bims-bac4me Biomed News
on Microbiome and trained immunity
Issue of 2025–10–05
forty-two papers selected by
Chun-Chi Chang, Lunds universitet



  1. mSphere. 2025 Sep 30. e0049325
      Nasal colonization by Staphylococcus aureus or Streptococcus pneumoniae is associated with an increased risk of infection by these pathobionts, whereas nasal colonization by Dolosigranulum species is associated with health. Human nasal epithelial organoids (HNOs) differentiated at air-liquid interface (ALI) physiologically recapitulate human nasal respiratory epithelium with a robust mucociliary blanket. Due to their natural stem-like properties, HNO lines are a long-term experimental resource that offers genetic diversity based on the different donors. To develop HNOs as a new model system for bacterial nasal colonization, we reproducibly monocolonized HNOs differentiated at ALI with S. aureus, S. pneumoniae, or Dolosigranulum pigrum for up to 48 h with varying kinetics across species. HNOs tolerated bacterial monocolonization with localization of bacteria to the mucus layer and with minimal cytotoxicity compared to uncolonized HNOs. Human nasal epithelium exhibited both species-specific and general cytokine responses, without induction of type I interferons, which is consistent with colonization rather than infection. Only live S. aureus colonization robustly induced epithelial cell production of interleukin-1 family cytokines, suggestive of inflammasome signaling. D. pigrum and live S. aureus decreased CXCL10, whereas S. pneumoniae increased CXCL11, chemokines involved in antimicrobial responses to both viruses and bacteria. Overall, HNOs are a new model system for uncovering microbe-epithelial cell dynamics at the human nasal mucosa.
    IMPORTANCE: Human nasal microbiota often includes highly pathogenic members, many of which are antimicrobial resistance threats, e.g., methicillin-resistant Staphylococcus aureus and drug-resistant Streptococcus pneumoniae. Preventing colonization by nasal pathobionts decreases infections and transmission. In contrast, nasal microbiome studies identify candidate beneficial bacteria that might resist pathobiont colonization, e.g., Dolosigranulum pigrum. Learning how these microbionts interact with the nasal epithelium and identifying new means to reduce pathobiont colonization are key goals in the field. As a tool to advance this research, we developed human nasal epithelial organoids (HNOs) differentiated at an air-liquid interface as a new model system of bacterial nasal colonization. HNOs accurately represent the mucosal surface of the human nasal passages, enabling exploration of bacterial-epithelial interactions, which is important since the epithelium is an instigator of the initial innate immune response to bacteria. Here, we identified differential epithelial cytokine responses to these three bacteria, setting the stage for future research.
    Keywords:  Dolosigranulum pigrum; HNO; MRSA; Staphylococcus aureus; Streptococcus pneumoniae; bacterial colonization; epithelial innate immune response; human nasal organoids; nasal colonization
    DOI:  https://doi.org/10.1128/msphere.00493-25
  2. Elife. 2025 Oct 02. pii: e106688. [Epub ahead of print]14
      The skin is frequently subjected to minor mechanical insults that may compromise its barrier integrity and permit the entry of pathogens. Therefore, the immune system of the skin needs to rapidly balance antimicrobial defense with tissue repair. To maintain homeostasis, the skin relies both on acute immune defenses and on mechanisms of innate memory or trained immunity. This enhanced inflammatory response to a second challenge has been well characterized in bone marrow cells, such as monocytes, monocyte-derived macrophages, and stem cells. Yet, the specific memory responses in skin-resident immune cells remain less understood. Importantly, the common skin colonizer Staphylococcus aureus has been identified as a potent inducer of trained immunity, triggering both metabolic and epigenetic changes at local sites such as the skin, and centrally in the bone marrow. This review explores the emerging understanding of trained immunity in the skin, that is how infection-driven cellular processes induce long-lasting immune adaptation and modulate skin barrier integrity.
    Keywords:  Dermis; S. aureus; immunology; inflammation; macrophages; skin; trained immunity
    DOI:  https://doi.org/10.7554/eLife.106688
  3. Exp Mol Med. 2025 Oct 01.
      The immune system has traditionally been divided into innate and adaptive branches, with immunological memory considered a hallmark of adaptive immunity. However, recent studies reveal that innate immune cells can also exhibit memory-like properties, known as trained immunity. This phenomenon involves the long-term functional reprogramming of innate immune cells following exposure to exogenous or endogenous stimuli, mediated by epigenetic and metabolic changes. Trained immunity enhances responses to subsequent unrelated challenges and serves as a protective mechanism against reinfection. Nonetheless, it may also contribute to the development of chronic inflammatory diseases such as autoimmune disorders, allergies and atherosclerosis. Whereas much of the research has focused on pathogen-associated molecular patterns as inducers of trained immunity, emerging evidence highlights that sterile inflammation, driven by damage-associated molecular patterns and lifestyle-associated molecular patterns, can similarly induce this immune adaptation. Here we examine the molecular mechanisms underlying damage-associated molecular pattern- and lifestyle-associated molecular pattern-induced trained immunity and their roles in chronic inflammation. This Review also discusses central trained immunity, characterized by the durable reprogramming of hematopoietic stem and progenitor cells, and its implications in disease progression. Finally, potential therapeutic strategies targeting metabolic and epigenetic pathways are considered. Understanding noninfectious stimuli-induced trained immunity offers new insights into chronic inflammatory disease management.
    DOI:  https://doi.org/10.1038/s12276-025-01542-w
  4. bioRxiv. 2025 Apr 10. pii: 2025.04.10.647998. [Epub ahead of print]
      Staphylococcus aureus is a leading cause of healthcare-associated pneumonia, contributing significantly to morbidity and mortality worldwide. As a ubiquitous colonizer of the upper respiratory tract, S. aureus must undergo substantial metabolic adaptation to achieve persistent infection in the distinctive microenvironment of the lung. We observed that fumC , which encodes the enzyme that converts fumarate to malate, is highly conserved with low mutation rates in S. aureus isolates from chronic lung infections. Fumarate, a pro-inflammatory metabolite produced by macrophages during infection, is regulated by the host fumarate hydratase (FH) to limit inflammation. Here, we demonstrate that fumarate, which accumulates in the chronically infected lung, is detrimental to S. aureus , blocking primary metabolic pathways such as glycolysis and oxidative phosphorylation (OXPHOS). This creates a metabolic bottleneck that drives staphylococcal FH (FumC) activity for airway adaptation. FumC not only degrades fumarate but also directs its utilization into critical pathways including the tricarboxylic acid (TCA) cycle, gluconeogenesis and hexosamine synthesis to maintain metabolic fitness and form a protective biofilm. Itaconate, another abundant immunometabolite in the infected airway enhances FumC activity, in synergy with fumarate. In a mouse model of pneumonia, a Δ fumC mutant displays significant attenuation compared to its parent and complemented strains, particularly in fumarate- and itaconate-replete conditions. Our findings underscore the pivotal role of immunometabolites in promoting S. aureus pulmonary adaptation.
    DOI:  https://doi.org/10.1101/2025.04.10.647998
  5. BMC Microbiol. 2025 Oct 02. 25(1): 621
       BACKGROUND: Staphylococcus epidermidis is a prevalent common microorganism found on the skin of most mammals, including humans. However, S. epidermidis is also known as an opportunistic pathogen that can cause diseases if the skin microbiome becomes skewed to favor its overgrowth. This situation can be worsened if the causative strains are antibiotic-resistant. To explore potential alternatives to replace conventional antibiotics, we aim to use probiotics, specifically Lacticaseibacillus rhamnosus and Lactiplantibacillus plantarum, in controlling the growth, colonization, and biofilm formation of a multidrug-resistant S. epidermidis.
