bims-imseme Biomed News
on Immunosenescence and T cell metabolism
Issue of 2025–07–13
sixteen papers selected by
Pierpaolo Ginefra, Ludwig Institute for Cancer Research



  1. Genes Immun. 2025 Jul 05.
      The long-term persistence of naive T lymphocytes is maintained by a state of relative quiescence. Upon antigenic stimulation, these naive T cells undergo rapid activation and proliferation, differentiating into effector cells with specific clonal expansion. Recently, in-depth studies have revealed a fundamental difference in the metabolic requirements of distinct T cell subsets. The fate of CD4 + T cells is influenced by glucose-mediated glycolysis and oxidative phosphorylation (OXPHOS). In this context, key enzymes and various glycolytic intermediates, in conjunction with transcription factors and cytokines, play a crucial role in CD4 + T cell differentiation and function. In our study, we investigated the mechanisms underlying glycolytic reprogramming in CD4 + T cells, with a particular focus on the role of glycolytic enzymes in modulating cytokines and transcription factors that govern T cell differentiation.Our aim is to provide novel insights into the treatment of clinically relevant immune diseases by thoroughly elucidating the characteristics and potential regulatory mechanisms of glucose metabolism in CD4 + T cells.
    DOI:  https://doi.org/10.1038/s41435-025-00340-8
  2. Cell Immunol. 2025 Jun 28. pii: S0008-8749(25)00086-3. [Epub ahead of print]414 105000
      Upon activation, naïve T cells undergo rapid proliferation and differentiation, giving rise to clonally expanded populations specifically tailored for an effective immune response. To meet the heightened bioenergetic and biosynthetic demands associated with activation, T cells adapt and reprogram both their metabolism and transcriptome. Beyond this, T cells are also able to dynamically adapt to fluctuations in the microenvironmental nutrient levels. While the adaptability of T cells is a well-established hallmark of their functionality, the molecular mechanisms by which metabolic responses underpin this flexibility remain incompletely defined. Acetyl-CoA, with its role as a central metabolite in mitochondrial ATP production, and a substrate for nuclear histone acetylation reactions, emerges as a key player in a metabolic-epigenetic axis. Recent evidence indicates that enzymes responsible for generating acetyl-CoA can translocate to the nucleus, supporting sub-cellular local acetyl-CoA production. Here, we explore the impact of acetyl-CoA metabolism on T cell functionality within different subcellular compartments and highlight the potential for intervention in acetyl-CoA metabolic pathways in T cell-driven autoimmune diseases and cancers.
    Keywords:  Acetyl-CoA; Epigenetic remodelling; Metabolic reprogramming; Nuclear metabolism; T cells
    DOI:  https://doi.org/10.1016/j.cellimm.2025.105000
  3. Front Med (Lausanne). 2025 ;12 1577906
       Introduction: CD8+ T cells are vital in the immune control of cancer and a key player in cell-based cancer immunotherapy. Recent studies have shown that microbial short-chain fatty acids (SCFA) can promote both effector and memory phenotypes in CD8+ T cells and may thereby enhance protection against cancer.
    Methods: In this study, we determined the effect of SCFA butyrate on mouse CD8+ T cell function in vitro and in vivo, using the OT-I model.
    Results: Butyrate co-culture with anti-CD3 + anti-CD28 activated T cells in vitro enhanced the frequency of effector CD8+ IFN-γ-producing cells, and the amount of cytokine produced per cell. Culture with butyrate also enhanced the activation, TCR expression, and levels of phosphorylated mTOR proteins within CD8+ T cells but reduced proliferation rate and increased apoptosis. Butyrate-treated activated cells conferred tumor protection after adoptive transfer. Butyrate-treated cells were present at higher frequencies within the tumor compared to non-butyrate treated cells, and expressed IFN-γ. When analyzed using high dimensional cytometry, the tumors of mice that received butyrate-treated cells were enriched in clusters displaying an effector memory phenotype with high expression of IL-15Rβ and T-bet.
