoid organs. These cells are characterized by the expression of surface markers including and CD103, which promote their retention and survival in local microenvironments. Unlike circulating memory T cells, T_RM cells provide rapid, localized immune and effector responses upon re-encounter, enhancing frontline protection at barrier sites. The development and maintenance of T_RM cells are critically driven by transforming growth factor-β (TGF-β) signaling, which induces CD103 expression and inhibits tissue egress by downregulating sphingosine-1-phosphate receptor 1 (S1PR1). This cytokine-mediated process ensures long-term residency in epithelial and mucosal tissues, where T_RM cells can rapidly produce cytokines and cytotoxic molecules to control infections. In barrier tissues like the skin and intestines, TGF-β-responsive T_RM populations exhibit enhanced persistence, contributing to sustained immunity without reliance on circulating precursors. T_RM cells play a pivotal role in viral control, particularly against (), by mounting localized responses that limit reactivation and dissemination. For instance, in the genital mucosa, HSV-specific + T_RM cells suppress more effectively than circulating T cells, reducing lesion severity during recurrent infections. Similarly, skin-resident T_RM cells patrol epithelial layers to detect and eliminate HSV at entry sites, demonstrating superior protective efficacy compared to central or effector subsets. Virtual memory T cells constitute an antigen-inexperienced subset of memory-like CD8+ T cells marked by KLRG1 expression, arising through homeostatic proliferation rather than prior antigen encounter. These cells exhibit innate-like properties, rapidly responding to cytokines such as IL-15, which drives their differentiation from naïve precursors and supports their maintenance in lymphoid and non-lymphoid tissues. Virtual memory T cells contribute to immune homeostasis by filling niches during lymphopenia and providing bystander protection against infections without TCR specificity. A 2025 study revealed that virtual memory T cells emerge rapidly in humans following for (SCID-X1), bridging innate and adaptive immunity by generating antigen-naïve yet responsive effectors within months post-treatment. This innate-adaptive continuum highlights their potential in early-life immunity, where they compensate for delayed antigen-driven memory formation. In , virtual memory T cells maintain T cell pool stability through IL-15-dependent proliferation, preventing immune gaps in antigen-naïve states. Virtual memory T cells also exhibit anti-tumor potential through mechanisms akin to molecular , where their cytokine-driven activation mimics adaptive responses to cross-reactive epitopes, enhancing tumor surveillance. Recent research in 2024 demonstrated that FLT3L-induced CD8+ T cells infiltrate tumors and boost effector functions, improving anti-tumor immunity via homeostatic-like expansion that parallels -driven . This positions them as a bridge for innate-like anti-tumor responses, particularly in settings with limited exposure. T_RM and T cells display notable heterogeneity, particularly between + and + subsets, with + T_RM often showing stronger cytotoxic profiles and CD103 dependency in epithelial tissues, while + T_RM prioritize production and residency in mucosal sites. + T_RM cells exhibit greater transcriptional diversity across tissues, adapting to local cues like TGF-β for effector maturation, whereas + counterparts maintain more helper-oriented functions with variable marker expression. Aging impacts this residency, leading to reduced T_RM accumulation and functionality, with + subsets showing accelerated decline in proliferation and viral control capacity in barrier tissues. Studies indicate that age-associated shifts increase +/+ ratios in intestinal T_RM, impairing responses and exacerbating susceptibility to infections.

Activation Processes

Antigen-Dependent Activation

Memory T cells are activated in an antigen-dependent manner through engagement of their (TCR) with peptide-major histocompatibility complex (pMHC) on antigen-presenting cells, a that typically requires lower TCR affinity compared to naive T cells. This reduced affinity threshold enables memory T cells to respond effectively to lower antigen densities encountered during secondary infections. Upon TCR ligation, proximal signaling events occur rapidly, including of the tyrosine kinase ZAP-70, which is expressed at higher levels in memory T cells than in naive counterparts, thereby lowering the activation threshold and facilitating quicker signal propagation. Downstream of ZAP-70, memory T cells exhibit accelerated activation of key transcription factors, such as NFAT and AP-1, which drive the expression of effector genes including interleukin-2 (IL-2). In memory CD4+ T cells, NFAT activation is notably rapid, leading to IL-2 production within hours of stimulation, in contrast to the delayed response observed in naive T cells. This swift transcriptional response supports autocrine and essential for amplifying the recall reaction. Co-stimulatory signals further modulate this activation, with memory T cells capable of initiating responses independently of CD28 engagement, unlike naive T cells that strictly require it for full activation. However, signals from alternative co-stimulators such as and 4-1BB enhance proliferation, survival, and secretion in memory populations, promoting a more robust secondary response. The outcomes of antigen-dependent activation include rapid , differentiation into short-lived effector cells, and eventual contraction to maintain . Recall responses in memory T cells are kinetically superior, with initiating more quickly and achieving higher peak expansion—often 10- to 100-fold greater in magnitude due to increased precursor frequency—compared to primary responses. High-avidity clones, characterized by stronger TCR-pMHC interactions, preferentially dominate these secondary expansions, optimizing pathogen-specific immunity.00179-6)