    RESULTS: Multidrug-resistant S. epidermidis was co-cultured with either L. rhamnosus or L. plantarum, and antimicrobial activity was assessed by counting S. epidermidis colonies at various timepoints. Under co-culture conditions, growth of S. epidermidis was decreased over time in the presence of either probiotic, specifically, a significant reduction was observed after 48 hours (p=0.0062). Additionally, S. epidermidis was treated with cell-free culture supernatant (CFCS) collected from probiotics to investigate the mechanism of inhibition. CFCS from 72-hour Lactobacilli cultures significantly inhibited S. epidermidis growth within 4 hours (p<0.05), and no S. epidermidis was detected after 72 hours (p=0.0003) when treated with 50% CFCS. Both CFCSs also reduced biofilm formation and bacterial counts after 24 hours, with complete inhibition by 48 h. Gene expression analysis revealed that exposure to CFCS led to downregulation of S. epidermidis genes associated with metabolism and cell division (arcC, gts, mutS, mur1, and ftsZ). Fluorescence microscopy indicated compromised cell wall integrity, evidenced by reduced HADA fluorescence and increased propidium iodide staining.
    CONCLUSION: L. plantarum and L. rhamnosus possess strong antimicrobial activity against multidrug-resistantS. epidermidis, supporting their potential as alternative strategies to control S. epidermidis colonization.
    Keywords:   Lactobacillus ; Staphylococcus epidermidis ; Antibiotic resistance; Biofilm; Probiotics
    DOI:  https://doi.org/10.1186/s12866-025-04382-w
  6. Immunol Res. 2025 Oct 01. 73(1): 139
      Sepsis, a life-threatening systemic infection, has long been recognized for its immediate risks, but its long-term consequences on health are increasingly evident, particularly in predisposing survivors to chronic cardiometabolic disorders (CMDs) such as atherosclerosis, insulin resistance, and dyslipidemia. Central to this process is trained immunity, where innate immune cells like monocytes, macrophages, and neutrophils undergo long-lasting epigenetic reprogramming after sepsis. This reprogramming, sustained by molecular pathways such as NF-κB, mTOR, and altered lipid metabolism, drives chronic inflammation, oxidative stress, and metabolic dysfunction, contributing to long-term cardiovascular diseases (CVDs) and metabolic disorders post-sepsis. This review explores the key mechanisms through which trained immunity bridges sepsis and CMDs, particularly focusing on epigenetic modifications such as histone acetylation, DNA methylation, and mitochondrial alterations. We discuss how trained immunity enhances immune cell activation, leading to persistent low-grade inflammation, lipid dysregulation, and impaired insulin sensitivity, all of which predispose sepsis survivors to CVDs. Additionally, we highlight potential therapeutic approaches targeting trained immunity, including statins, which reduce inflammation and immune reprogramming; metformin, which restores metabolic balance by activating AMPK and reducing oxidative stress; dimethyl fumarate (DMF), a potent Nrf2 activator that counteracts inflammation; and probiotics, which help restore gut microbiota balance and limit endotoxin-driven inflammation. These therapies offer promising strategies to mitigate long-term metabolic dysfunction and reduce the incidence of CMDs following sepsis. Understanding these mechanisms and developing targeted interventions may ultimately help prevent chronic cardiovascular and metabolic diseases in sepsis survivors and improve long-term outcomes.
    Keywords:  Cardiometabolic disorders; Epigenetic reprogramming; Inflammation; Sepsis; Trained immunity
    DOI:  https://doi.org/10.1007/s12026-025-09698-3
  7. bioRxiv. 2025 Sep 26. pii: 2025.09.24.678325. [Epub ahead of print]
      Staphylococcus aureus ( S. aureus ) is a highly virulent pathogen responsible for chronic infections such as osteomyelitis. Although its interaction with the host immune system has been widely studied, the specific role of inflammasomes in regulating the infection within macrophages remains unclear. We investigated this question using bone marrow-derived macrophages infected with S. aureus and observed a significant reduction in intracellular bacterial load beginning at 18 hours post-infection (hpi), which continued through 96 hpi. Notably, robust activation of the NLRP3 inflammasome-including inflammasome assembly, IL-1β and GSDMD maturation, and pyroptosis-occurred only after 18 hpi. This led us to hypothesize that S. aureus suppresses inflammasome activation during early infection. Supporting this, infected BMDMs failed to respond robustly to LPS and nigericin up to 18 hpi, with partial recovery at later timepoints, suggesting that S. aureus initially inhibits NLRP3 signaling to persist within macrophages but is later counteracted by the host response.
    DOI:  https://doi.org/10.1101/2025.09.24.678325
  8. Immunology. 2025 Oct 01.
      Understanding the innate immune memory induced by adjuvants provides an opportunity to improve vaccine efficacy by inducing nonspecific secondary responses alongside the intended adaptive defence against the target antigen. To understand the consequences of adjuvant-induced immune training, we treated mice with commercially available Sigma Adjuvant System (SAS) and performed functional assays of bone marrow-derived innate immune cells, assessed its functional consequences in vivo, determined the resulting haematopoietic stem and progenitor cell (HSPC) phenotypes, and extensively analyzed the HSPC transcriptome. SAS induced temporal shifts in HSPC frequencies, alterations in the circulating blood profile, and lowered proinflammatory output by macrophages. SAS-induced training caused disparate outcomes in models of inflammation and acute infection. Further, SAS enhanced antibody responses after primary immunisation, that were profoundly altered upon a secondary dose. Integrated transcriptional analysis revealed shifts in HSPCs defined by altered transcription factor activity and lineage-specific shifts in metabolic, epigenetic, myeloid, and kinase genes, resulting in enhanced antimicrobial neutrophil responsiveness and revealing regulators of central training. Together, these results contribute to the understanding of the plasticity and limitations of innate immune training.
    Keywords:  haematopoiesis; macrophage; memory; transcriptomics; vaccination
    DOI:  https://doi.org/10.1111/imm.70047
  9. Nat Microbiol. 2025 Oct;10(10): 2355
      
    DOI:  https://doi.org/10.1038/s41564-025-02151-z
  10. Int Forum Allergy Rhinol. 2025 Sep 30.
       BACKGROUND: Chronic rhinosinusitis (CRS) is a chronic inflammatory disease that is associated with polymicrobial infections, often involving S. aureus and P. aeruginosa. It is unclear whether the polymicrobial context plays a role in exacerbating epithelial damage, inflammation, and resistance to therapy.
    METHODS: S. aureus and P. aeruginosa (n = 3 each) biofilms were established in a Transwell system, followed by the extraction of P. aeruginosa conditioned media and application to an air-liquid interface (ALI) model of human nasal epithelial cells (HNECs). Transepithelial electrical resistance (TEER) and FITC dextran paracellular permeability tests evaluated the epithelial integrity. Colony-forming unit (CFU) counting, protease activity assay, and pyocyanin and pyoverdine quantification were used to test the proliferation and production of virulence factors of the bacteria.
    RESULTS: Cocultures of P. aeruginosa and S. aureus isolated from the same patient reduced HNEC TEER values, had an earlier onset of HNEC barrier disruption, and increased paracellular permeability compared to monocultures of P. aeruginosa. P. aeruginosa proliferation was enhanced, and protease activity increased significantly. The production of pyoverdine increased significantly in the same patient cocultures, while the pyocyanin levels remained unchanged.
    CONCLUSIONS: These results indicate a role of within-host evolution in shaping P. aeruginosa-mediated virulence in the context of polymicrobial biofilms. This supports the need to develop strategies directed at disrupting interspecies synergies that culminate in the formation of polymicrobial biofilms associated with CRS for the purpose of improving disease management and therapeutic efficacy.
