bims-traimu Biomed News
on Trained immunity
Issue of 2025–09–07
eighteen papers selected by
Yantong Wan, Southern Medical University



  1. Elife. 2025 Sep 02. pii: e106443. [Epub ahead of print]14
      The long-term functional adaptation of innate immune cells following an initial stimulation, referred to as trained immunity or innate immune memory, enhances responsiveness and protection against secondary infections. Toll-like receptors (TLRs), an evolutionarily conserved family, recognize microbial-associated molecular patterns, initiating innate and adaptive immune responses. TLR signaling cascades induce the production of pro-inflammatory cytokines, antimicrobial peptides, and interferons, promoting pathogen clearance, while also driving epigenetic and metabolic reprogramming that enhances immune responses and protection to subsequent challenges. However, TLRs also recognize endogenous ligands contributing to chronic inflammation and autoimmune diseases. This review examines the role of TLRs and their various agonists in mediating trained immunity across diverse immune cell types, with an emphasis on their dual role in protecting against infections and chronic inflammation. It highlights recent clinical trials of TLR agonists as immunomodulatory agents and their therapeutic potential in infectious diseases and cancer. By providing an in-depth analysis of TLR-driven trained immunity, this review highlights the extensive influence of TLRs on immune cell populations and their implications for the development of novel, broad-spectrum immunotherapies.
    Keywords:  Toll-like receptors; chronic inflammation; immunology; inflammation; innate immunity; microbial infection; trained immunity
    DOI:  https://doi.org/10.7554/eLife.106443
  2. Elife. 2025 Sep 04. pii: e106610. [Epub ahead of print]14
      Human and murine studies reveal that innate immune cells are able to mount enhanced responses to pathogens after primary inflammatory exposure. Innate immune memory has been shown to last for months to years, longer than the lifespan of most innate immune cells. Indeed, long-lived hematopoietic stem and progenitor cells (HSPCs) serve as a cellular reservoir for innate immune memory. In this review, we summarize the evidence that innate immune memory is epigenetically encoded in HSPCs, and we consider whether HSPC subpopulations with differentiation bias, cell autonomous epigenetic reprogramming, or both features underlie the phenomenon of central trained immunity. We further profile the significant implications of central trained immunity in stem cell transplant, aging, inflammatory diseases, and vaccination strategies for the future.
    Keywords:  epigenetics; hematopoietic progenitor cells; hematopoietic stem cells; immunology; inflammation; innate immune memory; metabolism; trained immunity
    DOI:  https://doi.org/10.7554/eLife.106610
  3. J Clin Invest. 2025 Sep 02. pii: e192469. [Epub ahead of print]135(17):
      Trained immunity (TRIM) is a form of long-lasting functional reprogramming of innate immune cells and their progenitors that enhances responsiveness to subsequent stimuli. Although first characterized in myeloid cells, TRIM was recently extended to nonmyeloid cell types, including endothelial and glial cells, which also exhibit stimulus-driven, memory-like behavior. While initially recognized as a protective mechanism, particularly in the context of vaccines and acute infections, TRIM can also become maladaptive, promoting chronic inflammation, immune dysfunction, and disease. This Review focuses on virus-induced TRIM while also addressing microbial, metabolic, and endogenous inducers. We examine key ligands and receptors that initiate TRIM and dissect the associated signaling and epigenetic pathways. Importantly, we argue that maladaptive TRIM arises not from a specific ligand, receptor, or molecular event, but from contextual factors such as stimulus persistence, dose, tissue microenvironment, and preexisting inflammation. The nature of the secondary challenge also shapes whether a trained response is adaptive or maladaptive. We further discuss TRIM induction in the bone marrow, involvement of both myeloid and nonmyeloid cells, and the role of lipid rafts in sustaining TRIM. We review maladaptive TRIM's potential contribution to systemic diseases, such as atherosclerosis, diabetes, sepsis, cancer, and autoimmunity, along with its influence on viral vaccine responses. Finally, we outline potential strategies to redirect maladaptive TRIM and propose key outstanding questions for future research.