    Discussion: Our findings show that butyrate promotes the effector activity of CD8+ T cells in culture, which can persist in vivo while also stimulating memory phenotypes. Consequently, butyrate treatment may have strong application in T cell-based immunotherapies to improve protective cell functions and patient outcomes.
    Keywords:   T cell memory; CD8+ T cells; activation; butyrate; cancer; effector function; immunotherapy; short-chain fatty acids
    DOI:  https://doi.org/10.3389/fmed.2025.1577906
  4. Immunity. 2025 Jul 01. pii: S1074-7613(25)00248-1. [Epub ahead of print]
      During persistent antigen stimulation, CD8+ T cell responses are maintained by progenitor exhausted CD8+ T (Tpex) cells. Tpex cells respond to blockade of the inhibitory receptor programmed cell death-1 (PD-1), and regulation of their differentiation is critical for immunotherapies. Tpex cells highly express inducible costimulator (ICOS), but how ICOS modulates PD-1+CD8+ T cells is not clear. During chronic infection, intrinsic ICOS deficiency increased number and quality of virus-specific CD8+ T cells. Loss of ICOS potentiated activity of the transcription factor forkhead box O1 (FoxO1) and memory-like features of Tpex cells. ICOS-deficient Tpex cells were poised to generate effecor-like cells with improved survival and cytokine production. ICOS-ligand (ICOSL) blockade expanded effector-like PD-1+CD8+ T cells, reduced viral load, and improved response to PD-1 blockade. Similarly, in a mouse model of hepatocellular carcinoma, ICOS inhibition enhanced tumor-specific CD8+ T cell responses and tumor control by PD-1 blockade. Overall, we show that sustained ICOS costimulation limits CD8+ T cell responses during chronic antigen exposure.
    Keywords:  CD8(+) T cells; FoxO1; ICOS; LCMV; PD-1; T cell differentiation; costimulation; exhaustion; immunotherapy; progenitor exhausted
    DOI:  https://doi.org/10.1016/j.immuni.2025.06.001
  5. Nat Commun. 2025 Jul 10. 16(1): 6394
      Memory T cells are a highly heterogeneous collection of antigen-experienced cells that undergo dynamic adaptations upon antigen re-encounter and environmental signals. This heterogeneity hinders studies on memory T cell durability and age-related dysfunction. Using chronic Epstein-Barr virus (EBV) infection and barcode-enabled antigen tracing, we assess the influence of age on memory states at the level of single antigen-specific CD8+ T cells. In young adults (<40 years), EBV-specific CD8+ T cells recognizing different antigenic peptides assume divergent preferred differentiation phenotypes. In older adults (>65-years), antigen-specific cells show largely distinct phenotypic and transcriptomic aging trajectories. Common to many albeit not all antigen-specific populations are maintained TCR diversity, gained natural killer cell-like, innate signatures and lost stem-like features while no evidence is seen for cellular senescence or exhaustion. TCR avidity contributes to these phenotypic differences and aging-related changes. Collectively, our data uncover divergent antigen-guided aging shifts in memory T cell phenotypes, which are informative for antigen selection in optimizing vaccine design and adoptive T cell therapy.
    DOI:  https://doi.org/10.1038/s41467-025-61627-y
  6. Immun Ageing. 2025 Jul 08. 22(1): 29
       BACKGROUND: Immunosenescence describes the gradual remodeling of immune responses, leading to disturbed immune homeostasis and increased susceptibility of older adults for infections, neoplasia and autoimmunity. Decline in cellular immunity is associated with intrinsic changes in the T cell compartment, but can be further pushed by age-related changes in cells regulating T cell immunity. Myeloid-derived suppressor cells (MDSCs) are potent inhibitors of T cell activation and function, whose induction requires chronic inflammation. Since aging is associated with low grade inflammation (inflammaging) and increased myelopoiesis, age-induced changes in MDSC induction and function in relation to T cell immunity were analyzed.
