bims-bac4me Biomed News
on Microbiome and trained immunity
Issue of 2025–05–04
35 papers selected by
Chun-Chi Chang, Universitäts Spital Zürich



  1. Cell Rep. 2025 Apr 27. pii: S2211-1247(25)00395-X. [Epub ahead of print]44(5): 115624
      Cutaneous leishmaniasis is a parasitic infection that causes a spectrum of pathology ranging from single, self-healing lesions to disfiguring chronic wounds. In severe disease, uncontrolled inflammation exacerbates tissue damage and delays healing, though the contributing factors are unclear. We previously observed that delayed healing was associated with Staphylococcus aureus in the lesional microbiota of patients with cutaneous leishmaniasis. To investigate how S. aureus impacts immunopathology during leishmania infection, we established a murine model of S. aureus colonization with clinical isolates followed by Leishmania infection. S. aureus triggered early production of interleukin (IL)-1β during Leishmania infection, which was critical for neutrophil recruitment and cutaneous inflammation. S. aureus isolates differentially induced IL-1β and neutrophil recruitment, and isolates that induced greater neutrophil recruitment were resistant to neutrophil killing and persisted longer. We reveal a mechanism whereby S. aureus mediates immunopathology during cutaneous leishmaniasis, suggesting IL-1β as a promising immunomodulatory target for non-healing infections.
    Keywords:  CP: Microbiology; Leishmania; Staphylococcus aureus; cutaneous leishmaniasis; microbiota; skin infection
    DOI:  https://doi.org/10.1016/j.celrep.2025.115624
  2. Elife. 2025 Apr 30. pii: e106854. [Epub ahead of print]14
      Specific host factors, such as immune cell activity, sex hormones and microbiota composition, influence the ability of Staphylococcus aureus bacteria to colonize the gut of mice.
    Keywords:  MRSA; Th17; gastrointestinal colonization; immunology; inflammation; microbiota; mouse; sex hormone
    DOI:  https://doi.org/10.7554/eLife.106854
  3. J Infect Dis. 2025 Apr 26. pii: jiaf220. [Epub ahead of print]
      We examined whether IFNG restricts Mycobacterium tuberculosis (Mtb) growth in human macrophages across a range of conditions and methods. We observed an IFNG-dependent enhancement of bacterial replication in MCSF-differentiated monocyte-derived macrophages (MDMs, 4.84 x 105 CFU IFNG stimulated versus 2.48 x 105 CFU untreated, P < 0.001) with four Mtb strains. Mtb replication was not restricted by IFNG treatment of alveolar macrophages. In agreement with previous studies, IFNG-stimulated murine bone marrow derived macrophages effectively restricted Mtb replication. These data suggest that Mtb resists IFNG-stimulated immunity within human macrophages with implications for distinct host species immune responses.
    Keywords:   Mycobacterium tuberculosis ; innate immunity; interferon gamma; macrophage
    DOI:  https://doi.org/10.1093/infdis/jiaf220
  4. Nature. 2025 Apr 23.
      
    Keywords:  Antibiotics; Microbiology; Microbiome
    DOI:  https://doi.org/10.1038/d41586-025-01313-7
  5. Mol Med. 2025 Apr 30. 31(1): 159
      Neonates, especially those born prematurely, are highly vulnerable to infection-induced mortality. Numerous observational and immunological studies in newborns have shown that live attenuated vaccines have beneficial, non-specific effects (NSEs) against secondary infections to unrelated pathogens. These beneficial effects have been attributed to trained immunity, and emergency granulopoiesis plays an essential role. However, trained immunity has been shown to affect multiple myeloid subsets and how trained immunity influences the host protective response is still undefined. Here we show that Bacillus-Calmette-Guérin (BCG) vaccination improves survival to polymicrobial sepsis by simultaneously reprogramming broad aspects of myelopoiesis. Specifically, BCG vaccination expands multiple myeloid subsets, including the lineage (Lin)-Sca- 1+c-kit+ (LSK) and granulocytic-macrophage progenitors (GMPs), and increases CD11b+Gr1+ cell number, as well as their oxidative metabolism and capacity to stimulate T-cell proliferation in response to sepsis. Single-cell RNA sequencing of neonatal splenocytes suggests that BCG-vaccination changes the broad transcriptional landscape of multiple myeloid subsets. The result is the maturation of various neutrophil and monocyte subsets, stimulation of antimicrobial processes, and suppression of inflammatory pathways and myeloid-derived suppressor cell transcription. These findings reveal that BCG administration early after birth fundamentally reorganizes the myeloid landscape to benefit the subsequent response to polymicrobial infection.