Bystander and TCR-Independent Activation

Bystander activation of memory T cells refers to the stimulation of these cells without engagement of their T cell receptor (TCR), primarily driven by inflammatory cytokines released during infections. This process allows pre-existing memory T cell pools to rapidly respond to non-specific danger signals, enhancing early immune defense before antigen-specific clones expand. Key cytokines involved include interleukin-15 (IL-15), which promotes proliferation and effector functions in memory CD8+ T cells, and type I interferons (IFNs), which induce antiviral states and cytokine production independently of TCR signaling. For instance, during viral infections, IL-15 trans-presentation by antigen-presenting cells activates bystander memory CD8+ T cells, leading to granzyme B expression and cytotoxic potential without antigen recognition. Type I IFNs further amplify this by upregulating interferon-stimulated genes in memory T cells, facilitating quick deployment of innate-like responses. In CD4+ memory T cells, bystander activation often manifests as Th1-like responses, triggered by cytokines such as IL-12 and IFN-γ, though this phenomenon is less extensively studied compared to CD8+ counterparts. IL-12, produced by innate immune cells during viral encounters, directly stimulates memory CD4+ T cells to secrete IFN-γ, mimicking antigen-driven Th1 polarization and contributing to antiviral containment. This activation is particularly relevant in viral contexts, where bystander CD4+ T cells bridge innate and adaptive immunity by amplifying inflammation and recruiting other effectors, despite their lower prevalence in literature relative to CD8+ studies. For example, in respiratory virus infections, such responses enable rapid cytokine bursts that limit pathogen spread before specific immunity dominates. The prevalence of bystander activation varies by infection model but can represent a substantial fraction of the early T cell response, often up to 50% in acute challenges like lymphocytic choriomeningitis virus (LCMV) infection, where it drives initial proliferation of non-specific memory s. These responses are characteristically rapid, peaking within hours to days post-infection, but short-lived, typically subsiding as antigen-specific T cells take over and dominate the sustained phase. In contrast to TCR-dependent activation, bystander effects wane quickly due to the absence of persistent antigenic stimulation, limiting their role to immediate threat mitigation. Recent advances from 2023 to 2025 have highlighted the role of bystander activation in heterologous immunity, where prior T cells cross-respond to unrelated pathogens via cues, enhancing protection against novel infections. In vaccine contexts, this mechanism contributes to boosting, as non-specific T cells activated by adjuvants or viral vectors amplify overall immunity, observed in vaccination studies where bystander + responses bolster durability. These findings underscore bystander activation's potential in broadening beyond epitope-specific targeting.

Drivers and Consequences of Bystander Activation

Bystander activation of T cells is primarily driven by inflammatory , including IL-12 and IL-18, which stimulate CD8+ T cells to produce effector molecules like IFN-γ in the absence of TCR engagement. For + T cells, IL-1 serves as a key inducer, promoting rapid secretion without specificity. Alarmins such as IL-33 further contribute by enhancing bystander responses in tissue environments, while microbial products like (LPS) from trigger IL-12 and IL-18 production to initiate these -independent activations. This process relies on innate-like sensing mechanisms, allowing T cells to respond swiftly to inflammatory cues during infections. The consequences of bystander activation include amplification of non-specific inflammation, as bystander-activated memory T cells release proinflammatory cytokines like IFN-γ and TNF-α, exacerbating tissue damage in severe infections. On the positive side, it enables cross-protection against heterologous pathogens by providing rapid, non-antigen-specific immunity, as demonstrated by T cells controlling early bacterial loads. Experimental evidence from models highlights these dynamics; for instance, in co-infections, bystander-activated memory + T cells produce IFN-γ to limit bacterial dissemination independently of cognate antigen.00096-X) In human studies, particularly those examining mRNA booster vaccinations in 2025, bystander activation contributes to transient + T effector responses and modulates overall T cell dynamics, supporting hybrid immunity without evidence of exhaustion in repeated dosing.00678-3) Regulation of bystander activation is particularly constrained in chronic settings by PD-1 expression on memory T cells, which inhibits excessive production and prevents during persistent infections like LCMV.00876-3)

Clinical and Pathological Roles

Protective Immunity and

Live-attenuated vaccines, such as the , elicit robust vaccine-induced memory T cell responses that contribute to long-term protective immunity. The induces both central memory T cells (T_CM), which circulate systemically to coordinate secondary responses, and tissue-resident memory T cells (T_RM), which localize to mucosal sites like the lungs for rapid pathogen control. In measles vaccination, + and + T_RM cells persist in the lungs and liver, secreting cytokines such as IFN-γ and TNF-α upon restimulation, thereby restricting viral replication at entry points. T_CM and T_RM subsets play complementary roles in mucosal immunity, with T_RM providing frontline defense against respiratory pathogens by producing and perforin to limit initial infection, while T_CM sustain broader surveillance. The of vaccine-induced T cells emphasizes response quality—such as multifunctionality in cytokine —over mere quantity, as multifunctional T cells correlate with superior against reinfection. vaccination strategies can enhance this durability through bystander of preexisting T cells, independent of specificity, leading to heterologous immunity via cytokine-driven and IL-17A by CD4+ T_RM in mucosal tissues. For instance, intranasal whole-cell pertussis vaccination activates bystander CD4+ T_RM to confer cross-protection against unrelated like by reducing nasal bacterial burden. Challenges to protective immunity include waning responses in certain vaccines, such as acellular pertussis () vaccines, which fail to induce sufficient Th1/Th17 + T_RM cells in respiratory tissues, resulting in short-lived protection and increased colonization risk. Recent data from 2023–2025 on booster vaccinations, particularly for , demonstrate that additional doses reactivate stable + and + memory T cell populations without inducing exhaustion, maintaining polyclonal responses and enhancing durability through diverse T cell subsets. Memory T cells are essential for efforts to control endemic diseases like (TB), where lung-resident T_RM cells provide rapid IFN-γ-mediated control of replication, potentially reducing the global burden of 10.8 million new cases in 2023. Targeting T_RM induction in TB vaccines could improve efficacy beyond the variable protection offered by BCG, supporting sustained immunity in high-prevalence regions.