    DOI:  https://doi.org/10.1172/JCI192469
  4. J Leukoc Biol. 2025 Aug 29. pii: qiaf123. [Epub ahead of print]
      Trained immunity amplifies innate immune responses in an antigen-independent manner. This study explored the ability of trained human primary macrophages to modulate the phenotype and function of T cells. Macrophages play an important role in antigen presentation, resulting in T-cell activation and antigen-specific clonal expansion; however, few studies have investigated whether trained immunity induction in macrophages modulates T cell activation. Here, through surface marker analysis of naive, β-glucan-, and BCG-trained macrophages, we identified eight distinct macrophage clusters following trained immunity induction. One of these populations showed an increase in surface activation markers CD40 and CD86, as well as MHC molecules. In vitro co-culture of T cells with autologous BCG-trained macrophages resulted in a skewing towards TH17 cells. We also observed an increase in TH17 percentage after BCG vaccination of human subjects. The bias towards TH17 triggered by trained macrophages required direct T cell to macrophage contact. Trained macrophages potentiated TH17 skewing independently of the antigen presented. While co-cultures of T cells and BCG-trained macrophages responded with higher production of interferon (IFN)-γ and interleukin (IL)-17 after stimulation, no clear shifts towards effector or memory T cells were observed. In conclusion, this study provides evidence that BCG-trained macrophages can modulate T cell function towards a TH17 phenotype, suggesting that BCG-induced trained immunity has the potential to enhance not only innate immune responses but also to modify adaptive T cell immunity.
    Keywords:  BCG; T cells; Trained Immunity; macrophages; vaccination
    DOI:  https://doi.org/10.1093/jleuko/qiaf123
  5. Commun Biol. 2025 Aug 30. 8(1): 1314
      Shrimp aquaculture is critically important for global food security, but viral diseases like white spot syndrome virus (WSSV) cause devastating economic losses, highlighting the urgent need for effective disease control strategies. While trained immunity has been observed in invertebrates like shrimp after exposure to pathogens, the underlying molecular mechanisms remain elusive. Here we reveal that lysine acetyltransferase KAT8-mediated histone H3K27ac is critical for antiviral defense in shrimp Marsupenaeus japonicus. We demonstrate that ultraviolet-inactivated WSSV (UV-WSSV) induces antiviral trained immunity in the shrimp via KAT8-dependent H3K27ac. UV-WSSV training enhances glycolysis and the tricarboxylic acid (TCA) cycle, increasing acetyl-CoA production. This acetyl-CoA fuels KAT8 activity, depositing H3K27ac marks at the promoter of the NF-κB-like transcription factor Dorsal. This epigenetic modification upregulates Dorsal expression, leading to the enhanced production of the antiviral cytokine Vago5 and antimicrobial peptides (AMPs) upon subsequent WSSV challenge. Furthermore, H3K27ac directly activates key glycolytic genes (Hk2, Pk, Ldh), creating a feedforward loop that sustains metabolic reprogramming. Our work reveals a conserved KAT8-H3K27ac axis driving trained immunity in invertebrates through integrated metabolic-epigenetic crosstalk, analogous to mammalian systems. These findings provide a crucial theoretical foundation for developing antiviral vaccines and sustainable immunostimulants to control disease in shrimp aquaculture.
