bims-traimu Biomed News
on Trained immunity
Issue of 2025–12–14
eighteen papers selected by
Yantong Wan, Southern Medical University



  1. Curr Allergy Asthma Rep. 2025 Dec 09. 25(1): 59
       PURPOSE OF REVIEW: Asthma represents a major global health burden, with incidence increasing substantially over the past 50 years. Epidemiologic studies have linked this rise in asthma to escalating air pollution; however, the biological mechanisms remain undefined. Recent evidence has highlighted a role for innate immune memory, or trained immunity, in asthma pathogenesis; however, the contribution of air pollution remains unclear. This review synthesizes emerging findings on how air pollution shapes trained immunity in asthma and related inflammatory lung diseases.
    RECENT FINDINGS: Air pollutants increase gene acetylation and methylation and disrupt innate immune cell metabolism. These immune mediated changes are consistent with features of trained immunity, a process that remains largely underexplored. Collectively, these data support the concept that air pollution imprints long-lasting epigenomic and immunometabolic changes on innate immune cells, thereby contributing to asthma susceptibility and severity. Future studies will focus on mechanistic investigations to further elucidate how pollutants dysregulate innate immune memory.
    Keywords:  Asthma; Ozone; Particulate matter; Trained immunity
    DOI:  https://doi.org/10.1007/s11882-025-01240-7
  2. Front Immunol. 2025 ;16 1669054
      Innate immune memory, traditionally underappreciated in contrast to adaptive immunity, is now recognized as a critical component of host defense, particularly in the context of sepsis and sterile inflammatory injury. Recent advances have identified a central role for metabolic and epigenetic reprogramming in driving trained immunity (TRIM), where monocytes, macrophages, and other innate cells develop enhanced or tolerized responses to secondary stimuli. This review synthesizes current knowledge of how damage-associated molecular patterns (DAMPs), including oxidized LDL, HMGB1, heme, urate crystals, and mitochondrial DNA, serve as potent inducers of immunometabolic rewiring, often through the mTOR/HIF-1α axis or alternative pathways such as SYK signaling. We highlight distinct epigenetic mechanisms, such as enhancer priming via H3K4me1/H3K27ac, and metabolic shifts like the Warburg effect, succinate accumulation, and fatty acid synthesis, that define the trained or tolerized states. Particular attention is given to the relevance of these mechanisms in the pathophysiology of sepsis, burns, trauma, and other critical illnesses where persistent DAMP exposure may sustain maladaptive inflammation or immunosuppression. We review data linking central (stem cell-level) and peripheral reprogramming to long-term immune dysfunction in various inflammatory disease models, and explore how DAMPs intersect with PAMPs to shape the immune trajectory. Finally, we identify pressing gaps in the field, including the need for standardized TRIM models, validated biomarkers of innate memory, and mechanistic clarity on mitochondrial DAMPs in immune tolerance. These insights provide a foundation for future therapeutic strategies aimed at modulating trained immunity to improve outcomes in critically ill patients.
    Keywords:  DAMPs; epigenetics; innate immunity; innate training; trauma
    DOI:  https://doi.org/10.3389/fimmu.2025.1669054
  3. JCI Insight. 2025 Dec 09. pii: e193686. [Epub ahead of print]
      Infection leads to durable cell-autonomous changes in hematopoietic stem and progenitor cells (HSPCs), resulting in production of innate immune cells with heightened immunity. The mechanisms underlying this phenomenon, termed central trained immunity, remain poorly understood. We hypothesized that infection induces histone modifications leading to changes in chromatin accessibility that are conserved during differentiation from HSPCs to myeloid progenitors and monocytes. We conducted genome-wide surveillance of histone marks H3K27ac and H3K4me3 and chromatin accessibility in hematopoietic stem cells, multipotent progenitor 3, granulocyte-monocyte progenitors, monocytes and macrophages of naïve and Mycobacterium avium infected mice. Interferon signaling pathways and related transcription factor binding motifs including IRFs, NF-κB, and CEBP showed increased activating histone marks and chromatin accessibility across cell types. However, histone marks and increased chromatin accessibility were conserved at only a few loci, notably Irf1 and Gbp6. Knock out of IRF1 disrupted enhanced mitochondrial respiration and bacterial killing in human monocyte cell lines, while GBP6 KO monocyte cell lines showed dysregulated mitochondrial respiration. In summary, this study identifies IRF1 and GBP6 as two key loci at which infection-induced systemic inflammation leads to epigenetic changes that are conserved from HSPCs to downstream monocytes, providing a mechanistic avenue for central trained immunity.