Involvement in Autoimmunity and Chronic Disease

Memory T cells play a detrimental role in by perpetuating self-reactive responses that contribute to disease relapses and progression. Self-reactive memory T cells, particularly effector memory subsets, maintain heightened reactivity to autoantigens, leading to recurrent inflammation in tissues such as the . In (MS), tissue-resident memory T (T_RM) cells infiltrate the brain parenchyma, where they exhibit signs of reactivation and promote chronic by sustaining local inflammation and formation. This persistence is exemplified by CD8+ T_RM cells in progressive MS lesions, which invade and correlate with disease severity. Furthermore, spreading amplifies as initial antigen-specific responses diversify to encompass additional self-epitopes, broadening the autoreactive T cell repertoire and driving disease chronicity, as observed in models of nonobese diabetic mice and human MS cohorts. In infections, memory T cells often succumb to exhaustion, impairing control while fostering . During persistent infections like and (HCV), antigen-specific + memory T cells progressively lose effector functions, upregulate inhibitory receptors such as PD-1, and exhibit altered transcriptional programs that limit proliferation and cytokine production. This exhaustion persists even after clearance in HCV, with epigenetic modifications in exhausted cells hindering full functional restoration. Virtual memory T cells, a subset of antigen-inexperienced cells with memory-like properties, can exacerbate in settings by mediating bystander activation and promoting excessive without specific recognition, as seen in models of persistence and autoimmune-like damage. Aging exacerbates the pathological potential of memory T cells through , a state of chronic low-grade driven by accumulated viral-specific memory pools. ()-specific memory T cells undergo memory inflation in older individuals, comprising up to 50% of the + memory compartment and secreting pro-inflammatory cytokines that contribute to and age-related comorbidities. This process is linked to T cell senescence markers, including shortened telomeres and increased PD-1 expression, which amplify . Recent 2023 studies highlight age-biased fate decisions in naïve T cells, where epigenetic adaptations favor differentiation into short-lived effector cells over long-lived memory cells, further skewing the toward and reducing adaptive immunity in the elderly. Therapeutic strategies targeting memory T cells aim to mitigate their role in autoimmunity by depleting pathogenic subsets. Alemtuzumab, a against , effectively depletes circulating memory T cells in relapsing-remitting , leading to a repopulation dominated by naïve and regulatory T cells that suppresses autoreactive responses and promotes long-term remission. However, this depletion can induce homeostatic of residual autoreactive clones, occasionally triggering secondary , underscoring the need for balanced immune reconstitution. Such approaches highlight the dual-edged nature of memory T cell targeting in autoimmune diseases.

Applications in Cancer Immunotherapy

Memory T cells play a crucial role in by providing long-term surveillance and rapid response against tumor antigens. Tumor-specific memory T cells can arise through molecular , where T cells primed by viral infections cross-react with structurally similar tumor antigens, enabling pre-existing immunity to contribute to anti-tumor responses. This mechanism has been observed in various cancers, highlighting how prior exposures may enhance endogenous memory T cell-mediated tumor control. Additionally, CD8+ tissue-resident memory T (T_RM) cells accumulate in tumors and correlate with improved patient survival by directly eliminating malignant cells and shaping the to favor anti-tumor immunity. Chimeric antigen receptor (CAR) T cell therapies leverage memory T cell properties to achieve durable remissions in hematologic malignancies. In CD19-targeted CAR-T treatments for B-cell leukemia, engineering CAR-T cells to adopt a memory-like phenotype enhances their persistence and expansion in vivo, reducing relapse rates compared to effector-dominant populations. For instance, clinical data from trials using less-differentiated CAR-T cells show prolonged circulation and better control of minimal residual disease in acute lymphoblastic leukemia patients. However, in solid tumors, challenges such as antigen heterogeneity, immunosuppressive microenvironments, and poor infiltration limit efficacy; advances in universal CAR-T platforms, which allow targeting of multiple antigens via adaptable receptors, aim to address these issues and improve outcomes in epithelial cancers. Stem-like memory T (T_SCM) cells, a CD8+ subset characterized by high self-renewal and multipotency, are particularly promising for CAR-T engineering due to their superior engraftment and long-term repopulation potential. When used as the starting population for CAR-T , T_SCM-enriched products demonstrate enhanced anti-tumor activity and reduced exhaustion in preclinical models. Clinical trials incorporating T_SCM-like CAR-T cells have reported improved relapse-free survival in B-cell malignancies, with one study showing over 50% of patients achieving durable remissions beyond two years post-infusion. Future directions in memory T cell-based include CAR delivery systems, which use viral vectors or nanoparticles to transduce T cells directly within the body, bypassing manufacturing challenges and potentially accelerating treatment for solid tumors. Combining these approaches with checkpoint inhibitors, such as anti-PD-1 antibodies, further boosts memory T cell function by alleviating exhaustion and promoting infiltration, as evidenced in ongoing trials yielding synergistic tumor regression.