    RESULTS: MDSC numbers and functions were compared between "healthy" young and old adults, who were negatively diagnosed for severe acute and chronic diseases known to induce MDSC accumulation. MDSCs were either isolated from peripheral blood or generated in vitro from blood-derived CD14 cells. Aging was associated with significantly increased MDSC numbers in the monocytic- (M-) and polymorphonuclear (PMN-) MDSC subpopulations. MDSCs could be induced more efficiently from CD14 cells of old donors and these MDSCs inhibited CD3/28-induced T cell proliferation significantly better than MDSCs induced from young donors. Serum factors of old donors supported MDSC induction comparable to serum factors from young donors, but increased immunosuppressive activity of MDSCs was only achieved by serum from old donors. Elevated immunosuppressive activity of MDSCs from old donors was associated with major metabolic changes and increased intracellular levels of neutral and oxidized lipids known to promote immunosuppressive functions. Independent of age, MDSC-mediated suppression of T cell proliferation required direct MDSC- T cell contact. Besides their increased ability to inhibit activation-induced T cell proliferation, MDSCs from old donors strongly shift the immune response towards Th2 immunity and might thereby further contribute to impaired cell-mediated immunity during aging.
    CONCLUSIONS: These results indicate that immunosenescence of innate immunity comprises accumulation and functional changes in the MDSC compartment, which directly impacts T cell functions and contribute to age-associated impaired T cell immunity. Targeting MDSCs during aging might help to maintain functional T cell responses and increase the chance of healthy aging.
    Keywords:  Immunosenescence; Myeloid-derived suppressor cells; T cell immunity
    DOI:  https://doi.org/10.1186/s12979-025-00524-w
  7. JCI Insight. 2025 Jul 08. pii: e174235. [Epub ahead of print]
      Hematopoietic Protein-1 (Hem1) is a component of the WASP-family verprolin-homologous protein (WAVE) actin regulatory complex, which is activated downstream of multiple immune receptors. Mutations in the NCKAP1L gene encoding HEM1 have recently been found to result in severe Primary Immunodeficiency Disease (PID), characterized by recurrent respiratory infections, hyperinflammation, autoimmunity, and high mortality. However, how loss of Hem1 results in PID is unclear. To define the importance of Hem1 specifically in T cells, we generated constitutive and T cell specific Hem1 null mice. Hem1 deficient T cells exhibited an increased shift from naïve to memory T cells, and increased ratio of immunosuppressive regulatory to effector T cells. Loss of Hem1 resulted in hallmarks of T cell exhaustion including T cell lymphopenia, decreased activation and proliferation, increased expression of PD-1 and Tim3, and increased IL-10 production. In vitro TCR stimulation of CD4 T cells resulted in increased production of Th1 (IFN), Th2 (IL-5, IL-13), Th17 (IL-17, IL-22), and Treg (IL-10) cytokines. This correlated with reduced F-actin, increased expression of CD107a, and increased granzyme release indicative of increased granule membrane fusion and exocytosis. These results suggest that Hem-1 is critical for maintaining T cell activation, homeostasis and regulated cytokine production following antigen encounter.