    Keywords:  Epigenetics; Hematology; Immunology; Innate immunity; Memory
    DOI:  https://doi.org/10.1172/jci.insight.193686
  4. J Control Release. 2025 Dec 05. pii: S0168-3659(25)01103-4. [Epub ahead of print] 114489
      Given the challenge of antigen selection in universal vaccine development, pathogenic microorganisms involved in diverse disease progressions have emerged as promising targets. Certain components of their glycocalyx can induce trained immunity, making them highly attractive for universal vaccine construction. Considering the limitations of current glycocalyx-mimicking strategies based on single glycans, we propose and construct host-pathogen hybrid glycocalyx-mimicking lipid nanoparticles incorporating endogenous sialic acid and exogenous lipopolysaccharide (SA-LPS@LNPs). By tuning the SA-to-LPS ratio, we balanced "self" and "non-self" features, enabling SA-LPS@LNPs to achieve efficient immune recognition, targeted uptake, and autonomic lysosomal escape, thus promoting trained immunity. SA-LPS@LNPs exhibited significant therapeutic efficacy in both solid tumors and liquid tumors (acute myeloid leukemia, AML), inhibited tumor metastasis. A 100 % cure rate in early-stage AML and improved survival after tumor rechallenge, with significantly improved survival in sepsis (87.5 % vs. 0 %-25 %) were observed in SA-LPS@LNPs group. We propose the Enhanced Level Evaluation Via Anatomic Tissue Weight (ELEVATE) index for quantifying anti-metastatic effects more scientifically. Besides, SA-LPS@LNPs provided prophylactic protection against sepsis in both healthy and tumor-bearing mice. To our knowledge, this is the first study to introduce the hybrid glycocalyx-mimicking concept in nanoparticle vaccine design, offering a new perspective for both glycocalyx-based formulation strategies and the advancement of universal vaccines.
    Keywords:  Hybrid glycocalyx mimicking; Lipid nanoparticles; Lipopolysaccharide; Trained immunity; Universal vaccine
    DOI:  https://doi.org/10.1016/j.jconrel.2025.114489
  5. J Vet Med Sci. 2025 Dec 05.
      Trained immunity (TI) is a memory-like response of the mammalian innate immune system. However, a convenient assay system for measuring TI is not yet available. Insects possess only innate immunity and therefore have the potential for evaluating innate immunity-enhancing vaccines or chemicals. In this study, larvae of the silkworm (Bombyx mori) were injected with heat-killed (HK) Escherichia coli to induce immune priming, the insect counterpart of mammalian TI, which was confirmed with reduction in the infected Bombyx mori nucleopolyhedrovirus copy numbers, and gene expression was analyzed by RNA-seq and reverse transcription quantitative PCR. Among the genes with significant expression changes, B. mori homologs of known human TI marker genes were identified. Changes in the expression of some of these marker genes in response to HK E. coli were also confirmed in mouse macrophage cell lines. Although B. mori responses were results of the insect immune priming and not equivalent to the mammalian TI, these results suggest the potential use of B. mori for detecting innate immunity-enhancing agents for mammals.
    Keywords:  assay system; gene expression; marker gene; silkworm; trained immunity
    DOI:  https://doi.org/10.1292/jvms.25-0535
  6. Nat Commun. 2025 Dec 13.
      Inhaled opportunistic pathogens such as Pseudomonas aeruginosa actively modify gene expression to meet the challenges of a new environment. In the infected airway the bacteria must respond to the immunometabolite itaconate, which is abundantly produced by macrophages and has anti-inflammatory and anti-oxidant functions that protect the host from airway damage and causes toxicity to bacteria. As a dicarboxylate that targets cysteine residues, itaconate can modify both bacterial and host proteins often altering metabolic activity. We demonstrate that itaconate promotes a global metabolic response in P. aeruginosa by enhancing the activity of the major alternative transcription factor RpoN. Itaconate is actively transported into the bacteria, induces σ54 rpoN expression and covalently binds cysteine residues 218 and 275 on RpoN helping to neutralize its toxicity. The S-itaconated RpoN exhibits a gain of function driving increased glucose catabolism and enhanced utilization of the bioenergetically efficient Entner-Doudoroff pathway. Thus, the accumulation of itaconate in the infected airway promotes the adaptation of P. aeruginosa to the lung by optimizing its metabolic activity and ability to cause pneumonia.