References

  1. [1]
    Defining the Molecular Hallmarks of T-Cell Memory - PubMed Central
    The first defining characteristic of memory T cells is that upon reencounter with antigen, the cells recall effector function faster than the first time the ...
  2. [2]
    Human memory T cells: generation, compartmentalization and ...
    The acquisition of antigen experience is marked by the generation and persistence of memory T cells which can provide life-long protection against pathogens.
  3. [3]
    Biochemical Signaling Pathways for Memory T cell Recall - PMC
    Memory T cells exhibit low activation thresholds and rapid effector responses following antigen stimulation, contrasting naive T cells with high activation ...
  4. [4]
  5. [5]
    Recent insights of T cell receptor-mediated signaling pathways for T ...
    May 21, 2020 · Naive T cells are initially activated through their TCRs by antigen/MHC complexes expressed by antigen-presenting cells. Subsequent signals, ...
  6. [6]
    Effector and memory T-cell differentiation: implications for vaccine ...
    Apr 1, 2002 · CD8+ T cells have a lower activation requirement, which permits the threshold for activation to be passed more rapidly than for CD4+ T cells.
  7. [7]
    Generation of effector CD8+ T cells and their conversion to memory ...
    Typically, the number of memory CD8+ T cells remains quite stable overtime, largely through interactions with interleukin-7 (IL-7) and IL-15 that mediate ...
  8. [8]
    Asymmetric cell division shapes naive and virtual memory T ... - Nature
    May 11, 2021 · Upon ACD, a single T cell can give rise to daughter cells inheriting distinct contents of fate determinants through the polarized segregation of ...
  9. [9]
    Asymmetric T-cell division: insights from cutting-edge experimental ...
    Mar 1, 2024 · As discussed below, naïve CD8+ T cells divide and develop into short-lived effector cells (SLECs) that are important for immunological ...
  10. [10]
    Inflammation Directs Memory Precursor and Short-Lived Effector ...
    As acute infections resolve, effector CD8 T cells differentiate into IL-7Rlo short-lived effector cells (SLECs) and IL-7Rhi memory precursor effector cells ...
  11. [11]
    Molecular regulation of effector and memory T cell differentiation
    Moreover, the IL-7Rhi subset seems to retain plasticity, while the KLRG1hi subset does not, because the former 'memory precursor' subset can also give rise to T ...
  12. [12]
    The role of mTOR in memory CD8+ T-cell differentiation - PMC
    1D). This progressive improvement of memory T-cell qualities occurs along with many phenotypic changes including upregulation of Bcl2 (anti-apoptotic) (24), re- ...
  13. [13]
    XIAP promotes the expansion and limits the contraction of CD8 T ...
    Interestingly, the CD8 T cell response observed at the peak of response (day 7) was significantly reduced in Xiap−/− mice as detected by OVA-dextramer staining ...
  14. [14]
    Differential expansion of T central memory precursor and effector ...
    Jan 8, 2020 · During the contraction phase 90–95% of all T cells present at the peak of expansion die, leaving mainly memory T cells that persist long term1, ...Missing: timeline | Show results with:timeline
  15. [15]
    Transcriptional control of effector and memory CD8+ T cell ...
    This paper illustrates that memory and effector T cell fates can arise from a single precursor T cell through asymmetric cell division. 27. Mousavi SF, et ...
  16. [16]
  17. [17]
  18. [18]
    MECHANISMS OF MEMORY T CELL ACTIVATION AND ...
    Reactivated memory T cells are also shown to secrete important amounts of proinflammatory cytokines and chemokines such as IFNγ, CCL3, CCL4 and CCL5, which ...
  19. [19]
  20. [20]
    Cellular immune correlates of protection against symptomatic ...
    Sep 22, 2013 · The role of T cells in mediating heterosubtypic protection against natural influenza illness in humans is uncertain. The 2009 H1N1 pandemic ...
  21. [21]
    Tissue reservoirs of antiviral T cell immunity in persistent human ...
    Jan 27, 2017 · Overall, our results reveal how CMV-specific T cells are distributed and function in multiple sites in the context of tissue location and viral ...
  22. [22]
    15 and IL-7 Jointly Regulate Homeostatic Proliferation of Memory ...
    The periodic turnover of memory T cells is MHC independent, at least for CD8+ cells (14), and occurs without any significant change in the total size of the ...Adoptive Transfer Of T Cells · Role Of Mhc In Homeostatic... · Role Of γ Cytokines In...
  23. [23]
    Memory Phenotype CD4 T Cells Undergoing Rapid, Nonburst-Like ...
    Indeed, both the work presented here and recent studies analyzing antigen-specific CD4 memory T cells ... 1% to ∼2.5% per day. The possibility that MP cells and ...
  24. [24]
    Ageing and life-long maintenance of T-cell subsets in the ... - Nature
    In summary, ageing is associated with a dramatic increase in the proportion of memory T cells in the blood, with a concomitant and equivalent decrease in the ...
  25. [25]
    Bcl-2 Allows Effector and Memory CD8+ T Cells To Tolerate Higher ...
    We found that survival of both subsets of effector and memory CD8 + T cells required Bcl-2 to combat the proapoptotic activity of Bim.
  26. [26]
    Mcl-1 Regulates Effector and Memory CD8 T-Cell Differentiation ...
    Mcl-1, an anti-apoptotic member of Bcl-2 family maintains cell viability during clonal expansion of CD8 T cells, but the cell intrinsic role of Mcl-1 in ...Missing: upregulation Birc3
  27. [27]