    Keywords:  Pseudomonas aeruginosa; Staphylococcus aureus; biofilms; chronic rhinosinusitis; epithelial barrier integrity; polymicrobial infections
    DOI:  https://doi.org/10.1002/alr.70036
  11. mBio. 2025 Sep 30. e0212425
      Staphylococcus aureus bacteremia (SAB) remains a significant clinical burden with high mortality rates. Kupffer cells (KCs) are integral to clearing Staphylococcus aureus (SA) from the bloodstream yet also serve as a pathogenic reservoir, driving persistence and treatment failure. KCs utilize their metabolic plasticity to coordinate successful antimicrobial responses, and this process may be therapeutically targeted. We investigated the immunometabolic effects of antistaphylococcal antibiotics (vancomycin, ceftobiprole, daptomycin, and tedizolid) on KCs relative to clearance of intracellular SA through gene expression analysis and optical metabolic imaging using fluorescence lifetime imaging microscopy. We observed differential effects of antibiotics on KC polarization and metabolic signatures, with ceftobiprole inducing the strongest pro-inflammatory M1 expression profile and shortest mean NAD(P)H fluorescence lifetime in KCs in the absence of infection, and a retained M1 expression profile but longest NAD(P)H lifetime during infection. Daptomycin and tedizolid most effectively cleared intracellular SA, induced significantly longer NAD(P)H lifetimes, and exhibited decreased M1 gene expression. Vancomycin failed to control intracellular infection showing blunted M1 expression without altering NAD(P)H lifetimes. Stratification of NAD(P)H lifetime signals revealed daptomycin significantly increased binding to PDH-like enzymes and nitric oxide synthase 2 compared to other antibiotics. These findings reveal novel insights into antibiotic modulation of KC immunometabolism during SA infection, which may influence treatment outcomes in SAB.IMPORTANCEStaphylococcus aureus bloodstream infection is a leading cause of sepsis and is associated with up to 30% mortality. Despite treatment with guideline-recommended antibiotics, persistent bacteremia develops in one in three patients, which may be attributed in part to survival of bacteria inside liver-resident macrophages known as KCs. Using gene expression and optical metabolic profiling, we demonstrated that antistaphylococcal antibiotics differentially affect KC metabolism and function, thereby contributing to the overall killing of intracellular bacteria. Daptomycin, ceftobiprole, and tedizolid exert distinct effects on KC metabolism that correspond to effective intracellular killing and prompt resolution of inflammatory response. Vancomycin, however, did not affect KC metabolism and was unable to control bacterial growth inside the cells. These findings suggest that choosing antibiotics based on direct antimicrobial activity as well as indirect effects on host immune function could improve treatment outcomes for patients with S. aureus bloodstream infection.
    Keywords:  Kupffer cells; Staphylococcus aureus; antibiotics; bloodstream infections; ceftobiprole; daptomycin; immunometabolism; macrophages; tedizolid; vancomycin
    DOI:  https://doi.org/10.1128/mbio.02124-25
  12. Front Immunol. 2025 ;16 1654034
      The metabolism of immune cells adapts to support the energy demands for their activation, differentiation, and effector functions through a process known as metabolic reprogramming. This metabolic plasticity is influenced by both extrinsic and intrinsic factors, including steroid hormones such as glucocorticoids, androgens, progestogens, and estrogens. These critical mediators modulate immune function and inflammatory responses through genomic and non-genomic regulation of intracellular metabolic pathways, including glycolysis, the tricarboxylic acid cycle, and oxidative phosphorylation. Interestingly, these effects appear to be dependent on cell type, hormonal concentration, and microenvironmental context. Herein, we discuss how steroid hormones regulate inflammation and immunometabolism and summarize recent studies highlighting immunometabolic regulation by steroid hormones as the key driver of their immunomodulatory effects. We also address potential mechanisms contributing to their seemingly dichotomous and context-specific regulation. Understanding the link between steroid hormone signaling, immunometabolism, host defense, chronic inflammation, and immunity will expand our understanding about how biological sex and stress influence the immune system and facilitate more precise therapeutic targeting of immune cell activity to mitigate inflammation- and immune-mediated diseases.
    Keywords:  glycolysis; immune; immunometabolism; inflammation; oxidative phosphorylation; steroid hormones
    DOI:  https://doi.org/10.3389/fimmu.2025.1654034
  13. Int Arch Allergy Immunol. 2025 Oct 01. 1-16
      Dendritic cells (DCs) represent a heterogeneous subpopulation of antigen-presenting cells that bridge the innate and acquired immune responses. Studies have reported that cellular metabolic reprogramming regulates the biological function of DCs, with distinct metabolic profiles characterizing different DC subsets and activation state. Particularly, blood monocyte-derived dendritic cells (mo-DCs) drive the skin inflammation. The differentiation, maturation, metabolism and function of mo-DCs are influenced by microenvironmental factors including pH, mechanical force, and temperature. The paper briefly reviewed the biological role of mo-DCs in skin diseases, focusing on their differentiation and phenotype, disease-associated metabolic adaptations, and the microenvironmental factors that influence their maturation, energy metabolism and function. Future researchers should explore the impact of the skin microenvironment on the metabolism and function of mo-DCs and identify specific targets, which may pave the way for precision medicine methods for treating refractory inflammatory skin diseases.
    DOI:  https://doi.org/10.1159/000547871
  14. Virulence. 2025 Dec;16(1): 2566242
      Neisseria meningitidis encounters a dynamic nutrient landscape during host colonization, which necessitates its metabolic adaptation to different host metabolites such as acetate. Acetate, a short chain fatty acid (SCFA) within the host milieu, found to regulate host defense and inflammation during bacterial infection. In macrophage acetate gets converted into acetyl-CoA to provide energy via TCA cycle. Also, acetate is a crucial metabolic intermediate that takes part in lipid biosynthesis and protein acetylation. Acetate acts as an immunomodulator which improves the bactericidal effect of macrophage by activating the inflammasome. Therefore, to persist in nutrient limited conditions encountered in macrophages pathogens should resort to effective utilization of energy sources. We demonstrate that N. meningitidis can potentially utilize host acetate as a carbon source and regulate its virulence. The utilization of acetate enhanced the survival of N. meningitidis in H2O2 induced oxidative stress which can be correlated with the macrophage infection assay. Moreover, our investigation into underlying mechanism suggests that acetate exposure upregulates bacterial oxidative stress response by significantly increasing catalase and superoxide dismutase activity. We demonstrated that acetate metabolism also upregulated expression of nitric oxide detoxification genes fnr, narQ which mitigate reactive oxygen and nitrogen species produced in macrophages. Therefore, we conclude that bacterial utilization of physiologically relevant host acetate levels represents an important strategy to consume or detoxify macrophage-mediated oxidative stress, thereby facilitating bacterial survival.
    Keywords:  Oxidative stress; acetate; carbon sources; nitric oxide detoxification; reactive oxygen species
    DOI:  https://doi.org/10.1080/21505594.2025.2566242
  15. Placenta. 2025 Sep 22. pii: S0143-4004(25)00700-3. [Epub ahead of print]
      Trained immunity, defined as the epigenetic and metabolic reprogramming of innate immune cells that shapes their subsequent responses, has emerged as a key paradigm in reproductive immunology. In pregnancy, trained immunity can act as a "double-edged sword." Beneficial training of decidual NK cells and macrophages promotes vascular remodeling, tissue repair, and host defense, thereby supporting implantation and placental development. Conversely, dysregulated training triggered by hypoxia, metabolic stress, or infection may sustain chronic inflammation and contribute to preeclampsia, fetal growth restriction, and recurrent pregnancy loss. In this review, we summarize current evidence for both protective and pathogenic roles of trained immunity during pregnancy, highlight the underlying molecular mechanisms, and discuss key research gaps. Considering pregnancy complications from the perspective of trained immunity may provide new insights into pathogenesis and suggest opportunities for biomarker discovery and targeted interventions.