    DOI:  https://doi.org/10.1038/s41467-025-67153-1
  7. J Clin Immunol. 2025 Dec 10.
       PURPOSE: Long COVID (LC) is a long-term debilitating disease of which the exact pathophysiology is unknown. A dysregulated immune response resulting in hyperresponsive immune cells is hypothesized as a key mechanism in the development of LC. Several studies suggest that acute infections can leave lasting epigenetic changes, which result in heightened immune reactivity. Upon stimulation, these primed immune cells may exhibit exaggerated responses. This form of epigenetic memory can contribute to altered immune dynamics, particularly in response to induction of type I Interferons (IFN-I) pathway activation using a viral mimic. Therefore, we investigated if LC patients exhibit a hyperresponsive response towards viral mimics in comparison with healthy controls (HC).
    METHODS: PBMCs of two distinct LC cohorts, characterized by a different disease course and duration, were collected and transfected using Lyovec with the cGAS and RIG-I agonists G3-YSD and 3p-RNA followed by measurement of IFN-I bioactivity with a reporter cell line.
    RESULTS: Transfection of PBMCs of LC patients with the cGAS and RIG-I agonists resulted in increased IFN-I bioactivity in comparison with HC. Unsupervised hierarchical clustering revealed two distinct clusters, each predominantly composed of either patients or HC. In addition, a moderate correlation between RIG-I stimulation with 3p-RNA and fatigue severity scores was found.
    CONCLUSION: These data show a hyperresponsive phenotype of immune cells of LC patients upon stimulation with viral mimics. The current availability of biologicals and small molecule inhibitors that interfere with aberrant IFN-I pathway activation underscores the importance of pursuing future investigations into this phenomenon.
    Keywords:  Epigenetic memory; Fatigue; Innate immune memory; Long COVID; PASC; RIG-I; Type I interferon; cGAS
    DOI:  https://doi.org/10.1007/s10875-025-01969-w
  8. Cell Death Discov. 2025 Dec 10.
      Endothelial dysfunction-driven vascular inflammation underlies sepsis and atherosclerosis. Piezo1 serves as a central mediator for endothelial mechanotransduction and inflammatory homeostasis. Nevertheless, the transcriptional pathways linking mechanical sensing to anti-inflammatory protection and the exact composition of its downstream signaling cascade remain incompletely resolved. Here, we identify BHLHE40 as an endothelial mechanosensitive transcription factor induced by Piezo1 that coordinates ferroptosis resistance and inflammation suppression. Mechanistically, shear stress activates Piezo1, triggering Ca²⁺ influx and calcineurin-dependent NFAT2 nuclear translocation. NFAT2 recruits HDAC1 to form a transcriptional complex that directly drives BHLHE40 expression. BHLHE40 then binds the SLC7A11 promoter, upregulating this cystine transporter to inhibit ferroptosis. Rescued mitochondrial integrity, reduced ROS, and reversed lipid peroxidation demonstrated this phenomenon. Crucially, mice with endothelial-specific BHLHE40 overexpression attenuate LPS-induced lung vascular leakage, neutrophil infiltration, and pro-inflammatory cytokine release. Our work establishes the Piezo1/Ca²⁺/calcineurin/NFAT2-HDAC1/BHLHE40/SLC7A11 axis as a master mechanotransduction pathway that transcriptionally maintains endothelial homeostasis.
    DOI:  https://doi.org/10.1038/s41420-025-02909-8
  9. Cell Death Discov. 2025 Dec 08.
      Sepsis-induced acute lung injury (ALI) involves a complex interplay between immune cells and the pulmonary endothelium. However, the molecular regulators that coordinate this interaction remain poorly defined. In a murine sepsis model, we identified a subset of lung-resident macrophages characterized by robust IL-1β expression as pivotal contributors to lung damage. Single-cell RNA sequencing (scRNA-seq) delineated a distinct IL-1β⁺ macrophage population with pronounced pro-inflammatory transcriptional features and enhanced endothelial communication. These macrophages exhibited intensified ligand-receptor interactions with pulmonary endothelial cells, corresponding with elevated vascular leakage and histopathological evidence of injury. Immunoassays, Western blotting, and histopathology confirmed IL-1β upregulation during lung injury. Furthermore, metabolomics and in vitro co-culture experiments demonstrated that IL-1β impairs endothelial integrity and modulates metabolic activity. This study reveals a novel immune-metabolic axis whereby IL-1β+ macrophages orchestrate endothelial dysfunction and tissue injury in sepsis. Our findings highlight IL-1β as a potential therapeutic target for mitigating ALI in septic patients.