    Keywords:  BCG; Myeloid-derived suppressor cells; Neonatal sepsis; Single-cell RNA sequencing; Trained immunity
    DOI:  https://doi.org/10.1186/s10020-025-01179-5
  6. Immunohorizons. 2025 Apr 26. pii: vlaf011. [Epub ahead of print]9(6):
      Phagocytosis requires the coordination of various classes of receptors and the activation of multiple signaling programs, culminating in actin cytoskeletal rearrangement and ingestion. Given the pleiotropic nature of the events necessary for proper microbial ingestion, identifying molecules that control distinct steps of phagocytosis could reveal potential strategies to enhance microbial clearance. PTEN is a lipid/protein phosphatase traditionally recognized as a tumor suppressor. While PTEN inhibits various arms of the innate immune response, its role during Staphylococcus aureus infection remains unclear. We hypothesize that PTEN inhibits the functions of scavenger receptors (SRs) and the actin cytoskeleton during methicillin-resistant S. aureus (MRSA) infection in macrophages. RNAseq analysis of PTEN KO immortalized bone marrow-derived macrophages (iBMDMs) unveiled increased expression of genes involved in actin polymerization, pathogen recognition, and SRs, which leads to enhanced MRSA phagocytosis in both iBMDMs and primary peritoneal macrophages lacking PTEN. PTEN is physically associated with 2 SRs, MARCO and CD36, and blocking these receptors prevents the increased phagocytosis seen in PTEN KO macrophages. PTEN binds to the actin depolymerizing factor cofilin-1 during infection, inhibiting F-actin (the essential form of actin for phagocytosis) while increasing G-actin pools. Cytometry by time of flight (CyTOF) analysis of human myeloid cell populations from a PTEN-haploinsufficient patient suggests that PTEN is necessary for generating specific monocyte and dendritic subclasses. This study identifies the role of PTEN in macrophage phagocytosis of a gram-positive pathogen and in the development of monocyte subsets. This highlights the spectrum of PTEN importance in host defense mechanisms in both murine and human phagocytes.
    Keywords:   Staphylococcus aureus ; PTEN; actin; phagocytosis; scavenger receptors
    DOI:  https://doi.org/10.1093/immhor/vlaf011
  7. Cell Mol Life Sci. 2025 Apr 28. 82(1): 182
      The role of immune metabolism, specific metabolites and cell-intrinsic and -extrinsic metabolic states across the time course of an inflammatory response are emerging knowledge. Targeted and untargeted metabolomic analysis is essential to understand how immune cells adapt their metabolic program throughout an immune response. In addition, metabolomic analysis can aid to identify pathophysiological patterns in inflammatory disease. Here, we discuss new metabolomic findings within the transition from inflammation to resolution, focusing on three key programs of immunity: Efferocytosis, IL-10 signaling and trained immunity. Particularly the tryptophan-derived metabolite kynurenine was identified as essential for efferocytosis and inflammation resolution as well as a potential biomarker in diverse inflammatory conditions. In summary, metabolomic analysis and integration with transcriptomic and proteomic data, high resolution imaging and spatial information is key to unravel metabolic drivers and dependencies during inflammation and progression to tissue-repair.