    Specific elimination of effector memory CD4 + T cells due to ... - Nature
    Dec 17, 2010 · The differential sensitivity of CD4+ T-cell subsets to Fas-induced apoptosis may be regulated by cell-intrinsic survival factors, such as bcl-2 ...
  28. [28]
    PPARβ/δ-orchestrated metabolic reprogramming supports the ...
    Aug 23, 2024 · Our findings indicate that PPARβ/δ is a master metabolic regulator orchestrating a metabolic switch that may be favorable for T cell longevity.
  29. [29]
    Metabolic Reprogramming and Longevity of Tissue-Resident ...
    Jun 18, 2018 · T RM upregulate expression of genes associated with lipid uptake and metabolism and utilize mitochondria fatty acid β-oxidation to support their long-term ...
  30. [30]
    PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis ...
    Mar 26, 2015 · ... T cells are unable to engage in glycolysis or amino acid metabolism but have an increased rate of fatty acid β-oxidation (FAO). PD-1 ...
  31. [31]
    Telomerase Activation and Rejuvenation of Telomere Length in ...
    Together, these results imply that one role for telomerase in T cells may be to renew or extend replicative potential via the rejuvenation of telomere length.
  32. [32]
    New mechanism to promote long-term T-cell immunity by telomere ...
    Nov 18, 2022 · Maintaining telomere length is important for preventing T cells from undergoing senescence [1]. Upon antigen stimulation, naive T cells ...
  33. [33]
    An intercellular transfer of telomeres rescues T cells from ...
    The common view is that T-lymphocytes activate telomerase to delay senescence. Here we show that some T cells (primarily naïve and central memory cells) ...
  34. [34]
    Memories that last: Dynamics of memory T cells throughout the body
    Apr 28, 2023 · Memory T cells form an essential part of immunological memory, which can last for years or even a lifetime. Much experimental work has shown ...INTRODUCTION · DYNAMICS OF CIRCULATING... · DYNAMICS OF MEMORY T...
  35. [35]
    Generation, persistence and plasticity of CD4 T-cell memories - PMC
    Once generated, memory CD4 T cells can persist for up to the lifetime of an individual via long-term homeostasis and turnover. In humans, anti-viral memory CD4 ...Missing: lifelong | Show results with:lifelong
  36. [36]
    Use it or lose it: establishment and persistence of T cell memory
    Moreover, it appears that once the memory T cells are established early in life, they can persist for the lifetime of an individual. This is in contrast to the ...Missing: lifelong | Show results with:lifelong
  37. [37]
    Memory CD8 T-Cell Differentiation during Viral Infection
    Jun 1, 2004 · During a CD8 T-cell response to infection, there are three characteristic phases: a period of initial activation and expansion, ...<|separator|>
  38. [38]
    Transcriptional and Epigenetic Regulation of Effector and Memory ...
    In this review, we will summarize the current understanding of CD8 T cell differentiation upon acute infection, with a focus on the transcriptional and ...Missing: 2020s | Show results with:2020s
  39. [39]
    Beyond T-cell subsets: stemness and adaptation redefining ... - Nature
    Jul 10, 2025 · While naïve T cells serve as the “ancestors” of all effector and memory subsets, T-cell stemness pertains to antigen-experienced T cells that ...Stem-Like Cd4 T Cells And... · Tumor Control And Immune... · In Peripheral Immune...
  40. [40]
    The role of TCF-1 + CD8 + exhausted progenitors and TCF-1 in graft ...
    Oct 8, 2025 · Upon competitive retransplantation, TetH60+CD39loTCF-1hi cells outcompeted TetH60+CD39hiTCF-1lo cells and underwent self-renewal, whereas CD39hi ...
  41. [41]
    'Stem-like' precursors are the fount to sustain persistent CD8 + T cell ...
    May 27, 2022 · A failure to generate proliferation-competent precursor cells in chronic infections and tumors results in the collapse of the T cell response.Missing: lifelong | Show results with:lifelong
  42. [42]
    T cell fate decisions during memory cell generation with aging
    Aug 7, 2025 · TCF7, whose gene product is TCF1, functions via the Wnt signaling ... Metabolic Signaling as a Driver of T Cell Aging. Article. Full-text ...
  43. [43]
    Human memory CD8 T cell effector potential is epigenetically ...
    May 10, 2017 · Among the DMRs that delineate the TEM, TCM, and TSCM CD8 T cells were subset-associated DMRs at CpG sites in the CCR7 and CD62L (SELL) loci.
  44. [44]
    DNA Methylation and Immune Memory Response - PMC
    Progesterone impairs the production of IFN-γ by memory CD8 T cells, inducing hypermethylation in the Ifng gene promoter region. This mechanism is suggested ...
  45. [45]
    Deciphering the role of histone modifications in memory ... - bioRxiv
    Apr 22, 2025 · ... TN are actively repressed in TMEM. In contrast, genes that became repressed during TEX differentiation gained repressive H3K27me3 at the ...
  46. [46]
    Histone acetylation is associated with differential gene expression in ...
    Nov 15, 2006 · We examined gene expression and chromatin modification by histone H3 lysine 9 (H3K9) acetylation in resting and activated human naive and memory CD8 + T cells.Gene Expression · Results · Increased H3k9 Acetylation...
  47. [47]
    Ezh2 phosphorylation state determines its capacity to maintain CD8 ...
    Dec 14, 2017 · Ezh2 regulates effector and memory T-cell persistence. To evaluate the impact of Ezh2 in CD8+ T-cell responses, we used an experimental ACT ...
  48. [48]
    Conserved epigenetic hallmarks of T cell aging during immunity and ...