    DOI:  https://doi.org/10.1038/s41420-025-02868-0
  10. J Immunol. 2025 Dec 12. pii: vkaf317. [Epub ahead of print]
      Human macrophages (MΦs) reside in tissues and develop tissue-specific identities. While studies in mice have identified molecular signatures for site-specific MΦ differentiation, less is known about the transcriptional profiles of human MΦs in distinct sites, including mucosal tissues and lymphoid organs during homeostasis and activation. Here, we use multimodal single-cell sequencing and ex vivo stimulation assays to define tissue signatures for populations of human MΦs isolated from lungs, small intestine, spleen, bone marrow, and lymph nodes obtained from individual organ donors. Our results reveal distinct tissue-adapted gene and protein profiles of metabolic, adhesion, and immune interaction pathways, which are specific to MΦs and not monocytes isolated from the same sites and exhibit homology to murine MΦs from the same sites. Tissue-adapted MΦs remained responsive to polarizing cytokine stimuli ex vivo, with upregulation of expected transcripts and secreted proteins, while retaining tissue-specific profiles. Patterns of chromatin accessibility in tissue MΦs identified from single-nucleus assay for transposase-accessible chromatin by sequencing reflected gene expression signatures and indicate that differential utilization of transcription factors may drive stable tissue-adapted profiles. Together, our findings show how human MΦ identity is coupled to their site of residence for mucosal and lymphoid organs and is intrinsically maintained during activation and polarization.
    Keywords:  gastrointestinal tract; human; lung; monocytes/macrophages; single-cell sequencing
    DOI:  https://doi.org/10.1093/jimmun/vkaf317
  11. J Neuroinflammation. 2025 Dec 10.
      Acute neuroinflammation rapidly activates brain immune responses, but its lasting effects on microglia are unclear. Using systemic LPS administration and LCMV-Armstrong infection, we found that blood-brain barrier disruption and cytokine shifts resolved within 30 days, yet microglial recovery was incomplete-marked by persistent numerical loss and an IFN-γ-low phenotype in the LPS model and reduced relative abundance in the LCMV model. Single-cell RNA sequencing revealed sustained transcriptional alterations, including disease-associated microglia (DAM) features and a distinct recovery-biased population. These acute signatures overlapped with profiles from Alzheimer's model mice and were enriched in human microglia from multiple sclerosis, Alzheimer's disease, and other neuroinflammatory conditions. Although our observation period was shorter than the chronic course of these diseases, the persistence of disease-like microglial states suggests that transient inflammation can prime the brain for long-term vulnerability. Targeting this primed state may offer new strategies to prevent or mitigate neurodegenerative pathology.
    Keywords:  Acute neuroinflammation; Microglia; Transcriptomic reprogramming
    DOI:  https://doi.org/10.1186/s12974-025-03572-7
  12. Proc Natl Acad Sci U S A. 2025 Dec 16. 122(50): e2525296122
      Innate immunity provides the critical first line of defense against infection and sterile triggers. Inflammatory cell death is a key component of the innate immune response to clear pathogens, but excessive or aberrant cell death can induce inflammation, cytokine storm, and pathology, making it a central molecular mechanism in inflammatory diseases. Alcohol-associated liver disease (ALD) is one such inflammatory disease, and the specific innate immune mechanisms driving pathology in this context remain unclear. Here, by leveraging RNA-seq and protein expression analyses in tissues from clinical samples, we identified increased expression of the innate immune sensor ZBP1 in patients with ALD. ZBP1 expression correlated with ALD progression in patients and that ethanol induced ZBP1-dependent lytic cell death, PANoptosis, in immune (macrophages, monocytes, and Kupffer cells) and nonimmune cells (hepatocytes). Mechanistically, the interferon regulatory factors (IRFs) IRF9 and IRF1 upregulated basal ZBP1 expression. Activation of ZBP1 led to PANoptosis via caspase-8 and cell membrane rupture through NINJ1, independent of gasdermin D, gasdermin E, and MLKL. In mouse models of ALD, ZBP1-deficient mice were significantly protected from disease pathology and liver damage. Furthermore, the expressions of ZBP1 and NINJ1 were upregulated in both liver and serum samples from patients with ALD, implicating these molecules as potential biomarkers. Overall, our findings establish the critical role of the IRF-ZBP1-caspase-8-NINJ1 axis in driving inflammatory cell death, PANoptosis, suggesting that targeting these molecules will have therapeutic potential in ALD and other inflammatory conditions.