    DOI:  https://doi.org/10.1038/s42003-025-08767-5
  6. Front Immunol. 2025 ;16 1672030
      
    Keywords:  KLF4; alternative splicing; immune cells; inflammation; stem cells; trained immunity
    DOI:  https://doi.org/10.3389/fimmu.2025.1672030
  7. Elife. 2025 Aug 28. pii: e106654. [Epub ahead of print]14
      Originally defined in the context of influenza vaccines by Thomas Francis Jr. in the late 1950s, original antigenic sin (OAS) refers to the tendency of the immune system to preferentially recall B cell memory against primary antigen after secondary exposure to different but related antigen. This competes with the elicitation of de novo antibodies by lowering the frequency of antigen reception by the naïve B cell lymphocyte pool residing within secondary lymphoid organs. Consequently, OAS imposes a 'primary addiction' that modulates the target epitope specificity of the secondary antibody response and has wide-reaching consequences for vaccines that require seasonal updating, including influenza and SARS-CoV-2. Rationally designed vaccines that preferentially stimulate the production of de novo antibodies rather than those derived from recalled B cell memory are of central interest, particularly for universal vaccine formulations tasked with directing robust humoral immunity against these viruses which, due to their ongoing evolution, have 'resisted' conventional vaccine approaches. Largely absent from this discussion is an integrated evaluation of what Janeway famously called 'the immunologists dirty secret', that humoral immune reactions require stimulation by the innate immune system. In this perspective piece, we present a hypothesis that innate immune cells and trained immunity, a collective term for the epigenetic reprogramming that enhances responsiveness upon re-stimulation, provides a template for promoting de novo expansion of the naïve B cell repertoire over recallable memory. This natural control axis may inform the design of vaccines that seek to avoid primary addiction and OAS.
    Keywords:  B cell; immunology; imprinting; infectious disease; inflammation; microbiology; repertoire
    DOI:  https://doi.org/10.7554/eLife.106654
  8. J Cardiol. 2025 Aug 29. pii: S0914-5087(25)00217-5. [Epub ahead of print]
      The cardiopulmonary system is not only a pump-respirator network but also a sophisticated sensor-effector circuit. Recent findings have highlighted how mechanical and inflammatory stress in the heart and lungs is transmitted via afferent nerves-including vagal, glossopharyngeal, and spinal fibers-to the brainstem and higher autonomic centers. These afferent signals trigger adaptive or maladaptive efferent responses via the sympathetic nervous system, which in turn modulate immune cell dynamics in the bone marrow and peripheral organs. This review discusses the cardiopulmonary afferent-efferent axis, focusing on three major components: (1) the molecular and functional basis of afferent pathways linking the heart and lungs to the brain; (2) the impact of these inputs on central autonomic regulation; (3) the downstream effects of sympathetic outflow on hematopoietic stem cells, leading to myeloid skewing, epigenetic memory, and inflammatory macrophage expansion. Finally, we explore how this axis contributes to cardiovascular disease progression and multimorbidity, and how recent studies-especially those on innate immune memory-open new therapeutic avenues targeting neuro-immune crosstalk.
    Keywords:  Bone marrow niche; Cardiac afferents; Clonal hematopoiesis; Interoception; Trained immunity
    DOI:  https://doi.org/10.1016/j.jjcc.2025.08.013
  9. J Neurochem. 2025 Sep;169(9): e70219
      Polar metabolic profiling, as well as bioenergetic assays, were used to characterize microglial responses to lipopolysaccharide, which induces a pro-inflammatory state, and interleukin-4, which is associated with an anti-inflammatory phenotype. BV2 microglial cells and primary microglia were used for these investigations. Results revealed that lipopolysaccharide-treated microglia exhibited an increased aerobic glycolytic activity measured by extracellular flux analysis, accompanied by increased levels of endogenous itaconate, a metabolite produced by the IRG1 enzyme. Increased itaconate levels observed by LC-HRMS were found to be associated with a stabilization of the NF2L2/NRF2 transcription factor. Attenuation of the Acod1 gene leads to increased pro-inflammatory cytokine production, as measured by ELISA, while having no effect on LPS-induced oxidative stress or neurotoxicity, an effect only observed upon silencing Nfe2l2. This suggests that an IRG1/itaconate/NRF2 axis functions as a feedback mechanism. The study underscores the dual role of metabolic reprogramming in microglial activation, balancing inflammation and neuroprotection, and suggests potential therapeutic targets for neuroinflammatory diseases by modulating itaconate and NF2L2/NRF2-related pathways. This work highlights the complexity and therapeutic potential of targeting microglial metabolism in CNS disorders.