    Keywords:  Chronic inflammatory disease; Efferocytosis; IL-10; Kynurenine; Metabolomics; Spatial resolution; Trained immunity
    DOI:  https://doi.org/10.1007/s00018-025-05715-8
  8. Front Immunol. 2025 ;16 1599215
      
    Keywords:  inflammation; innate immunity; macrophage; macrophage activation; macrophage polarization
    DOI:  https://doi.org/10.3389/fimmu.2025.1599215
  9. Recent Adv Antiinfect Drug Discov. 2025 ;20(1): 5-29
      The human microbiome, a diverse microorganism community, crucially defends against pathogens. Probiotics, postbiotics, and paraprobiotics alone and in combination are potent in countering fungal and waterborne infections, particularly against viral threats. This review focuses on the mechanisms of the microbiome against viral infections, emphasizing probiotic interventions. Certain Lactic Acid Bacteria (LAB) strains effectively eliminate toxic aflatoxin B1 (AFB1) from microfungi-produced mycotoxins. LAB binding to AFB1 persists post-gastric digestion, and pre-incubation with mycotoxins reduces probiotic adhesion to mucus. Oral probiotic administration in animals increases mycotoxin excretion, reducing associated health risks. Bifidobacterium longum and Lactobacillus rhamnosus show exceptional efficacy in removing cyanobacterial toxin microcystin-LR from drinking water. Engineered probiotics promise advanced therapeutic applications for metabolic disorders, Alzheimer's, and type 1 diabetes, serving as diagnostic tools for detecting pathogens and inflammation markers. In antimicrobial peptide production, genetically modified probiotics producing human β-defensin 2 (HBD2) treat Crohn's disease with implemented biocontainment strategies preventing unintended environmental impacts.
    Keywords:  Probiotics; antimicrobial peptides.; fungal infections; immunomodulation; viral infections; waterborne pathogens
    DOI:  https://doi.org/10.2174/0127724344308638240530065552
  10. Nat Immunol. 2025 May;26(5): 639
      
    DOI:  https://doi.org/10.1038/s41590-025-02157-z
  11. Microb Biotechnol. 2025 May;18(5): e70147
      The respiratory tract microbiome (RTM) is a multi-kingdom microbial ecosystem that inhabits various niches of the respiratory system. While previously overlooked, there is now sufficient evidence that the RTM plays a crucial role in human health related to immune system training and protection against pathogens. Accordingly, dysbiosis or disequilibrium of the RTM has been linked to several communicable and non-communicable respiratory diseases, highlighting the need to unveil its role in health and disease. Here, we define the RTM and its place in microbiome medicine. Moreover, we outline the challenges of RTM research, emphasising the need for combining methodologies, including multi-omics and computational tools. We also discuss the RTM's potential for diagnosing, preventing and treating respiratory diseases and developing novel microbiome-based therapies to improve pulmonary health.
    Keywords:  human health; methods in microbiome research; microbiome biotechnology; microbiome‐based therapies; respiratory tract microbiome
    DOI:  https://doi.org/10.1111/1751-7915.70147
  12. Cell Rep. 2025 Apr 25. pii: S2211-1247(25)00394-8. [Epub ahead of print]44(5): 115623
      Tissue-resident macrophages (TRMs) populate throughout various tissues, and their homeostatic metabolism is heavily influenced by these microenvironments. Peroxisomes are organelles that contribute to lipid metabolism. However, the involvement of these organelles in the bioenergetics of TRMs remains undetermined. We conducted a developmental screen of TRMs using a conditional peroxisomal biogenesis factor 5 (Pex5) knockout mouse model that lacks functional peroxisomes in all immune cell subsets. Pulmonary alveolar macrophages (AMs) appeared as the only subset of TRMs that required functional peroxisomes for their development. Pex5 deficiency resulted in reduced AM survival due to increased sensitivity to lipotoxicity, in line with an excess accumulation of ceramides. The absence of peroxisomes had a significant effect on overall mitochondrial fitness and altered their metabolic program, allowing them to engage in glycolysis in addition to oxidative phosphorylation. Our results revealed that AMs have a unique metabolic regulation, where peroxisomes play a central role in their homeostatic development and maintenance.