    Keywords:  ZBP1; alcohol; alcohol-associated liver disease; caspase; inflammation
    DOI:  https://doi.org/10.1073/pnas.2525296122
  13. JCI Insight. 2025 Dec 08. pii: e177004. [Epub ahead of print]10(23):
      Mitochondrial dysfunction is a major mechanism of acute kidney injury (AKI), and increased circulating interleukin 6 (IL-6) is associated with systemic inflammation and death due to sepsis. We tested whether kidney mitochondrial DNA (mtDNA) contributes to IL-6 release in sepsis-associated AKI via Toll-like receptor 9 (TLR9). In a murine model of sepsis via cecal ligation and puncture (CLP), we used next-generation sequencing of plasma mtDNA to inform the design of optimal target sequences for quantification by droplet digital PCR, and to identify single-nucleotide polymorphisms (SNPs) to infer tissue origin. We found significantly higher concentrations of plasma mtDNA after CLP versus shams and that plasma mtDNA SNPs matched kidney SNPs more than other organs. Kidney mtDNA contributed directly to IL-6 and mtDNA release from dendritic cells in vitro and kidney mitochondria solution led to higher IL-6 concentrations in vivo. IL-6 release was mitigated by a TLR9 inhibitor. Finally, plasma mtDNA was significantly higher in septic patients with AKI compared with those without AKI and correlated significantly with plasma IL-6. We conclude that AKI contributes to increased circulating IL-6 in sepsis via mtDNA release. Targeting kidney mitochondria and mtDNA release are potential translational avenues to decrease mortality from sepsis-associated AKI.
    Keywords:  Inflammation; Innate immunity; Mitochondria; Nephrology
    DOI:  https://doi.org/10.1172/jci.insight.177004
  14. Phytomedicine. 2025 Nov 29. pii: S0944-7113(25)01276-0. [Epub ahead of print]150 157641
       BACKGROUND: Acute Lung Injury (ALI) significantly contributes to mortality in septic patients and lacks specific therapies. Although multicenter trials have confirmed that Xuebijing injection (XBJ) reduces sepsis mortality, the mechanisms underlying its anti-inflammatory effects remain unclear.
    PURPOSE: This study aims to investigate the pharmacological efficacy of XBJ against ALI.
    METHODS: Histopathology and RT-PCR were used to evaluate the therapeutic effects of XBJ. Monocyte phenotype, viability, and function were assessed by flow cytometry and immunofluorescence. Western blotting was performed to analyze inflammatory, ferroptosis, and cell survival-related signaling molecules.
    RESULTS: By employing cecal ligation and puncture (CLP) and LPS-induced murine sepsis models in WT, chemokine C-C-motif receptor 2 (CCR2)-/-, and nuclear factor erythroid 2-related factor 2 (Nrf2)-/- mice, we demonstrated that XBJ ameliorated sepsis-induced ALI by specifically reducing CCR2hi monocytes in the lungs without affecting neutrophils or the CCR2lo monocytes, thereby blocking the shift of pulmonary macrophages toward the M1 (classically activated macrophages) phenotype. This reduction was independent of the bone marrow mobilization of monocytes or improved tight junction protein expression. Mechanistically, XBJ triggers ferroptosis in LPS-stimulated monocytes, suppressing their pro-inflammatory responses, including chemotaxis, cytokine secretion, and differentiation into pro-inflammatory macrophages. We demonstrated that ferroptosis is a critical mechanism underlying the anti-inflammatory function of XBJ as evidenced by its effective reversal with the ferroptosis inhibitor ferrostatin-1. The Nrf2/heme oxygenase-1 (HO-1) axis, a central regulator of ferroptosis, was closely associated with the anti-inflammatory action of XBJ. In Nrf2-knockout models, the ferroptosis-inducing effect of XBJ on LPS-stimulated monocytes was abolished; co-administration of hemin (a pharmacological HO-1 inducer) and XBJ inhibited LPS-induced monocyte ferroptosis in vitro, counteracted the reduction of CCR2hi pro-inflammatory monocyte infiltration mediated by XBJ injection, and exacerbated pulmonary pro-inflammatory cytokine production in vivo.