    Keywords:  IRG1/ACOD1; NF2L2/NRF2; central nervous system; immune activation; itaconate; metabolic reprogramming; microglia; neuroinflammation
    DOI:  https://doi.org/10.1111/jnc.70219
  10. Cell Host Microbe. 2025 Aug 23. pii: S1931-3128(25)00297-5. [Epub ahead of print]
      Bacteria of the genus Shigella replicate in intestinal epithelial cells and cause shigellosis, a severe diarrheal disease that resolves spontaneously in most healthy individuals. During shigellosis, neutrophils are abundantly recruited to the gut and have long been thought to be central to Shigella control and pathogenesis. However, how shigellosis resolves remains poorly understood due to the longstanding lack of a tractable and physiological animal model. Here, using our newly developed Nlrc4-/-Casp11-/- mouse model of shigellosis, we unexpectedly find no major role for neutrophils in limiting Shigella or in disease pathogenesis. Instead, we uncover an essential role for macrophages in the host control of Shigella. Macrophages respond to Shigella via Toll-like receptors (TLRs) to produce IL-12, which then induces IFN-γ, a cytokine that is essential to control Shigella replication in intestinal epithelial cells. Collectively, our findings reshape our understanding of the innate immune response to Shigella.
    Keywords:  IFN-g; IL-12; Shigella; Shigellosis; innate immunity; intestinal epithelial cells; macrophages; mouse model; neutrophils
    DOI:  https://doi.org/10.1016/j.chom.2025.08.001
  11. Nat Commun. 2025 Aug 30. 16(1): 8119
      The metabolic flexibility of tissues determines the degree and reversibility of organ damage during inflammatory challenges. However, effective treatments for myocardial metabolic dysfunction in septic cardiomyopathy (SCM) are unavailable. Nicotinamide adenine dinucleotide-dependent signaling is fundamental to cellular metabolic homeostasis and inflammatory responses. Here, using male mice models, we reveal that both genetic and pharmacological inhibition of mono-ADP-ribosyl hydrolase MacroD1 which is predominantly enriched in cardiomyocytes alleviates myocardial metabolic impairment, inflammation, dysfunction, and the risk of mortality caused by lipopolysaccharide and cecal ligation and puncture. Mechanistically, MacroD1 selectively modulates the activity of mitochondrial complex I (MCI), which is particularly vulnerable at the early stages of sepsis. Inhibition of MacroD1 preserves MCI activity and bioenergetic reserves of cardiomyocytes by enhancing mono-ADP-ribosylation of Ndufb9 protein, thereby mitigating sepsis-induced myocardial pyroptosis and dysfunction. These preclinical results indicate that MacroD1 dictates cardiac tolerance to sepsis by configuring MCI-coupled bioenergetic reserve and cardiomyocyte pyroptosis.
    DOI:  https://doi.org/10.1038/s41467-025-62384-8
  12. Cell Rep. 2025 Aug 26. pii: S2211-1247(25)00964-7. [Epub ahead of print]44(9): 116193
      Hematopoietic aging is characterized by chronic inflammation associated with myeloid bias, hematopoietic stem cell (HSC) accumulation, and functional HSC impairment. Yet it remains unclear how inflammation promotes aging phenotypes. Nuclear factor κB (NF-κB) both responds to and directs inflammation, and we present an experimental model of elevated NF-κB activity ("inhibitor of κB deficient" [IκB-]) to dissect its role in hematopoietic aging phenotypes. We find that while elevated NF-κB activity is not sufficient for HSC accumulation, HSC-autonomous NF-κB activity impairs their functionality, leading to reduced bone marrow reconstitution. In contrast, myeloid bias is driven by the IκB- proinflammatory bone marrow milieu, as observed functionally, epigenomically, and transcriptomically. A single-cell RNA sequencing (scRNA-seq) HSPC labeling framework enables comparisons with aged murine and human HSC datasets, documenting an association between HSC-intrinsic NF-κB activity and quiescence but not myeloid bias. These findings delineate separate regulatory mechanisms that underlie the three hallmarks of hematopoietic aging, suggesting that they are specifically and independently therapeutically targetable.