    Keywords:  CP: Immunology; CP: Metabolism; immunometabolism; innate immunity; peroxisomes; tissue-resident macrophages
    DOI:  https://doi.org/10.1016/j.celrep.2025.115623
  13. J Transl Med. 2025 Apr 29. 23(1): 488
      The mucosal system serves as the primary barrier against respiratory diseases and plays a crucial role in combating viral infections through mucosal immunity. The resident microbial community constitutes the main component of the mucosal system and exerts a significant inhibitory impact on the invasion of exogenous agents. However, the precise relationship between resident microbiota, mucosal immunity, and viral infections remains incomplete. This review aims to summarize the regulatory interactions between the resident microbiota of the mucosal system and innate immune components such as mucosal immunity and trained immunity. By clarifying these complex relationships, this review seeks to identify potential targets for augmenting respiratory disease prevention strategies and developing novel vaccine formulations. Furthermore, we propose the possibility of integrating the fields of microbiome-based therapeutics and vaccine development to create multifunctional vaccine formulations capable of targeting mucosal immunity induction. Such an approach holds great potential in offering novel pathways and strategies for the prevention and treatment of respiratory diseases.
    Keywords:  Microbiota; Mucosal immune; Respiratory tract; Trained immunity; Viral infection
    DOI:  https://doi.org/10.1186/s12967-025-06433-2
  14. Immunity. 2025 Apr 24. pii: S1074-7613(25)00167-0. [Epub ahead of print]
      Crosstalk between the immune system and the microbiome is critical for maintaining immune homeostasis. Here, we examined this communication and the impact of immune-suppressive IL-10 signaling on pulmonary homeostasis. We found that IL-10 sensing by interstitial macrophages (IMs) is required to prevent spontaneous lung inflammation. Loss of IL-10 signaling in IMs initiated an inflammatory cascade through the activation of classical monocytes and CD4+ T cell subsets, leading to chronic lung inflammation with age. Analyses of antibiotic-treated and germ-free mice established that lung inflammation in the animals lacking IL-10 signaling was triggered by commensal bacteria. 16S rRNA sequencing revealed Delftia acidovorans and Rhodococcus erythropolis as potential drivers of lung inflammation. Intranasal administration of these bacteria or transplantation of human fecal microbiota elicited lung inflammation in gnotobiotic Il10-deficient mice. These findings highlight that IL-10 sensing by IMs contributes to pulmonary homeostasis by preventing lung inflammation caused by commensal dysbiosis.
    Keywords:  Delftia acidovorans; IL-10; IL-10R; Rhodococcus erythropolis; Th1 and Th17 cells; commensal bacteria; gut-lung axis; interstitial macrophages; lung inflammation; monocyte-derived macrophages
    DOI:  https://doi.org/10.1016/j.immuni.2025.04.004
  15. Immunol Lett. 2025 Apr 28. pii: S0165-2478(25)00062-8. [Epub ahead of print] 107029
      Monocytes are circulating immune cells that migrate to inflamed tissues and differentiate into macrophages, where they play a dual role in regulating pro-inflammatory and pro-resolving responses through cytokine and lipid mediator secretion. Platelet-derived microvesicles (PMVs), released during platelet activation, infiltrate inflamed areas and interact with monocytes and macrophages, facilitating the transfer of bioactive contents. While these interactions have been observed, their functional consequences on monocyte/macrophage inflammatory profiles remain poorly understood. In this study, PMVs are shown to be internalized by human THP-1 monocytes. The interaction with THP-1 cells occurs rapidly, with 60% of cells interacting with PMVs within one hour. When cells are differentiated to M0 and M1 macrophages, interactions with PMVs only peak after 24 hours. Interaction of cells with PMVs resulted in an increased capacity to synthesize cyclooxygenase- and lipoxygenase-derived lipid mediators of inflammation, especially in M1 cells. Cytokine production was also influenced in a cell-state-dependent manner. PMVs had no impact on undifferentiated THP-1 cells but enhanced the production of several cytokines in M0 cells as well as IL-23 and IL-6 in M1 macrophages. When stimulated with lipopolysaccharides, PMV-treated M0 macrophages demonstrated elevated production of the anti-inflammatory cytokine IL-10, while M1 macrophages exhibited increased secretion of IL-1β, MCP-1, and IL-6, highlighting an effect on pro-inflammatory cytokine production. These findings reveal that PMVs selectively modulate the inflammatory cytokine and lipid mediator profiles of monocytes and macrophages depending on their differentiation state. This study underscores the role of PMVs as key players in intercellular communication and immune regulation, particularly in the context of inflammation.