    Keywords:  Adaptive immunity; Cytoskeleton; Immunology; Inflammation; T cells
    DOI:  https://doi.org/10.1172/jci.insight.174235
  8. Cell Mol Immunol. 2025 Jul 10.
      T cells often acquire diverse phenotypes and functional states following activation. CD4+ T cells are traditionally classified into distinct effector subsets, such as Th1, Th2, Th17, and Tfh, on the basis of their cytokine profiles and functional roles. While this framework has advanced our understanding of adaptive immunity, it has limitations in explaining the persistence of T-cell responses in settings of autoimmunity and transplant rejection, in contrast to its limited efficacy in cancer. Moving beyond this subset-based framework, recent studies have revealed that stemness and adaptation are fundamental to CD4+ T-cell fate and function. Central to this new understanding is the TCF1+ stem-like CD4+ T-cell population, which emerges early after activation and serves as a reservoir for effector differentiation. These cells dynamically integrate environmental cues to direct effector differentiation and shape functional outcomes at target tissue sites, a process we define as clonal adaptation. By balancing self-renewal with effector differentiation, stem-like CD4+ T cells continue to replenish short-lived effector cells to sustain autoimmunity, transplant rejection, chronic infections, and allergic diseases. However, under tolerogenic conditions or within the tumor microenvironment, these cells often fail to differentiate into effectors, instead entering dysfunctional states or regulatory T-cell differentiation. Targeting stem-like CD4+ T cells offers great therapeutic potential: disrupting their persistence could mitigate autoimmune pathology and transplant rejection, whereas enhancing their effector capacity could improve antitumor immunity.
    Keywords:  Autoimmunity; CD4+ T cells; Cancer immunotherapy; Effector differentiation; Stem-like T cells; Transplant rejection
    DOI:  https://doi.org/10.1038/s41423-025-01321-7
  9. J Vis Exp. 2025 Jun 20.
      The widespread accessibility of clustered regularly interspaced short palindromic repeat (CRISPR)-Cas9 technology has made gene targeting in primary cells a routine method for evaluating gene function in T cells. Given the cost and limited availability of knockout (KO) mouse strains, testing preliminary hypotheses involving gene function in T cells can be prohibitive using gene-targeted animal models. However, using commercially available resources, including predesigned guide RNAs (gRNAs), researchers can conveniently generate gene-targeted naïve T cells that can be used for T cell activation and differentiation studies. Here we outline a protocol for using nucleofection-delivered CRISPR-Cas9 ribonucleoprotein complexes (RNPs) to efficiently generate gene KO murine naïve CD4 T cells that can be used to evaluate gene function in CD4 T cell differentiation, in vitro. Isolation of naïve CD4 T cells from mouse secondary lymphoid organs, followed by nucleofection with Cas9-gRNA complexes ensures gene KO is initiated before downstream T cell activation, offering a strategic advantage over retroviral-mediated gRNA delivery, which typically requires preactivation of T cells, preventing the evaluation of effects in naïve T cells. Furthermore, this nucleofection-based method bypasses potential developmental issues associated with gene KO animals. Following Cas9-gRNA delivery, we describe protocols for studying CD4 T cell differentiation into Th1, Th2, Th17, and Treg lineages using in vitro polarization. In addition, this protocol is adaptable to using gene-targeted CD4 or CD8 T cells for numerous downstream applications, including other T cell activation studies in vitro and adoptive transfer studies in vivo. The use of CRISPR-Cas9 methods has streamlined our ability to evaluate gene function in T cells and allows for the routine KO of many genes of interest, freeing researchers from limitations associated with studying gene KO animals.
    DOI:  https://doi.org/10.3791/67380
  10. bioRxiv. 2025 Apr 11. pii: 2025.04.09.647991. [Epub ahead of print]
      Nicotinamide adenine dinucleotide (NAD + ) is an essential redox cofactor and signaling molecule linked to age-dependent metabolic decline, with its compartmentalization regulated by the mitochondrial carrier SLC25A51. The mechanisms contributing to declining NAD + levels during aging and the consequences of altered NAD + homeostasis across tissues are poorly understood. Here, we show that SLC25A51 is upregulated in aging and aging-associated conditions, particularly in senescent cells. In a mouse model of beta-cell senescence, upregulated SLC25A51 was associated with beta-cell identity loss, senescence progression, and a reduced NAD + /NADH ratio. SLC25A51 was elevated following p16 INK4a -, replicative-, irradiation-, and H 2 O 2 -induced senescence, with NRF2 implicated as a potential transcriptional regulator. Overexpression of SLC25A51, but not a transport-dead mutant, induced senescence factors, while its deletion prevented this effect. Beta-cell-specific deletion of SLC25A51 lowered p16 INK4a levels in pancreatic islets, circulating insulin, and glucose levels, improving insulin sensitivity and indicating its role in cellular senescence and the metabolic control of beta-cell function.