    Keywords:  C/EBP; CP: Immunology; CP: Stem cell research; NF-κB; bone marrow cytokine milieu; bone marrow niche; hematopoietic aging; hematopoietic stem cell impairment; inflammaging; microenvironment; myeloid bias; stem cell aging
    DOI:  https://doi.org/10.1016/j.celrep.2025.116193
  13. J Clin Invest. 2025 Sep 02. pii: e184721. [Epub ahead of print]
      The persistent challenge of sepsis-related mortality underscores the necessity for deeper insights, with our multi-center cross-age cohort study identifying insulin-like growth factor binding protein 6 (IGFBP6) as a critical regulator in sepsis diagnosis, prognosis, and mortality risk evaluation. Mechanistically, IGFBP6 engages in IGF-independent binding to prohibitin2 (PHB2) on epithelial cells, driving PHB2 tyrosine phosphorylation during sepsis. This process disrupts STAT1 phosphorylation, nuclear translocation, and its recruitment to the CCL2 promoter, ultimately impairing CCL2 transcription and macrophage chemotaxis. Crucially, PHB2 silencing via siPHB2 and STAT1 activation using 2-NP restored CCL2 expression in vitro and in vivo, improving bacterial clearance and survival in septic mice. Concurrently, IGFBP6 compromises macrophage bactericidal activity by inhibiting Akt phosphorylation, reducing ROS/IL-1β production and phagocytic capacity - defects reversible by Akt agonist SC79. Collectively, IGFBP6 emerges as an endogenous driver of sepsis pathogenesis, positioning it as a dual diagnostic biomarker and therapeutic target. Intervention strategies targeting IGFBP6-mediated signaling may offer transformative approaches for sepsis management.
    Keywords:  Immunotherapy; Infectious disease; Inflammation; Macrophages
    DOI:  https://doi.org/10.1172/JCI184721
  14. bioRxiv. 2025 Aug 22. pii: 2025.08.18.670953. [Epub ahead of print]
      Macrophages are capable of eliminating cancer cells by phagocytosis, particularly in the presence of monoclonal antibody (mAb) therapies targeting tumor antigens. Paradoxically, tumor-associated macrophages are typically associated with poor patient outcome, and can promote tumor growth by secretion of immunosuppressive cytokines and growth factors. The mechanisms by which these pro-tumor macrophage states arise are poorly understood, and it is unclear how mAb-induced cancer cell phagocytosis may contribute to these states. To understand how antibody-dependent cancer cell phagocytosis (ADCP) alters macrophage state and function, we profiled gene expression and chromatin accessibility changes over time after ADCP. We observed that after ADCP, macrophages upregulate an anti-inflammatory gene regulatory program, characterized by expression of pro-angiogenic and immunosuppressive chemokine genes, and increased activity by cellular, oxidative, and lysosomal stress transcription factors. This gene regulatory program was shared among phagocytic macrophages following either ADCP or apoptotic cancer cell phagocytosis, in addition to substrate-specific pathways. Conditioned media from macrophages promoted EMT in cancer cells, but this pro-EMT macrophage phenotype was attenuated following ADCP, but not following apoptotic cancer cell phagocytosis. The phagocytic gene signature we identified in vitro is also expressed by tumor-associated macrophages across numerous cancer types in vivo . Together, this work identifies an anti-inflammatory and immunosuppressive epigenetic program in macrophages following ADCP upon mAb treatment, and expands our understanding of how phagocytosis influences macrophage heterogeneity in the tumor microenvironment.
    DOI:  https://doi.org/10.1101/2025.08.18.670953
  15. Sci Immunol. 2025 Sep 05. 10(111): eadv4810
      RNA modifications regulate phenotype and function of macrophages by regulating RNA translation, splicing, and stability. However, the role of N7-methylguanosine (m7G) modification in macrophages and inflammation remains unexplored. In this study, we observed elevated levels of the methyltransferase METTL1 and m7G modifications in macrophages from mouse and human tissues during acute kidney injury (AKI). METTL1 deficiency in myeloid cells mitigated multiorgan inflammation induced by cecal ligation and puncture and renal ischemia/reperfusion. Genetic deletion of METTL1 inhibited macrophage proinflammatory responses. We identified internal Sarm1 messenger RNA (mRNA) as a target of m7G modification that controls macrophage metabolic reprogramming. METTL1 deficiency in macrophages inhibited metabolic reprogramming, which was reversed by SARM1 overexpression that induced NAD+ decline. Pharmacologically, SA91-0178, a specific METTL1 inhibitor, effectively alleviated tissue injury during septic inflammation. Collectively, our findings suggest that m7G modification enhances the stability of Sarm1 mRNA, thereby resulting in NAD+ imbalance in macrophages, indicating that METTL1 may serve as a potential therapeutic target for systemic inflammation.