    Jun 12, 2024 · Here we found that memory T cell epigenetic clocks tick independently of host age and continue through four lifetimes.
  49. [49]
    Role of TCF-1 in differentiation, exhaustion, and memory of CD8+ T ...
    TCF-1 can serve as a potential target of immunotherapy and may provide promising novel treatment strategies for patients with cancer and infections. Moreover, ...
  50. [50]
    NFκB-Pim-1-Eomesodermin axis is critical for maintaining CD8 T ...
    Feb 28, 2017 · We also found that NFκB-Pim-1 signals were required at memory to maintain memory CD8 T-cell longevity, effector function, and Eomes expression.
  51. [51]
    Runx3 programs CD8+ T cell residency in non-lymphoid tissues and ...
    Dec 14, 2017 · Tissue-resident memory CD8 + T (T RM ) cells are found at common sites of pathogen exposure, where they elicit rapid and robust protective ...
  52. [52]
    The degree of T cell stemness differentially impacts the potency of ...
    Mar 10, 2023 · We find that the level of stemness T cells possess in vitro differentially impacts their potency upon transfer in three tumor models.
  53. [53]
    The role of metabolic reprogramming in T cell fate and function - PMC
    Mar 27, 2017 · In this review, we describe the basic metabolic features of T cells and new findings on how these correlate with various differentiation fates and functions.
  54. [54]
    Enforced PGC-1α expression promotes CD8 T cell fitness, memory ...
    Feb 13, 2020 · Specifically, memory CD8 T cells engage oxidative phosphorylation and fatty acid oxidation to fulfill their metabolic demands. In contrast ...
  55. [55]
    Mitochondrial respiration is necessary for CD8+ T cell proliferation ...
    Jul 16, 2025 · By contrast, mitochondrial complex III ROS are necessary for optimal memory CD8+ T cell differentiation and persistence. Thus, T cell ...
  56. [56]
    Mitochondrial isocitrate dehydrogenase impedes CAR T cell ...
    Jan 2, 2024 · IDH2 limits the cytosolic citrate level to impede memory CAR T cell formation through epigenetic regulation. IDH isoenzymes mainly catalyze ...
  57. [57]
    MiR-146a and NF-κB1 regulate mast cell survival and T lymphocyte ...
    We show that in mast cells it contributes to the regulation of cell homeostasis and survival, while in T lymphocytes it modulates T cell memory formation.
  58. [58]
    Central memory self/tumor-reactive CD8+ T cells confer superior ...
    Jun 24, 2005 · Central memory CD8+ T cells (TCM) and effector memory CD8+ T cells (TEM) are found in humans and mice; however, their relative contributions ...Sign Up For Pnas Alerts · Materials And Methods · Results<|control11|><|separator|>
  59. [59]
    Ratios of effector to central memory antigen-specific CD4(+) T cells ...
    Feb 4, 2014 · CMV responses in healthy donors showed a Tem-dominant response with a Tem/Tcm ratio of 1.2, whereas the tetanus toxoid responses were dominated ...
  60. [60]
    Tissue-Resident Memory T Cells - PMC - PubMed Central
    Tissue-resident memory T (Trm) cells constitute a recently identified lymphocyte lineage that occupies tissues without recirculating.Missing: seminal | Show results with:seminal
  61. [61]
    Tissue resident memory T cells in the respiratory tract - Nature
    Oct 20, 2021 · Although the stable long-term persistence of Trm has been documented in a variety of tissues including the nose, skin, liver, and intestinal ...Memory T-Cell Subsets · Ii) Local Cytokine Milieu · Respiratory Viral InfectionsMissing: lifelong | Show results with:lifelong<|control11|><|separator|>
  62. [62]
    TGF-β: Many Paths to CD103+ CD8 T Cell Residency - PMC
    This review focuses on the diverse role of TGF-β in the development and maintenance of CD103 + CD8 T RM cells by skewing the population towards a memory ...Missing: seminal papers
  63. [63]
    T-box Transcription Factors Combine with the Cytokines TGF-β and ...
    Transforming growth factor-β signaling controls the formation and maintenance of gut-resident memory T cells by regulating migration and retention.<|separator|>
  64. [64]
    The Role of Tissue Resident Memory CD4 T Cells in Herpes ...
    Upon HSV-2 re-infection, mice relying on only circulating CD4+ memory T cells could not fully suppress viral replication, whereas mice harbouring HSV-2 ...
  65. [65]
    Tissue-resident memory CD8+ T cells continuously patrol skin ...
    Nevertheless, tissue-resident memory T cells provide superior protection against local virus reactivation relative to the circulating memory T-cell pool (3, 6, ...
  66. [66]
    Tissue resident memory T cells and viral immunity - PubMed Central
    Dec 14, 2016 · Tissue resident memory T cells (T RM ) constitute a recently identified T cell lineage that is responsible for frontline defense against viral infections.Tissue Resident Memory T... · Introduction · Figure 3. Cd8+ T Functions
  67. [67]
    Control of memory CD8+ T cell longevity and effector functions by IL ...
    Finally, IL-15 is sufficient to convert naïve CD8+ T cells into “virtual” memory cells and this unique subset of memory CD8+ T cells is absent in IL-15−/− mice ...Missing: papers | Show results with:papers
  68. [68]
    Virtual memory T cells develop and mediate bystander protective ...
    Apr 21, 2016 · Previous studies indicated that VM cells were the result of homeostatic proliferation (HP) resembling the proliferation observed in a ...
  69. [69]
    Virtual memory CD8 T cells display unique functional properties
    Considerable evidence suggests this population, termed “virtual memory” (VM) CD8 T cells, arise via physiological homeostatic mechanisms. However, the antigen- ...