    DOI:  https://doi.org/10.1101/2025.04.09.647991
  11. Immunity. 2025 Jul 08. pii: S1074-7613(25)00282-1. [Epub ahead of print]58(7): 1620-1622
      Tumor-infiltrating CD8+ T cells with a tissue-resident memory phenotype have been described in both mice and humans. In this issue of Immunity, Green et al. provide a detailed analysis of the transcriptional and epigenetic regulation of these cells in relation to bona fide tissue-resident and exhausted T cells found in acutely resolving and chronic viral infections.
    DOI:  https://doi.org/10.1016/j.immuni.2025.06.013
  12. Sci China Life Sci. 2025 Jul 08.
      Tumor-infiltrating lymphocyte (TIL) therapy was recently approved for melanoma patients; however, the dynamic changes in T cell subpopulations during TIL production remain poorly understood. Here, we analyzed epithelial ovarian cancer samples at various stages of ex vivo TIL culture using paired single-cell RNA and TCR sequencing. We also assessed the expansion potential and tumor reactivity of the identified TIL subpopulations. Single-cell transcriptomic analysis revealed that CD8+ TILs exhibited reduced cellular diversity following ex vivo expansion, selectively expanding stem-like TCF7+ precursors of exhausted T cells (Tpex) and effector-like tissue-resident memory (Trm) cells. TCR clonotype analysis showed that Tpex cells accumulated through self-renewal, while Trm cells primarily originated from TCF7+GZMK+ early effector memory cells in tumors. Additionally, TCR tracing identified preferential activation and reprogramming of CD4+ T follicular helper (Tfh)-like cells, especially TCF7+ ones. All three TCF7+ subpopulations showed robust expansion potential and tumor reactivity in vitro. Notably, CCR7+CD200+ T cells, enriched for TCF-1+CD8+ Tpex and CD4+ Tfh-like cells in the tumor microenvironment, exhibited self-renewal during in vitro expansion and demonstrated tumor reactivity both in vivo and in vitro. These findings highlight the selective expansion of tumor-reactive TCF7+ T cells during TIL culture and suggest that CCR7 and CD200 serve as important surface markers for generating stem-like, tumor-reactive cells, potentially improving TIL therapy in cancers.
    Keywords:  adoptive cell therapy; expansion; ovarian cancer; tumor reactivity; tumor-infiltrating lymphocyte
    DOI:  https://doi.org/10.1007/s11427-025-2958-3
  13. Cell. 2025 Jul 04. pii: S0092-8674(25)00684-1. [Epub ahead of print]
      Exercise improves immune checkpoint inhibitor (ICI) efficacy in cancers such as melanoma; however, the mechanisms through which exercise mediates this antitumor effect remain obscure. Here, we identify that the gut microbiota plays a critical role in how exercise improves ICI efficacy in preclinical melanoma. Our study demonstrates that exercise stimulates microbial one-carbon metabolism, increasing levels of the metabolite formate, which subsequently enhances cytotoxic CD8 T cell (Tc1)-mediated ICI efficacy. We further establish that microbiota-derived formate is both sufficient and required to enhance Tc1 cell fate in vitro and promote tumor antigen-specific Tc1 immunity in vivo. Mechanistically, we identify the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) as a crucial mediator of formate-driven Tc1 function enhancement in vitro and a key player in the exercise-mediated antitumor effect in vivo. Finally, we uncover human microbiota-derived formate as a potential biomarker of enhanced Tc1-mediated antitumor immunity, supporting its functional role in melanoma suppression.