    DOI:  https://doi.org/10.1126/sciimmunol.adv4810
  16. Adv Sci (Weinh). 2025 Sep 06. e09952
      Mitigating myocardial ischemia-reperfusion (IR) injury is essential for enhancing the success of heart transplantation (HT) and improving patient outcomes. During HT, infiltrating neutrophils are influenced and regulated by various other cell types, contributing to myocardial IR injury through the excessive release of neutrophil extracellular traps (NETs). Nonetheless, the precise mechanisms underlying the interactions between neutrophils and other non-cardiomyocytes remain largely unexplored. Single-cell RNA sequencing is employed to characterize the cellular landscape and to explore the crosstalk between neutrophils and other non-cardiomyocytes. The role of AT-rich interactive domain-containing protein 3A (ARID3A) during HT is further examined using myeloid-specific ARID3A-knockout mice. Molecular docking analyses are conducted to identify the target of 4-octyl itaconate (4-OI). These results reveal that M1 macrophages recruited during the reperfusion of HT promote NETs formation and myocardial IR injury through THBS1/CD47 axis, whereas CD47 induces NETosis by activating the p38 MAPK signaling. Exogenous administration of 4-OI specifically inhibits ARID3A in macrophages, thereby suppressing NETosis and alleviating myocardial IR injury. These findings indicate that THBS1/CD47 signaling is a critical bridge mediating the interaction between M1 macrophages and NETs-associated neutrophils, and identify 4-OI as a promising therapeutic candidate for the treatment of myocardial IR injury following HT.
    Keywords:  heart transplantation; intercellular crosstalk; ischemia‐reperfusion injury; neutrophil extracellular traps (NETs); single‐cell RNA sequencing
    DOI:  https://doi.org/10.1002/advs.202509952
  17. Proteomics. 2025 Sep 02. e70030
      Innate immune signaling relies heavily on phosphorylation cascades to mount effective immune responses. Although traditional innate immune signaling cascades following TLR4 stimulation have been investigated through a temporally quantitative phosphoproteomic lens, far fewer studies have applied these methods to distinct signaling following the inflammasome trigger leading to IL-1β release. Here, we conducted time-resolved phosphoproteomic profiling to investigate kinase signaling downstream of the inflammasome trigger nigericin. We found that nigericin induces rapid and potent alterations in the phosphorylation landscape where immune-related signaling, mitogen-activated protein kinases (MAPKs), and PKC signaling are prevalent. We also found significant evidence of phospho-modified metabolic cascades, suggesting that phosphosignaling plays a role in previously described immunometabolic regulation. These signaling events preceded robust phosphorylation of DNA damage and chromatin reorganization proteins before pyroptotic rupture. Lastly, by performing temporal clustering of phospho-dynamics, we revealed novel ontology-level shifts in phosphosignaling cascades following nigericin treatment that highlight abrupt changes in cellular behavior during early and late intracellular inflammatory events. SUMMARY: Protein phosphorylation is critical to convey innate immune signaling information to specific effector arms of the cellular immune response. This study focuses on characterizing phosphoproteomic alterations stemming from the inflammasome trigger nigericin. By gaining a deeper understanding of global kinase phosphodynamics in response to inflammasome activation, we aim to identify novel pharmacological targets to treat chronic inflammatory diseases driven by inflammasome-dependent IL-1β release.
    Keywords:  MAPK; inflammasome; inflammation; phosphoproteomics; pyroptosis
    DOI:  https://doi.org/10.1002/pmic.70030