    CONCLUSION: This study confirms that XBJ treats sepsis by inducing ferroptosis in pro-inflammatory monocytes, identifies monocytes as key cellular targets in ALI, and underscores the translational potential of targeting ferroptosis to regulate dysregulated inflammation in sepsis.
    Keywords:  Acute Lung Injury; CCR2; Ferroptosis; Monocyte; Nrf2; XBJ
    DOI:  https://doi.org/10.1016/j.phymed.2025.157641
  15. Autophagy. 2025 Dec 08.
      Accumulation of lipid droplets (LDs) in cardiomyocytes contributes to developmentof septic cardiomyopathy, a fatal complication of critical illness in patients.Lipophagy is a selective autophagic mechanism for LD degradation. This processis inhibited by MTOR, but is activated by PNPLA2 via its binding with LC3-II toform LD-containing autophagosomes. However, optimum lipophagic interventions tomanage septic cardiomyopathy have not been developed, thus furtherinvestigation is required to identify novel regulators of lipophagy in theseptic heart. HSPA12A (heat shockprotein 12A) encodes an atypical member of the HSPA/HSP70family. Here, we report that sepsis decreased HSPA12Aexpression in cardiomyocytes, whereas cardiomyocyte-specific HSPA12Aoverexpression aggravated sepsis-induced cardiomyocyte death and cardiacdysfunction in mice. Notably, HSPA12A promoted sepsis-induced LD accumulationin cardiomyocytes. By contrast, HSPA12A inhibited lipophagy in septiccardiomyocytes, as reflected by a decreased level of LD-containing autophagosomes,a reduced content of LC3-II, and an increased level of SQSTM1/p62. In-depthmolecular analysis revealed that HSPA12A increased phosphorylation of MTOR andthus its binding to PNPLA2 on LDs. MTOR thereby competed against LC3-II inbinding with PNPLA2 to suppress LD-containing autophagosome formation subsequentlyimpairing lipophagy and ultimately promoting cardiomyocyte death to exaggerate septiccardiomyopathy. We demonstrated that MTOR competed against LC3-II in bindingwith PNPLA2 to inhibit lipophagy and also identified HSPA12A as a driver ofthis competition with MTOR to impair lipophagy for exaggerating septic cardiomyopathy. Strategiesthat inhibit HSPA12A in cardiomyocytes might be a potential therapeuticintervention for septic cardiomyopathy.
    Keywords:  Cardiomyocyte; HSPA12A; MTOR; PNPLA2; lipophagy; septic cardiomyopathy
    DOI:  https://doi.org/10.1080/15548627.2025.2600895
  16. JCI Insight. 2025 Dec 08. pii: e185743. [Epub ahead of print]10(23):
      Sepsis is a life-threatening organ dysfunction caused by a dysregulated host response to infection. During early sepsis, kinins are released and bind to B1 (BDKRB1) and B2 (BDKRB2) bradykinin receptors, but the involvement of these receptors in sepsis remains incompletely understood. This study demonstrated that the genetic deletion of Bdkrb2 had no significant impact on sepsis induced by cecal ligation and puncture (CLP) compared to wild-type (WT) mice. In contrast, Bdkrb1-/- mice subjected to CLP exhibited decreased lethality and bacterial load, associated with an increased influx of neutrophils into the peritoneal cavity, compared with WT mice. Neutrophils from CLP-Bdkrb1-/- mice partially restored CXCR2 expression and reduced the upregulation of P110γ observed in WT CLP neutrophils. Pharmacologic inhibition of BDKRB1 combined with imipenem treatment substantially improved survival compared with antibiotic therapy alone. In human neutrophils, stimulation with LPS led to the upregulation of BDKRB1 expression, and antagonism of BDKRB1 restored neutrophil migration in response to CXCL8. These findings identify BDKRB1 as an important modulator of neutrophil dysfunction in sepsis and a promising therapeutic target whose inhibition improves bacterial clearance, restores neutrophil migration, and increases the efficacy of antibiotic treatment.