    Keywords:  CD8 T cells; FMT; Nrf2; exercise; formate; immune checkpoint inhibitor; melanoma; microbiota; microbiota metabolite
    DOI:  https://doi.org/10.1016/j.cell.2025.06.018
  14. bioRxiv. 2025 Jul 03. pii: 2025.06.27.660220. [Epub ahead of print]
      Unresolved inflammation and fibrosis are the two key features of metabolic dysfunction-associated steatohepatitis (MASH), a progressive form of steatotic liver disease that can evolve into cirrhosis and liver cancer. Although innate immunity has been well studied in MASH, the role of CD4⁺ T cells remains underexplored despite their potential to coordinate immune responses by providing help to other immune cells, promoting inflammation, or regulating immune activity through effector and regulatory subsets. To better understand the role of CD4 + T cells in the pathogenesis of MASH, we comprehensively characterized hepatic CD4 + T cells in murine and human MASH at a single-cell protein, transcriptional, and functional level. Mass cytometry and CITE-sequencing revealed a marked shift in intrahepatic CD4⁺ T-cell composition in MASH, with enrichment of Th1, regulatory, and cytotoxic CD4⁺ T cells. Similar phenotypic changes were mirrored in the peripheral blood and validated in human MASH samples. Functional assays demonstrated increased production of IFNγ and TNFα by hepatic CD4⁺ T cells, highlighting their proinflammatory effector activity. Transcriptomic profiling identified Tnfrsf4 (OX40) upregulation in hepatic CD4⁺ T cells during MASH. Therapeutic blockade of the OX40L-OX40 axis reversed hepatic fibrosis and improved histologic disease scores in mice with established MASH, and also decreased inflammatory markers in a human ex vivo liver model. Together, these studies provide a proteogenomic single-cell atlas for hepatic CD4⁺ T cells and uncover a CD4⁺ T cell-dependent immunopathogenic circuit as a promising immunotherapeutic target to alleviate MASH and liver fibrosis.
    DOI:  https://doi.org/10.1101/2025.06.27.660220
  15. PLoS Pathog. 2025 Jul 09. 21(7): e1013218
      West Nile Virus (WNV), a mosquito-borne neurotropic flavivirus, is a major cause of viral encephalitis in the United States, posing a continuous threat to public health. Unfortunately, no vaccine or specific therapeutic intervention is available against WNV infection. Previous studies, including ours, demonstrated that interleukin-17A (IL-17A) signaling promotes the cytotoxicity of CD8+ T cells to facilitate WNV and parasite clearance; however, the molecular mechanism is not understood. IL-17 receptor C (IL-17RC) is an obligatory co-receptor with IL-17 receptor A (IL-17RA) for signaling induced by IL-17A, IL-17A/F, and IL-17F. In this study, we found that IL-17RC deficient (Il17rc-/-) mice were more susceptible to WNV infection with a higher viral load in the brain than wild-type (WT) control mice. The number of infiltrating WNV-specific CD8+ T cells and the expression levels of cytotoxicity mediators, such as perforin, in the T cells in the brain of Il17rc-/- mice were reduced. In addition, WNV-specific CD8+ T cells from IL-17RA deficient (Il17ra-/-) mice and CD8+ cell-specific Il17ra conditional knockout (cre-KO) mice expressed lower levels of perforin than their counterpart controls. Moreover, supplementing mouse recombinant IL-17A ex vivo increased the perforin production in WNV-specific CD8+ T cells from the WT mice but not Il17rc-/- or cre-KO mice. Interestingly, we found that IL-17A signaling activated the phosphatidylinositol-3-kinase/mammalian target of rapamycin (PI3K-mTOR) signaling pathway in CD8+ T cells, leading to increased metabolism of CD8+ T cells to cope with the higher energy demand for WNV clearance in the brain. In summary, our findings reveal a novel IL-17A-PI3K-mTOR signaling axis in promoting the effector functions of CD8+ T cells, suggesting potential broader implications in stimulating immune responses to combat WNV and other intracellular infections.
    DOI:  https://doi.org/10.1371/journal.ppat.1013218