    Keywords:  cytokines; extracellular microvesicles; inflammation; lipid mediators; macrophages; monocytes
    DOI:  https://doi.org/10.1016/j.imlet.2025.107029
  16. bioRxiv. 2025 Apr 07. pii: 2025.04.06.647500. [Epub ahead of print]
      After ingestion into macrophage phagosomes, some bacterial pathogens such as Mycobacterium tuberculosis ( Mtb ) evade killing by preventing phagosome acidification and fusion of the phagosome with a lysosome. Mtb accumulates extracellular polyphosphate (polyP), and polyP inhibits macrophage phagosome acidification and bacterial killing. In Dictyostelium discoideum , polyP also inhibits bacterial killing, and we identified some proteins in D. discoideum that polyP requires to suppress the killing of ingested bacteria. Here, we find that pharmacological inhibition of human orthologues of the D. discoideum proteins, including P2Y1 receptors, mammalian Target of Rapamycin (mTOR), and inositol hexakisphosphate kinase, enhances the killing of Mtb , Legionella pneumophila , and Listeria monocytogenes by human macrophages. Mtb inhibits phagosome acidification, expression of the proinflammatory marker CD54, and autophagy, and increases expression of the anti-inflammatory marker CD206. In Mtb -infected macrophages, the polyP-degrading enzyme polyphosphatase (ScPPX) and inhibitors reversed these effects, with ScPPX increasing CD54 expression more in female macrophages compared to male macrophages. In addition, Mtb inhibits proteasome activity, and some, but not all, inhibitors reversed these effects. While the existence of a dedicated polyP signaling pathway remains uncertain, our findings suggest that pharmacological inhibition of select host proteins can restore macrophage function and enhances the killing of intracellular pathogens.
    Importance: Human macrophages engulf bacteria into phagosomes, which then fuse with lysosomes to kill the bacteria. However, after engulfment, pathogenic bacteria such as Mycobacterium tuberculosis , Legionella pneumophila , and Listeria monocytogenes can block phagosome-lysosome fusion, allowing their survival. Here, we show that pharmacological inhibition of specific macrophage proteins reverses these effects and enhances bacterial killing. These findings suggest that targeting host factors involved in these processes may provide a therapeutic strategy to improve macrophage function against infections such as tuberculosis, Legionnaires' disease, and listeriosis.
    DOI:  https://doi.org/10.1101/2025.04.06.647500
  17. Eur J Dent. 2025 May 02.
      Dental caries is an infectious disease that develops through biofilm. Streptococcus mutans is a cariogenic bacterium that can be found in dental plaque. Streptococcus mutans regulates biofilm formation and communicate with other microbes through a process called quorum sensing. Dental caries prevention can be achieved by inhibiting quorum sensing. This study aimed to investigate the ability of Lactobacillus plantarum and Candida albicans to inhibit the formation of S. mutans polymicrobial biofilms. This study aims to investigate the ability of biofilm formation analyzed through the crystal violet (CV) assay and bacterial metabolic activity analyzed through the methylthiazol tetrazolium (MTT) assay. The bacteria used are S. mutans (serotype C), L. plantarum (FNCC 0020), and C. albicans.CV assay results show that in the presence of L. plantarum, biofilm formation in S. mutans decreases (9.5%). Meanwhile, the formation of S. mutans biofilms increased with the presence of C. albicans (28.8%). MTT assay results showed an increase in the metabolic activity of S. mutans in the presence of L. plantarum (20.2%) and C. albicans (19.4%). Lactobacillus plantarum can inhibit the formation of S. mutans biofilms, while C. albicans can increase S. mutans biofilms.