    Keywords:  Decidual NK cells; Epigenetic reprogramming; Gestational diabetes mellitus; Metabolic adaptation; Preeclampsia; Trained immunity
    DOI:  https://doi.org/10.1016/j.placenta.2025.09.015
  16. Inflamm Regen. 2025 Oct 01. 45(1): 29
      Macrophages are highly plastic immune cells that adopt diverse functional states in response to the local microenvironment. The traditional M1/M2 polarization model that has long been used to describe macrophage activation is insufficient to capture the full spectrum of macrophage diversity observed in vivo. Advances in single-cell RNA sequencing (scRNA-seq) have revealed that macrophages exist in a continuum of transcriptional states formed by tissue-specific and disorder-specific cues. This insight has led to the recognition of disorder-specific macrophages, defined as macrophage subpopulations that emerge in response to pathological stimuli and play unique roles in disease progression. These macrophages exhibit distinct transcriptional signatures, epigenetic modifications, and functional properties shaped by their ontogeny and microenvironmental signals, arising from the reprogramming of resident macrophages or the differentiation of bone marrow-derived progenitors. Notable examples include macrophages in chronic infections (e.g., tuberculosis), immunosuppressive tumor-associated macrophages, lipid-associated macrophages in obesity, and disease-associated microglia in neurodegeneration. These subsets exhibit unique regulatory mechanisms, including enhancer remodeling driven by histone H3 lysine 27 acetylation in non-alcoholic steatohepatitis, CCAAT enhancer binding protein α-mediated differentiation in obesity, and Jmjd3-IRF4 axis control in allergic inflammation. Additionally, their function and fate are strongly influenced by their subtissular niche, as evidenced by crown-like structures in adipose tissue, tumor microenvironments, fibrotic lesions, and granulomas, where distinct microenvironmental cues shape macrophage behavior. Furthermore, interindividual heterogeneity in macrophage function, driven by genetic polymorphisms, is increasingly recognized, highlighting the role of host genetic background in disease susceptibility and macrophage-driven pathology. Here, we review the conceptual evolution of the disorder-specific macrophage, tracing its origins from the limited M1/M2 model to its refinement through scRNA-seq-based classification. We summarize the ontogeny, transcriptional regulation, and spatial heterogeneity of these macrophages across various disorders, emphasizing how the subtissular niche dictates functional specialization. Finally, we discuss potential therapeutic strategies targeting disorder-specific macrophage subsets, highlighting the need for integrative multi-omics approaches to refine their classification and functional characterization. Understanding the regulatory networks that govern disorder-specific macrophages will advance our knowledge of macrophage biology while facilitating the development of precision medicine for immune-related disorders.
    Keywords:  Disorder-specific macrophage; Interindividual diversity of macrophages; M1/M2 polarization model; Multidimensional spectrum model
    DOI:  https://doi.org/10.1186/s41232-025-00390-5
  17. Semin Immunopathol. 2025 Sep 30. 47(1): 37
      Conventional dendritic cells (cDCs) play a pivotal role in orchestrating the delicate balance between immunity and tolerance within the gastrointestinal tract by interacting with other cell types, particularly T cells. Meanwhile, the microbiota is critical for the induction and modulation of the immune system in the gut and plays a key role in the function of cDCs. So far, the study of intestinal cDCs has been encumbered by their limited numbers and phenotypic overlap with other myeloid cells. Recent advancements in single-cell sequencing technology have helped define cDCs and their subsets, while also providing valuable insights into the contribution of cDCs to Inflammatory Bowel Disease (IBD). However, the exact role of cDCs in IBD remains unclear, particularly in terms of how the microbiota influences their function in this context. In this review, we summarize the functions of cDCs in the intestine and during IBD, and the role of the microbiota in cDC biology. We also describe the current limitations in the study of cDCs and the microbiota, as well as new methods for studying DC-T cell communications in vivo, which can help increase our understanding of the function of cDCs in the intestine and develop new therapeutic strategies against IBD.
    Keywords:  Crohn’s disease; Dendritic cells; IBD; Microbiota; Ulcerative colitis
    DOI:  https://doi.org/10.1007/s00281-025-01062-9
  18. Nat Plants. 2025 Sep 30.
      Plants face constant microbial threats and have evolved highly effective immune systems characterized by inducible defence mechanisms. On recognizing microbial patterns and/or effectors, plants activate localized pattern-triggered immunity and/or effector-triggered immunity, which culminate in systemic acquired resistance-a broad-spectrum immune response that enhances protection throughout the plant. Systemic acquired resistance shares striking similarities with mammalian trained immunity, particularly in defence priming, which equips organisms with an enhanced capacity to respond to subsequent infections. This Review explores the cross-kingdom similarities between systemic acquired resistance and trained immunity, emphasizing their potential to transform agricultural practices and medical therapies. These insights present innovative opportunities for developing new plant-protection strategies, producing disease-resistant crops and optimizing vaccine approaches, while also highlighting critical knowledge gaps to inspire future research.
    DOI:  https://doi.org/10.1038/s41477-025-02119-1
  19. Inflamm Res. 2025 Oct 03. 74(1): 138
       OBJECTIVE: Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains a significant global health burden, characterized by complex host-pathogen interactions that drive heterogeneous clinical outcomes. While pulmonary epithelial cells are increasingly recognized as active participants in innate immunity during Mtb infection, how host defense are altered when the epithelial barrier is compromised remains unclear.
    METHODS: In this study, we developed a murine model combining naphthalene-induced pulmonary epithelial injury with Mtb infection and mapped the pulmonary cells landscape through single-cell RNA sequencing (scRNA-seq), followed by in vitro stimulation assays to validate macrophage functional changes.
    RESULTS: Notably, we found a pronounced impairment in pulmonary bacterial clearance. Transcriptomic analysis revealed a widespread suppression of epithelial immune functions and showed that macrophages transitioned from an antimicrobial to an antigen-presenting phenotype, indicating waning pulmonary innate defenses and heightened adaptive immune activation. In vitro experiments further suggested that this macrophage transition may be linked to epithelial cell alterations.
    CONCLUSIONS: These findings indicate that pulmonary epithelial integrity may influence early host immune responses to Mycobacterium tuberculosis and provide a transcriptomic framework for exploring epithelial-immune crosstalk as a potential therapeutic target.
    Keywords:   Mycobacterium tuberculosis ; Macrophages; Naphthalene; Pulmonary epithelial injury
    DOI:  https://doi.org/10.1007/s00011-025-02106-4
  20. Annu Rev Physiol. 2025 Oct 03.
      Plasticity of myeloid cells, characterized by their ability to undergo reprogramming in response to environmental cues, is a fundamental feature enabling their versatile functions during immune responses. Macrophages and neutrophils, the primary myeloid cell types, exhibit distinct polarization states. Classical polarization states of macrophages and neutrophils are associated with antimicrobial activity, inflammation promotion, and tissue remodeling. Pathological polarization, observed in chronic inflammation, cancer, and other conditions, is marked by enhanced immune-suppressive activity, aberrant enzymatic activity, and atypical cytokine production, diverging from their classical functions. This review delves into the most up-to-date characterization of those polarization states, the transcriptional and epigenetic factors, and the metabolic pathways governing myeloid cell reprogramming, highlighting the influence of cytokines and tissue-specific conditions, such as hypoxia in tumors, on this process. Understanding the mechanisms underlying the pathological polarization of myeloid cells offers a promising avenue to modulate their activity for targeted therapeutic interventions.
    DOI:  https://doi.org/10.1146/annurev-physiol-050824-111031
  21. Front Immunol. 2025 ;16 1584791
       Introduction: Macrophage polarization into M1 or M2 phenotypes is a complex process influenced by various factors. However, existing literature and ongoing research support the view that Cyclin-Dependent Kinase 5 (CDK5) may play an important role in this process. CDK5 is a protein kinase that requires association with regulatory, co-activating proteins, p35 (CDK5R1) or p39 (CDK5R2), for functional activation.
    Purpose: This study investigated the role of the p35 protein in regulating M1 and M2 polarization.
    Methods: We compared bone marrow derived macrophages from wild type (WT) and p35 knockout (KO) mice under both M1 (IFNγ + LPS) and M2 (IL4) conditions, differentiated with M-CSF or GM-CSF. The expression of surface markers (CD86, CD206), enzyme expression (Arginase-1 and iNOS), metabolism and antigen process and presentation were compared.