    Keywords:  Bacterial infections; Cell migration/adhesion; Inflammation; Microbiology; Neutrophils
    DOI:  https://doi.org/10.1172/jci.insight.185743
  17. JAMA. 2025 Dec 08.
    ImmunoSep Study Group
       Importance: Sepsis is heterogeneous, and the optimal strategy for tailoring immunotherapy is uncertain.
    Objective: To investigate whether precision immunotherapy guided by the presence of macrophage activation-like syndrome or sepsis-induced immunoparalysis improves organ dysfunction by day 9.
    Design, Setting, and Participants: A randomized, double-blind, double-dummy, placebo-controlled clinical trial conducted in 6 countries. Patients with sepsis, defined by Sepsis-3, were included if they had community-acquired or hospital-acquired pneumonia or ventilator-associated pneumonia or bacteremia and sepsis and had displayed either macrophage activation-like syndrome (blood ferritin >4420 ng/mL) or sepsis-induced immunoparalysis (blood ferritin ≤4420 ng/mL and <5000 human leukocyte antigen DR receptors on CD45/CD14 monocytes). The first patient was enrolled August 5, 2021, and the last follow-up, April 29, 2024.
    Interventions: Eligible patients were randomized to receive standard care and precision immunotherapy or standard care and placebo. Those in the precision immunotherapy group with macrophage activation-like syndrome received anakinra intravenously (IV) and placebo subcutaneously, and those with sepsis-induced immunoparalysis received subcutaneous recombinant human interferon gamma and IV placebo. Those in the placebo group received both IV and subcutaneous placebo. Treatment was administered for up to 15 days.
    Main Outcomes and Measures: The primary end point was a decrease of at least 1.4 points in the mean Sequential Organ Failure Assessment (SOFA) score from baseline by day 9. The SOFA score evaluates 6 organ systems, ranging from 0, no dysfunction, to 4, failure, and the total score ranges from 0, normal, to 24, most severe form of multiorgan failure. Key secondary outcomes included 28-day mortality.
    Results: Of 672 patients assessed for eligibility, 281 were randomized and 276 were included in the primary analysis population (mean [SD] age, 70 [13] years; 93 females [33.7%]; median baseline SOFA score, 9 [IQR, 7-11]). The SOFA decrease end point was attained by 46 of 131 patients (35.1%) in the precision immunotherapy group and by 26 of 145 patients (17.9%) in the placebo group (difference, 17.2% [95% CI, 6.8% to 27.2%]; P = .002). Mortality at 28 days was not statistically significantly different between groups. A total of 1069 serious treatment-emergent adverse events (88.8%) were reported; increased incidence of anemia was noted in the anakinra group; and hemorrhage in the recombinant human interferon gamma group.
    Conclusions and Relevance: Among patients with sepsis, precision immunotherapy targeting macrophage activation-like syndrome and sepsis-induced immunoparalysis improved organ dysfunction by day 9 compared with placebo.
    Trial Registration: ClinicalTrials.gov Identifier: NCT04990232.
    DOI:  https://doi.org/10.1001/jama.2025.24175
  18. Cell Chem Biol. 2025 Dec 09. pii: S2451-9456(25)00354-X. [Epub ahead of print]
      The ability of innate immune cells to sense and respond to their physical environment is essential for regulating immune function. Macrophages, key players in inflammation, pathogen defense, and tissue repair, are influenced not only by biochemical cues but also by the mechanical properties of their microenvironment, including extracellular matrix stiffness, shear stress, and cyclic stretch. While the effects of soluble factors, such as cytokines on macrophage behavior are well characterized, the mechanisms underlying macrophage mechanotransduction remain poorly understood. This review synthesizes current understanding of how distinct mechanical forces shape macrophage activation, migration, polarization, and cytokine production. We also explore emerging insights into the roles of mechanosensitive ion channels (e.g., Piezo1, TRPV4), integrins, and cytoskeletal dynamics in transducing mechanical signals into pro- or anti-inflammatory responses. A deeper understanding of these pathways may uncover new therapeutic targets for treating cardiovascular diseases, including atherosclerosis.
    Keywords:  cyclic stretch; macrophages; mechanical forces; mechanotransduction; shear stress; stiffness
    DOI:  https://doi.org/10.1016/j.chembiol.2025.11.004