  70. [70]
    Innate-like memory T cells rapidly emerge in humans after gene ...
    Jul 31, 2025 · Here, we describe the genesis of virtual memory T cells during early human life. A longitudinal analysis of peripheral T cells after gene ...
  71. [71]
    FLT3L-induced virtual memory CD8 T cells engage the immune ...
    Jan 29, 2024 · T cells with memory phenotype are equipped with superior antitumor functions and proliferative ability compared to effector T cells [2, 3]. Many ...
  72. [72]
    Molecular mimicry as a driver of T cell-mediated tumour immunity
    Aug 15, 2025 · ... mimicry between viral epitopes and TAAs may possibly elicit crossreactive memory T cells with the potential to contribute to anticancer immunity ...Missing: virtual | Show results with:virtual
  73. [73]
    Heterogeneity and plasticity of tissue-resident memory T cells in skin ...
    May 7, 2024 · CD4+ Trm or CD8+ Trm cells play a harmful role in autoimmune related skin diseases such as psoriasis and vitiligo, while CD8+Trm cells play a ...<|control11|><|separator|>
  74. [74]
    The Multifaceted Role of Tissue-Resident Memory T Cells
    Jun 28, 2024 · Of these, tissue-resident memory T (TRM) cells are strategically positioned in barrier tissues, where they enable efficient frontline defense ...
  75. [75]
    Asynchronous aging and turnover of human circulating and tissue ...
    Memory T cells across peripheral sites exhibited continuous turnover with mean lifespans of 1–2 years, while the spleen contained longer-lived T cells. Over age ...Missing: rate | Show results with:rate
  76. [76]
    Age-related decline in CD8+ tissue resident memory T cells ...
    Nov 26, 2024 · We report that aging impairs the generation of CD8 + tissue resident memory T (T RM ) cells in nonlymphoid tissues in mice, thus compromising the antitumor ...
  77. [77]
  78. [78]
  79. [79]
  80. [80]
    The CD28-Related Molecule ICOS Is Required for Effective T Cell ...
    Taken together, these data suggest that the primary response is largely CD28 dependent, and the secondary response is CD28 independent and at least in part ICOS ...
  81. [81]
    Costimulation of human CD28– T cells by 4‐1BB ligand
    Jan 27, 2003 · ... CD28-independent pathways. Here, we ... The accumulation of CD28– memory T cells in humans may be due to their mode of prior activation.2 Results · 3 Discussion · 4 Materials And Methods
  82. [82]
    Functional differences between memory and naive CD8 T cells - PNAS
    On antigen stimulation in vivo, however, they proliferated more rapidly than naive cells. These findings suggest that, unlike naive T cells, CD8 memory T cells ...
  83. [83]
    Memory T cells: strategies for optimizing tumor immunotherapy
    Mar 27, 2020 · First, memory T cells have lower activation thresholds than naïve T cells: these cells can respond to 100-fold lower doses of antigen and ...
  84. [84]
    The activation of bystander CD8 + T cells and their roles in viral ...
    Dec 11, 2019 · ” Cytokines, including type I interferons, interleukin-18, and interleukin-15, are the most important factors that induce bystander ...
  85. [85]
    IL-15 in T-Cell Responses and Immunopathogenesis - PMC - NIH
    Feb 16, 2024 · During viral infection, the upregulation of IL-15 triggers bystander activation of memory CD8+ T cells (8,127). IL-15 induces polyclonal ...
  86. [86]
    Mechanisms governing bystander activation of T cells - Frontiers
    Under steady-state conditions, naive CD8 T cells can also acquire a memory-like phenotype in the presence of IL-15 and the transcription factor EOMES (75–77).
  87. [87]
    The activation of bystander CD8 + T cells and their roles in viral ...
    Dec 11, 2019 · Heterologous immune responses can include both antigen-dependent T cell activation by cross-reactive memory T cells and antigen-independent ...
  88. [88]
    Bystander activation of CD4 + T cells - Wiley Online Library
    Apr 14, 2010 · Furthermore, three other cytokines, namely IFN-γ, IL-12 and IL-18, led to bystander activation of MP CD8+ T cells; the bystander activation ...
  89. [89]
    Bystander CD4+ T cells: crossroads between innate and adaptive ...
    Aug 28, 2020 · Effector/memory CD4+ T cells (TH1, TH2, and TH17) can undergo bystander activation by directly responding to inflammatory cytokines and TLR ...
  90. [90]
    Pathogenic function of bystander-activated memory-like CD4+ T ...
    Feb 12, 2019 · Our findings suggest that antigen non-related memory-like CD4+ T cells are an important source of IL-17A and IFN-γ in inflammatory tissues in ...
  91. [91]
    Regulation of human and mouse bystander T cell activation ...
    Sep 22, 2023 · Bystander activation of memory T cells occurs via cytokine signaling alone in the absence of T cell receptor (TCR) signaling and provides a ...
  92. [92]
    Heterologous immunity and antibody-dependent enhancement in ...
    Oct 29, 2025 · Heterologous immunity arises when memory T or B cells generated during infection with one virus cross-recognize and respond to antigens from ...
  93. [93]
    Role of antiviral CD8+ T cell immunity to SARS-CoV-2 infection and ...
    Mar 3, 2025 · Low-dose mRNA-1273 COVID-19 vaccine generates durable memory enhanced by cross-reactive T cells. ... bystander-activated memory CD8 T cells. J ...
  94. [94]
    Evolution of SARS-CoV-2 T cell responses as a function of multiple ...
    Jul 22, 2025 · We investigate the long-term impact of repeated COVID-19 vaccinations on adaptive immunity through a 3-year study of 78 individuals without reported ...