    Results: While p35 had modest effect on phenotype during M1 or M2 polarization, p35 expression was important for Arginase1 induction after M2 polarization. The absence of p35 significantly increased glycolysis during M1 polarization, while it also enhanced mitochondrial oxidative phosphorylation in the context of M2 polarization. While p35 was important for antigen processing by M0 and M2, M1 were able to maintain capacity to process antigen albeit with a reduction due to decreased stability of peptide: MHC II complex.
    Conclusion: While loss of p35 resulted in minor changes in phenotype, there were decreases in ARG-1 production and STAT3 phosphorylation, increased metabolism, and dramatically reduced antigen processing by M0, M1 or M2. The absence of p35 enhanced antigen uptake, but it had no effect on degradation of antigen, suggesting an inability to produce peptide: MHC II complexes in the absence of p35 in M0 and M2. In contrast, p35-deficient M1 maintained an ability to rapidly produce peptide: MHC II complexes but showed a reduction in the stability of these complexes on the surface. Our findings reveal a crucial role for p35 in regulating macrophage metabolism and antigen function, with implications for the development of novel therapeutic strategies.
    Keywords:  CDK5; antigen processing; macrophage; p35; polarization
    DOI:  https://doi.org/10.3389/fimmu.2025.1584791
  22. Med. 2025 Sep 29. pii: S2666-6340(25)00274-0. [Epub ahead of print] 100847
      The human skin serves as a major reservoir of mutualists that penetrate skin appendages and contribute to skin development, barrier repair, appendage health, and wound healing. The skin microbiota exhibits significant shifts in community composition in response to pathological skin conditions, such as impaired barrier integrity, follicular-sebaceous-related diseases, and wound healing, which contribute to the progression of skin diseases. Crosstalk among bacteria, fungi, and viruses and their collective effects on the host are critical determinants of skin health and disease. This review discusses the changes in the skin microbiota under both physiological and pathological conditions, with a particular focus on bacterial strains, virulence factors, and pathogenic genes, and their impact on host outcomes. Additionally, we preview the emerging clinical applications of specific bacterial strains, microbial competition, fungal cooperation, phage therapy, and engineered microbial interventions.
    Keywords:  microbial homeostasis and dysbiosis; skin diseases; skin microbiota
    DOI:  https://doi.org/10.1016/j.medj.2025.100847
  23. J Inorg Biochem. 2025 Sep 24. pii: S0162-0134(25)00265-X. [Epub ahead of print]274 113085
      Iron (Fe) is an essential nutrient for S. aureus survivability and pathogenesis, but excess Fe can catalyze the formation of toxic oxygen radicals, emphasizing the importance of maintaining proper Fe homeostasis. The essentiality of Fe for bacteria is exploited by host immunity strategies, which employ metal-binding proteins to decrease the availability of metal ions such as Fe. S. aureus responds to Fe limitation using the ferric uptake regulator (Fur) and the Fur protein antagonist (Fpa). During Fe-replete conditions, Fur functions as a transcriptional repressor of target genes. Upon Fe deprivation, Fur repression is relieved with the aid of Fpa, allowing for the increased expression of Fur-regulated genes such as iron uptake systems. We demonstrate that fur inactivation is required during Fe-limited growth and is independent of the described high-affinity Fe uptake systems. Using transcriptomic and metabolomic analyses, we demonstrate that fur inactivation or Fe limitation triggers a decrease in respiration and an increase in fermentation. Triggering fermentative growth allows S. aureus to cope with Fe limitation by having a metabolism less reliant on Fe but allowing for redox balance. Our work provides insight into how S. aureus adapts to iron limitation; a common stress encountered during infection.
    Keywords:  Fpa; Fur; Iron; Staphylococcus aureus; Sulfur; YlaN
    DOI:  https://doi.org/10.1016/j.jinorgbio.2025.113085
  24. J Med Microbiol. 2025 Sep;74(9):
      Introduction. Sputum is the most used sample type to monitor the lower respiratory tract microbiota in cystic fibrosis (CF), but young patients often cannot expectorate.Hypothesis. We hypothesized that throat swabs could reflect lower airway colonization and assessed the concordance of bacterial community composition between paired sputum and throat swab samples from children with CF.Aim. We aimed to compare bacterial community diversity and composition between sputum and throat swabs in the full cohort and in patients with paired samples from the same visit.Methodology. The prospective longitudinal multicentre MUCOVIB cohort included 379 samples from 61 CF children. Using V3-V4 16S rRNA amplicon metagenomics, we compared bacterial community diversity and composition between sputum and throat swabs in the full cohort and in 11 patients with paired samples from the same visit.Results. Sputum and throat swabs exhibited similar bacterial diversity, regardless of the exacerbation status, and presented a substantial agreement for detecting pathogens (Cohen's kappa: 0.6). Differences in bacterial abundance were observed (P=0.001), but not presence/absence (P=0.098). Community typing revealed three distinct community types, with 86% of paired samples falling into the same cluster, highlighting the homogeneity between sputum and throat swab microbiota. Network analysis demonstrated slight, non-random similarities in microbial interactions between sample types (adjusted Rand index=0.08 and 0.10). The average beta-diversity distances between samples collected from the same visit were shorter (0.505±0.056 95% confidence interval), compared with sputum (0.695±0.017) or throat swab (0.704±0.045) from the same patient collected during different visits.Conclusion. Throat swabs can provide representative information on lower respiratory microbiota. Clinicians should collect throat swabs rather than relying on sputum samples from previous visits to guide antibiotic prescriptions in CF children unable to expectorate.
    Keywords:  community profiling; cystic fibrosis; lower respiratory microbiota; metabarcoding; pathogen detection
    DOI:  https://doi.org/10.1099/jmm.0.002079
  25. Cell Rep. 2025 Sep 25. pii: S2211-1247(25)00973-8. [Epub ahead of print]44(10): 116202
      We developed an in vitro methodology to study trained immunity using murine bone-marrow-derived macrophages stimulated with β-glucan and lipopolysaccharide (LPS). Longitudinal analysis of interleukin (IL)-6 and tumor necrosis factor (TNF) production demonstrates that trained macrophages secrete higher cytokine levels following primary stimulation with β-glucan compared to unstimulated macrophages (step 1). After a resting period, trained macrophages return to basal levels of cytokine production (step 2) but rapidly produce enhanced levels of IL-6 and TNF after secondary stimulation with LPS, compared to macrophages individually stimulated with either β-glucan (step 3) or LPS (step 4) alone. The combined cytokine production of macrophages after single stimulation with β-glucan (stimulus 1) and LPS (stimulus 2) is significantly lower than the cytokine levels produced by trained macrophages sequentially stimulated with both β-glucan and LPS (stimulus 1 + 2) (step 5). These results experimentally reproduce the distinctive functional stages that macrophages undergo during the training process.
    Keywords:  CP: Immunology; SAA3; T cell proliferation; mTOR; mouse strains; sample cryopreservation; trained immunity
    DOI:  https://doi.org/10.1016/j.celrep.2025.116202
  26. Virulence. 2025 Dec;16(1): 2566255
      Within mammalian gastrointestinal ecosystems, trillions of microorganisms generate sophisticated ecological networks through bacterial extracellular vesicles (bEVs). Emerging evidence positions these bEVs as pivotal mediators in gut homeostasis maintenance and host-microbiota crosstalk, capable of transporting bioactive cargo including virulence determinants, genetic transfer components, and host-derived defense molecules. This molecular payload enables bEVs to exert multifaceted immunomodulatory effects through: 1) Immune cell activation and differentiation. 2) Microbial community regulation. 3) Epithelial barrier reinforcement. This review systematically examines current understanding of bEV biology: First, we characterize the structural complexity and compositional diversity of bEVs across bacterial species. Second, we elucidate the molecular machinery governing bEV biogenesis and secretion pathways. Third, we analyze mechanisms underlying bEV-immune interactions through receptor-mediated signaling and cargo delivery processes. By integrating recent advances in bEV immunobiology with mechanistic insights into host-pathobiont communication networks, our framework not only clarifies current knowledge gaps but also proposes standardized methodologies for future investigations.
    Keywords:  Bacteria; extracellular vesicles; immunity; intestinal tract
    DOI:  https://doi.org/10.1080/21505594.2025.2566255
  27. Sci Adv. 2025 Oct 03. 11(40): eady1640
      The uterine cervix is a critical mucosal interface that balances immune defense and reproductive function, yet how its distinct epithelial compartments coordinate responses to infection remains unclear. Here, we integrate patient-derived three-dimensional cervical organoids, single-cell transcriptomics, and native tissue analysis to construct a high-resolution atlas of epithelial cell diversity and immune dynamics during Chlamydia trachomatis infection. We demonstrate that cervical organoids closely resemble native tissue at transcriptional and cellular levels, identifying epithelial subtypes with region-specific immune specializations. Upon infection, ectocervical epithelia reinforce barrier integrity, whereas endocervical epithelia, particularly uninfected bystander cells, exhibit extensive transcriptional reprogramming characterized by robust interferon activation, antigen presentation, and antimicrobial defense. Infection profoundly reshapes epithelial intercellular communication, positioning bystander cells as major contributors to signaling networks coordinating immune responses and tissue regeneration. Our findings highlight a sophisticated epithelial-intrinsic immune network critical for cervical mucosal defense and establish a physiologically relevant platform for studying host-pathogen interactions and guiding targeted mucosal therapies against reproductive tract infections and pathologies.
    DOI:  https://doi.org/10.1126/sciadv.ady1640
  28. Inflammopharmacology. 2025 Sep 28.
      Respiratory disorders such as asthma, chronic obstructive pulmonary disease (COPD), acute respiratory distress syndrome (ARDS), pulmonary fibrosis, and infectious conditions including COVID-19 and tuberculosis continue to rank among the foremost causes of illness and death worldwide. Although vaccines, antimicrobial treatments, and anti-inflammatory agents have improved disease management, their overall impact remains limited because of the intricate regulation of immune responses at epithelial surfaces. Within this context, the interleukin-10 (IL-10) cytokine family (comprising IL-10, IL-19, IL-20, IL-22, IL-24, and IL-26) has been identified as a key immunological axis in the respiratory tract. These cytokines possess structural homology and predominantly transmit signals through heterodimeric class II receptors via the JAK-STAT cascade. However, their functions are far from uniform: IL-10 primarily exerts suppressive effects on inflammation, whereas IL-19, IL-20, IL-24, and IL-26 are commonly associated with tissue injury, chronic inflammation, and airway remodeling. IL-22 occupies an intermediate role, promoting epithelial regeneration under certain conditions but aggravating inflammation or tumorigenesis in others. This article reviews recent findings on the IL-10 family in a range of respiratory diseases, emphasizing their context-dependent activity, value as potential biomarkers, and relevance as therapeutic targets. A clearer understanding of how protective versus pathogenic signals are balanced within this cytokine network is essential for designing targeted interventions that preserve host defense while restoring airway integrity.
    Keywords:  ARDS/COVID-19; Asthma; COPD; Cystic fibrosis; Epithelial barrier; IL-10 family; IL-19; IL-20; IL-22; IL-24; IL-26; JAK–STAT; Pulmonary fibrosis; Pulmonary infections; Respiratory diseases; Therapeutic targets
    DOI:  https://doi.org/10.1007/s10787-025-01975-9
  29. Virol Sin. 2025 Sep 27. pii: S1995-820X(25)00134-8. [Epub ahead of print]
      Hepatitis B virus (HBV) establishes chronic infection through strategic manipulation of host metabolic networks, driving a spectrum of hepatic pathologies ranging from hepatitis to cirrhosis and hepatocellular carcinoma. Mechanistically, HBV reprograms core metabolic pathways, including glycolysis, tricarboxylic acid (TCA) cycle, oxidative phosphorylation, and lipid homeostasis, to fuel its replication machinery and evade immune surveillance. This review systematically synthesizes current evidence on HBV-induced glucose/lipid metabolic rewiring, with particular emphasis on how viral-host crosstalk at the metabolic interface sustains viral pathogenesis.
    Keywords:  HBV; TCA cycle; glycolysis; lipid metabolism; metabolic rewiring; oxidative phosphorylation
    DOI:  https://doi.org/10.1016/j.virs.2025.09.008
  30. Immunol Lett. 2025 Sep 25. pii: S0165-2478(25)00125-7. [Epub ahead of print]277 107092
      Inflammatory responses often involve metabolic rewiring within immune cells to support effector functions. Targeting metabolic pathways in immune cells therefore represents a promising strategy to modulate inflammatory diseases and improve therapeutic outcomes. Acyl-CoA synthesis by fatty acid transporter 2 (FATP2/SLC27A2) facilitates the transport of long-chain fatty acids into the cell. It represents a key step in fatty acid metabolism and the subsequent production of bioactive lipid mediators (LMs) with immunoregulatory functions. While the FATP2 inhibitor Lipofermata is currently evaluated for lipid-lowering therapies in metabolic diseases, and to revert the suppressive nature of myeloid cells in cancer, its effect on inflammatory responses in human macrophages remains elusive. Here, we show that Lipofermata reduced LPS-induced inflammatory responses in whole blood and human monocytes. This anti-inflammatory effect was paralleled by a decreased biosynthesis of arachidonic acid-derived inflammatory LMs, including prostaglandin E2 (PGE2) and thromboxane 2 (TxB2). These findings suggest an anti-inflammatory effect mediated by Lipofermata-mediated redirection of lipid metabolism in monocytes. Conversely, in mature human monocyte-derived macrophages, Lipofermata treatment enhanced LPS-induced cytokine production and induced cell death, likely through inflammasome activation. Together, these results underscore the cell type-specific effects of FATP2 inhibition and highlight the dual role of Lipofermata in modulating inflammatory immune responses. As such, targeting lipid metabolism with Lipofermata could have therapeutic potential with both anti- and pro-inflammatory applications, depending on the target cell type and context.
    Keywords:  Fatty acid metabolism; Human monocyte-derived macrophages; Immunology; Immunometabolism; Metabolism
    DOI:  https://doi.org/10.1016/j.imlet.2025.107092
  31. Adv Sci (Weinh). 2025 Sep 30. e08725
      Acute respiratory distress syndrome (ARDS) is a severe clinical condition characterized by widespread inflammation and fluid accumulation in the lungs. Endothelial cell (EC) metabolic changes in acute lung injury (ALI) and their relationship to injury remain unclear. Transcriptomic and lipidomic analyses revealed downregulation of PUFA synthesis pathways, particularly omega-3 PUFAs, in pulmonary ECs during LPS-induced ALI. Activation of the PUFA metabolic pathway, through FADS1/2 overexpression or omega-3 fatty acid supplementation, protected ECs from ferroptosis and restored barrier function. In vivo, pulmonary EC-specific overexpression of FADS1/2 contributed to the alleviation of ALI. Overexpression of whole lung FADS1/2, combined with alpha-linolenic acid (ALA) supplementation, also significantly mitigated ALI. PARK7 is identified as an endogenous regulator of FADS1/2, acting through the BMP-BMPR-SMAD1/5/9 signaling. Driven by histone H3K14 lactylation, which is also promoted by the downregulation of FADS1/2, PARK7 upregulation restored FADS1/2 expression and counteracted ferroptosis, thereby forming a protective feedback loop. This study elucidates a novel regulatory axis involving the two major metabolic changes-downregulation of PUFA synthesis and upregulation of histone lactylation-in ALI pathogenesis, which are interconnected through the PARK7-BMP signaling pathway. Targeting this axis offers potential therapeutic strategies for mitigating endothelial dysfunction and ferroptosis in ARDS/ALI.