  95. [95]
    Regulation of innate CD8+ T-cell activation mediated by cytokines
    We found that IL-2 was able to synergistically enhance IFNγ production by virus-specific T cells in combination with IL-12, IL-18, IL-33, and TNFα (Fig. 3).
  96. [96]
    Bystander CD4 + T cells: crossroads between innate and adaptive ...
    Aug 28, 2020 · An increasing body of evidence suggests that effector/memory T cells can be activated in the absence of antigen stimulation by pro-inflammatory ...
  97. [97]
    No evidence of immune exhaustion after repeated SARS-CoV-2 ...
    Jun 5, 2025 · Repeated SARS-CoV-2 vaccination was not associated with increased T cell exhaustion in older frail adults, immunosuppressed individuals, or healthy adults.
  98. [98]
  99. [99]
  100. [100]
  101. [101]
  102. [102]
  103. [103]
    Tissue-resident memory T cells invade the brain parenchyma in ...
    Jun 1, 2020 · Inflammatory active multiple sclerosis lesions are populated by CD8+ tissue-resident memory T cells, which show signs of reactivation and infiltration of the ...
  104. [104]
    Tissue-resident memory T cells in the multiple sclerosis brain and ...
    Jan 26, 2023 · Presence of EBV infected B cells and EBV-specific CD8 T cells in the multiple sclerosis (MS) brain suggests a role for virus-driven ...
  105. [105]
    Antigen presentation by B cells enables epitope spreading across ...
    Oct 31, 2023 · Circumstantial evidence suggests that B cells may instruct T cells to break tolerance. Here, to test this hypothesis, we used a murine model ...
  106. [106]
    Repertoire dynamics of autoreactive T cells in multiple sclerosis ...
    Repertoire dynamics of autoreactive T cells in multiple sclerosis patients and healthy subjects: epitope spreading versus clonal persistence. Brain. 2000 Mar ...
  107. [107]
    T‐cell exhaustion in HIV infection - PMC - PubMed Central
    In this review, we will discuss the role of T‐cell exhaustion in HIV disease progression, the long‐term defect of T‐cell function even in aviremic patients on ...
  108. [108]
    Reversal of T Cell Exhaustion in Chronic HCV Infection - PMC
    Most of studies demonstrated that DAA treatment may result in at least partial restoration of T cell immune function.
  109. [109]
    Epigenetic scars of CD8+ T cell exhaustion persist after cure of ...
    Here we show that exhausted CD8+ T cells responding to HCV and HIV share a core regulatory program that is largely irreversible after cure of infection, leading ...<|control11|><|separator|>
  110. [110]
    Roles of Virtual Memory T Cells in Diseases - PMC - PubMed Central
    Virtual memory T cells develop and mediate bystander protective immunity in an IL-15-dependent manner. Nat Commun. 2016;7:11291. doi: 10.1038/ncomms11291 ...
  111. [111]
    Virtual Memory CD8+ T Cells: Origin and Beyond - PMC
    Oct 17, 2022 · One concern that arises from TVM cells is whether they could trigger autoimmunity ... Virtual memory T cells orchestrate extralymphoid responses ...
  112. [112]
    Inflammageing: chronic inflammation in ageing, cardiovascular ...
    This situation might explain why the human CMV-specific memory T cells can comprise up to 50% of the total memory T cell compartment in older individuals ...
  113. [113]
    T cell fate decisions during memory cell generation with aging - PMC
    We review age-sensitive, molecular pathways and gene regulatory networks that bias naïve T cell differentiation towards effector cell generation.
  114. [114]
    Alemtuzumab-induced immune phenotype and repertoire changes
    Alemtuzumab-induced changes including increased numbers of naïve CD4+ T cells and B cells as well as a clonal renewal of CD4+ T- and B-cell repertoires were ...
  115. [115]
    Human autoimmunity after lymphocyte depletion is caused by ...
    Here, we show that T-cell recovery after alemtuzumab is driven by homeostatic proliferation, leading to the generation of chronically activated (CD28−CD57+), ...
  116. [116]
    TSTEM-like CAR-T cells exhibit improved persistence and tumor ...
    Apr 5, 2023 · Patients who receive chimeric antigen receptor (CAR)–T cells that are enriched in memory T cells exhibit better disease control as a result ...
  117. [117]
    CAR-T cell therapy for cancer: current challenges and future directions
    Jul 4, 2025 · In the following pages, we will examine the current application of CAR-T cells in the treatment of another disease, which, similar to solid ...
  118. [118]
    Recent advances in universal chimeric antigen receptor T cell therapy
    Aug 29, 2025 · CAR T and UCAR-T face similar challenges in treating solid tumors, including tumor heterogeneity and the TME, which can lead to inadequate ...
  119. [119]
    Stem cell like memory T cells: A new paradigm in cancer ...
    Stem cell like memory T (TSCM) cells have emerged as the apex of memory T cell differentiation for their properties of self-renewal and replenishing ...Review Article · 4. Strategies To Induce T... · 5. T Cells In The...
  120. [120]
    CAR-T Cell Performance: How to Improve Their Persistence?
    Clinical results have shown that less differentiated memory T cells are required for the sustained in vivo persistence of adoptively transferred CAR-T cells, ...Abstract · CAR-T Cell Differentiation... · T-Cell Subsets · Strategies to Improve the...
  121. [121]
    In vivo CAR-T cell therapy: New breakthroughs for cell-based tumor ...
    Here, we review the key platforms of in vivo gene delivery and the progress of in vivo CAR-T cell therapy for cancers. We discuss the challenges of in vivo CAR- ...