    Keywords:  PARK7; acute lung injury (ALI); endothelial cell; ferroptosis; histone lactylation; polyunsaturated fatty acid (PUFA)
    DOI:  https://doi.org/10.1002/advs.202508725
  32. bioRxiv. 2025 Sep 25. pii: 2025.09.23.677925. [Epub ahead of print]
      The microbiota influence disease pathogenesis and treatment, however we have limited ability to assess patient status in relation to the microbiota. Here we find that the nitric oxide generating enzyme, nitric oxide synthase 2 (Nos2), is transcriptionally primed in intestinal epithelial cells (IECs), as opposed to immune cells, in inflammatory bowel disease (IBD) patients. Generation of IEC-specific Nos2 knockout mice revealed that epithelial Nos2 activity promoted susceptibility to intestinal disease and sustained a colitogenic microbiota. Epithelial Nos2 increased levels of nitric oxide-derived nitrates and nitrate-metabolizing bacteria in the intestine. Unexpectedly, extra-intestinal nitrates also reflected IEC-intrinsic Nos2 expression, and systemic nitrate concentrations in patients paralleled intestinal Nos2 activation. In fact, temporally inhibiting epithelial Nos2 was sufficient to alter intestinal nitrate homeostasis and inflammation in mice, as well as restrict nitrate production by human intestinal organoids. These data reveal that epithelial nitric oxide metabolism directs host-microbiota dynamics that can alter disease and that monitoring and targeting this axis may benefit patients with IBD.
    DOI:  https://doi.org/10.1101/2025.09.23.677925
  33. Environ Toxicol Pharmacol. 2025 Sep 26. pii: S1382-6689(25)00204-2. [Epub ahead of print]119 104829
      Airborne pollutants harm human health, but the mechanisms involved remain unclear. Impaired epithelial barrier function is, as in respiratory diseases, one possible pathomechanism. To investigate this, carbon black (CB) as a model for ultrafine particles (UFP), was applied to respiratory mucosa models of primary fibroblasts and epithelial cells cultured at the air-liquid interface (ALI). Models were assessed for the mucociliary phenotype. Cytotoxicity, DNA damage, and barrier integrity were evaluated by the lactate dehydrogenase (LDH) and comet assays, and by transepithelial electrical resistance (TEER) measurements. Cilia movement and ultrastructure, secretory cells, and intact cell-cell contacts were confirmed. Subtle changes were observed: the LDH release had increased 2 h post exposure and barrier disturbance 24 h post exposure was detected, both without mucosal damage or genotoxic effects. Donor-specific differences were present. Barrier disruption without cell detachment or death suggests model feasibility for long-term studies of, e.g., tissue regeneration or fibrosis following UFP exposure.
    Keywords:  Carbon black; air-liquid interface; barrier; primary cell model; respiratory mucosa; toxicology; ultrafine particles
    DOI:  https://doi.org/10.1016/j.etap.2025.104829
  34. Ann Dermatol. 2025 Oct;37(5): 276-285
       BACKGROUND: The skin microbiome plays a critical role in regulating epidermal differentiation and immune responses. Understanding of how individual microbial species influence the expression of barrier proteins and lipid synthesis pathways is essential for elucidating their contributions to skin barrier function.
    OBJECTIVE: This study aimed to investigate the distinct effects of Staphylococcus aureus (S. aureus), Staphylococcus hominis (S. hominis), and Cutibacterium acnes (C. acnes) on the skin barrier protein expression and lipid synthesis, thereby clarifying their roles in maintaining skin barrier integrity and homeostasis.
    METHODS: Keratinocyte 2-dimensional monolayer cultures and self-assembled 3-dimensional skin models were treated with S. aureus, S. hominis, or C. acnes. Alterations in skin barrier proteins and lipid synthesis were assessed using quantitative real-time polymerase chain reaction, immunofluorescence staining, and Oil Red O staining.
    RESULTS: S. aureus significantly downregulated the messenger ribonucleic acid expression of skin barrier proteins and lipid synthesis enzymes, resulting in reduced lipid accumulation. In contrast, S. hominis upregulated barrier protein expression and enhanced lipid accumulation. Similarly, C. acnes increased the expression of both skin barrier proteins and lipid synthesis enzymes, leading to a marked increase in lipid accumulation.
    CONCLUSION: Collectively, these findings suggest that S. aureus compromises the skin barrier function by downregulating the expression of barrier-associated proteins and lipid synthesis enzymes, whereas S. hominis and C. acnes enhance barrier integrity by upregulating these components. These differential microbial effects elucidate potential mechanisms by which the skin microbiome contributes to barrier homeostasis.
    Keywords:  In vitro 3D model; Keratinocytes; Lipids; Skin barrier function; Skin microbiome
    DOI:  https://doi.org/10.5021/ad.25.020
  35. PLoS Pathog. 2025 Sep 30. 21(9): e1013545
      Opportunistic pathogens switch from a commensal to pathogenic state by sensing and responding to a variety of environmental cues, including temperature fluctuations. Minor temperature oscillations can alert the pathogen to a changing niche ecosystem, necessitating efficient sensing and rapid integration to trigger behavioral change. This is typically achieved through master regulators, dictating pleiotropic phenotypes. Here, we uncover a pivotal role of minor temperature shifts in transition of Streptococcus pneumoniae (SPN) from commensal to virulent lifestyles, mediated via an RNA thermosensing (RNAT) element within the untranslated region of the global regulator CiaR. By positively regulating the expression of the surface adhesin, Phosphorylcholine (PCho), in response to elevated temperature, CiaR potentiates pneumococcal infection. Engineering the RNAT structure to create translation restrictive or permissive versions allowed us to demonstrate how modulation in expression of CiaR could alter pneumococcal invasion capability, influencing infection outcomes. Moreover, intranasal administration of PCho mitigated SPN-induced bacteraemic pneumonia. Since a majority of opportunistic respiratory bacterial pathogens decorate their surface with PCho, this signaling arm could be exploited for anti-infective interventions.
    DOI:  https://doi.org/10.1371/journal.ppat.1013545
  36. Nat Commun. 2025 Sep 29. 16(1): 8547
      The severity of allergic asthma is driven by the balance between allergen-specific T regulatory (Treg) and T helper (Th)2 cells. However, it is unclear whether specific subsets of conventional dendritic cells (cDCs) promote the differentiation of Tregs. We have identified a subset of lung resident type 2 cDCs (cDC2s) that display high levels of CD301b and have potent Treg-inducing activity ex vivo. Single-cell RNA sequencing and adoptive transfer experiments show that during allergic sensitization, many CD301b+ cDC2s transition in a stepwise manner to CD200+ cDC2s that selectively promote Th2 differentiation. GM-CSF augments the development and maintenance of CD301b+ cDC2s in vivo, and also selectively expands Treg-inducing CD301b+ cDC2s derived from bone marrow. Upon their adoptive transfer to recipient mice, lung-derived CD301b+ cDC2s confer immunological tolerance to inhaled allergens. Thus, GM-CSF maintains lung homeostasis by increasing numbers of Treg-inducing CD301b+ cDC2s.
    DOI:  https://doi.org/10.1038/s41467-025-63547-3
  37. J Microbiol Biotechnol. 2025 Sep 23. 35 e2506020
      SHR02, a derivative of homoisoflavonoid, exhibits potent anti-inflammatory activity in innate immune cells. In this study, we investigated the immunomodulatory effects of SHR02 in dendritic cells (DC2.4) and macrophages (RAW 264.7) under Toll-like receptor (TLR) stimulation. SHR02 significantly suppressed the secretion of pro-inflammatory cytokines (TNF-α and IL-6), reduced nitric oxide (NO) and reactive oxygen species (ROS) production, and downregulated the expression of iNOS and COX-2. Mechanistically, SHR02 inhibited NF-κB phosphorylation in dendritic cells while enhancing Nrf2 nuclear translocation in both cell types. These findings suggest that SHR02 modulates inflammatory and oxidative responses through both NF-κB and Nrf2 signaling pathways and may serve as a promising candidate for the treatment of inflammatory disorders.
    Keywords:  Nrf2; SHR02; dendritic cells; homoisoflavonoid; inflammation; macrophages
    DOI:  https://doi.org/10.4014/jmb.2506.06020
  38. Nat Immunol. 2025 Sep 29.
      Plasmacytoid dendritic cells (pDCs) are major producers of type I/III interferons. As interferons are crucial for antiviral defense, pDCs are assumed to play an essential role in this process; however, robust evidence supporting this dogma is scarce. Genetic or pharmacological manipulations that eliminate pDCs or disrupt their interferon production often affect other cells, confounding interpretation. Here, to overcome this issue, we engineered pDC-less mice that are specifically and constitutively devoid of pDCs by expressing diphtheria toxin under coordinated control of the Siglech and Pacsin1 genes, uniquely coexpressed in pDCs. pDC-less mice mounted protective immunity against systemic infection with mouse cytomegalovirus and showed higher survival and less lung immunopathology to intranasal infection with influenza virus and SARS-CoV-2. Thus, contrary to the prevailing dogma, we revealed that pDCs and their interferons are dispensable or deleterious during several viral infections. pDC-less mice will enable rigorously reassessing the roles of pDCs in health and disease.
    DOI:  https://doi.org/10.1038/s41590-025-02288-3
  39. PLoS Pathog. 2025 Oct 03. 21(10): e1013568
      Pseudomonas aeruginosa is a hallmark pathogen of cystic fibrosis (CF) airway infections, capable of reaching high cell densities despite its limited ability to directly utilize mucin glycoproteins as a nutrient source. In the CF lung, however, P. aeruginosa may access preferred carbon sources (e.g., amino acids and short-chain fatty acids) through metabolic cross-feeding with co-colonizing mucin-degrading microbes. Although host-derived enzymes such as neutrophil elastase can also degrade mucins, the extent to which host-mediated mucin breakdown supports P. aeruginosa growth remains unclear. Thus, here we compared the nutritional impact of microbial versus host mucolytic activity on P. aeruginosa physiology. Analyses of CF sputum revealed patient-specific variability in mucin integrity that is shaped by both host and microbial factors. We demonstrate that mucin degradation by anaerobic bacteria through proteolysis, glycolysis, and fermentation, promotes robust P. aeruginosa growth, unlike mucin processed by neutrophil elastase alone. Targeted metabolomics identified acetate and propionate as key metabolites driving this cross-feeding, while transcriptomic and phenotypic analyses revealed that P. aeruginosa engages in diauxic growth on a broader set of mucin-derived substrates. Unexpectedly, cross-feeding with anaerobes triggered the induction of P. aeruginosa denitrification and fermentation pathways, suggesting redox remodeling despite being cultured under oxygen-replete conditions. Finally, the transcriptional profile of P. aeruginosa grown on anaerobe-conditioned mucins more closely resembled its in vivo gene expression, more so than when grown on intact or neutrophil-degraded mucins. Together, these findings provide new insight into the potential role of interspecies metabolic interactions in shaping pathogen physiology in the inflammatory, polymicrobial, and mucus-rich environment of the CF airways.
    DOI:  https://doi.org/10.1371/journal.ppat.1013568
  40. Clin Sci (Lond). 2025 Oct 01. pii: CS20257363. [Epub ahead of print]138(19):
      Pulmonary arterial hypertension (PAH) is a syndrome characterized by a mean pulmonary artery pressure >20 mmHg and elevated pulmonary vascular resistance >2 Wood Units in the absence of left heart disease, chronic lung disease or hypoxia, and chronic thromboembolic disease. PAH is an obliterative pulmonary arteriopathy that leads to morbidity and mortality, often due to right ventricular failure (RVF). Emerging evidence from preclinical research, using chemical inhibition or genetic depletion of inflammatory mediators, reveals a role for inflammation in the adverse pulmonary vascular remodelling in PAH. More recently, studies have also identified inflammation of the right ventricle (RV) as a potential contributor to RV decompensation and failure. While inflammation contributes to the pathogenesis of PAH, no approved PH-targeted therapies specifically target inflammation. Macrophages are myeloid cells that play a critical role in inflammation and PAH. Their cellular plasticity enables the acquisition of tissue-specific phenotypes and functions that may promote either resolution or exacerbation of inflammatory signalling. Macrophage plasticity in PAH is poorly understood. We examine how alterations in glucose metabolism, particularly the uncoupling of glycolysis from glucose oxidation-a notable feature of PAH observed in various cell populations-impact macrophage polarization and the inflammatory phenotype associated with PAH. The study of immune cell metabolism, known as immunometabolism, is an emerging field that has yet to be explored in PAH. Improving understanding of the inflammatory mechanisms in PAH, particularly novel pathways related to macrophage immunometabolism, may identify new targets for anti-inflammatory therapies for PAH.
    Keywords:  NOD-like receptor protein 3 (NLRP3); interleukin 1 beta (IL-1β); itaconate; mitochondrial metabolism; pyruvate dehydrogenase kinase; succinate dehydrogenase
    DOI:  https://doi.org/10.1042/CS20257363
  41. Nat Commun. 2025 Sep 29. 16(1): 8565
      Gut microbiota influence the severity of pneumonia by producing metabolites that enhance systemic and pulmonary immune responses. Preclinical studies suggested that gut microbiota-derived indoles have protective effects against numerous diseases, including influenza and abdominal infections. However, the precise role of tryptophan metabolites during pneumonia is unknown. Here, we perform translational analyses in a large general-population cohort (n = 13,464), critically ill patients with severe community-acquired pneumonia (CAP; n = 158; NCT01905033), a randomized human intervention trial on antibiotic-mediated microbiota modulation (NCT03051698), and mice to investigate the effects of tryptophan metabolites, specifically indole-3-acetic acid (IAA), on pneumonia. In the population-based cohort, baseline IAA is associated with a higher risk of future hospital admission for pneumonia (cause-specific hazard ratio 1.15, 95% confidence interval 1.09-1.22 p < 0.0001). In patients with severe CAP higher levels of IAA are associated with increased mortality, independent from potential confounders (hazard ratio 1.30 per log2 increase, 95% confidence interval 1.02-1.68, p = 0.037). In a mouse model of bacterial pneumonia, IAA supplementation aggravates pulmonary damage while reducing systemic dissemination, which is mediated by the aryl hydrocarbon receptor (AhR) and increased release of reactive oxygen species from neutrophils. In summary, these findings from general population and severe pneumonia cohorts, and murine pneumonia experiments, show that the gut microbiota-derived tryptophan metabolite IAA affects pneumonia, suggesting that various indoles may have diverging, context-dependent effects.
    DOI:  https://doi.org/10.1038/s41467-025-63611-y
  42. Immunology. 2025 Oct 02.
      Sepsis is a life-threatening condition characterised by a dysregulated host response to infection, resulting in systemic inflammation, immune dysfunction, and multi-organ failure. Kupffer cells (KCs), the largest population of tissue-resident macrophages in the body, are essential for pathogen clearance, endotoxin detoxification, and maintaining hepatic immune homeostasis during sepsis. However, sepsis induces substantial KC depletion, contributing to increased bacterial burden and mortality. In this study, we demonstrate that β-glucan treatment effectively protects against sepsis-induced KC loss and reduces circulating bacterial load. Mechanistically, β-glucan attenuates KC death by suppressing NLRP3 and gasdermin D (GSDMD)-mediated pyroptosis triggered by bacterial infections. Notably, we identify a previously unrecognised function of β-glucan in markedly enhancing KC self-renewal during sepsis through downregulation of the transcriptional repressors c-Maf and MafB, which are known to inhibit macrophage proliferation. This discovery reveals a novel mechanism of hepatic macrophage regeneration and supports β-glucan as a promising immunomodulatory therapy to preserve liver immune integrity, enhancing antibacterial defence, and reducing the risk of secondary infections in immunocompromised septic hosts.
    Keywords:  immune homeostasis; macrophage; sepsis
    DOI:  https://doi.org/10.1111/imm.70043