    DOI:  https://doi.org/10.1055/s-0045-1806962
  18. PLoS Pathog. 2025 Apr 28. 21(4): e1013114
      Candida auris is an emerging multidrug-resistant skin-tropic fungal pathogen that causes serious human infections. However, the factors that regulate C. auris skin infection in vivo are still unclear. In this study, we identified that, unlike Candida albicans, which induces IL-17-secreting protective effector Th17 cells, C. auris predominately induces IFNγ-secreting pathogenic Th1 cells during reinfection. Surprisingly, we found that IFNγ enhances skin infection of C. auris but not C. albicans. Mechanistically, IFNγ enhances skin infection of C. auris by dampening the protective IL-17 responses and increasing dermal damage. Furthermore, we identified that the development of Th1 cells occurs through IL-12, produced by C. auris-induced inflammatory macrophages and monocyte-derived dendritic cells. In addition, our findings reveal that C. auris unique cell wall outer mannan layer regulates the development of Th1 and Th17 cells. Collectively, our findings, for the first time, identified that C. auris induces IFNγ to persist in the skin. These findings help explain why C. auris but not C. albicans preferentially persist in the skin long-term, with the potential to identify novel therapeutic approaches to prevent and treat this emerging fungal pathogen in humans.
    DOI:  https://doi.org/10.1371/journal.ppat.1013114
  19. Vet Microbiol. 2025 Apr 27. pii: S0378-1135(25)00173-7. [Epub ahead of print]305 110538
      Salmonella is one of the most common zoonotic pathogens, posing a significant threat to both animal and human health. Our previous study demonstrated that autophagy plays a crucial role in restricting the intracellular growth of Salmonella. This study aims to investigate the effect of autophagy in Salmonella Typhimurium (S. Typhimurium)-induced ferroptosis. First, we found that S. Typhimurium induced lipid peroxidation by increasing intracellular Fe2 + levels, promoting lipid oxidation, and inhibiting the antioxidant pathway. S. Typhimurium-induced lipid peroxidation led to ferroptosis in macrophages. Further results revealed that S. Typhimurium triggered ferritin degradation by NCOA4-mediated ferritinophagy. Additionally, S. Typhimurium-induced chaperone-mediated autophagy (CMA) degraded GPX4 through TAK1-HSC70 signaling pathway. Notably, GPX4 is involved in intracellular S. Typhimurium release. Overall, autophagy was essential for S. Typhimurium induced-ferroptosis, TAK1 not only facilitated autophagy to eliminate intracellular bacteria but also promoted bacterial release.
    Keywords:  Autophagy; Ferroptosis; Macrophages; Salmonella Typhimurium
    DOI:  https://doi.org/10.1016/j.vetmic.2025.110538
  20. J Extracell Vesicles. 2025 May;14(5): e70077
      Bacterial extracellular vesicles (EVs) are natural reservoirs of biological active substances. They exhibit promising application in developing bioproducts such as vaccine, drug-delivery system and anticancer agent. However, the low yield of naturally secreted EVs during bacterial growth is a bottleneck factor that restricts EV applications. In this study, we showed that sub-minimum inhibitory concentration (MIC) of β-lactams boosted EV production in various Staphylococcus aureus strains. The expression of penicillin-binding protein (PBP) genes increased after β-lactam treatment, and the inactivation of alternative PBPs promoted EV secretion of S. aureus. We also demonstrated that sub-MIC β-lactams promoted EV production via a reactive oxygen species (ROS)-dependent pathway. Deletion of redundant pbp genes enhanced oxacillin (OXA)-stimulated ROS levels. Transcriptomic and lipidomic analyses revealed that OXA-induced ROS triggered lipid metabolic reprogramming in S. aureus. Particularly, ROS promoted lipid peroxidation (LPO) and increased the biosynthesis of phosphatidic acid (PA) and lipoteichoic acid (LTA) that contributed to EV generation. Furthermore, OXA treatment altered the diversity of EV-loaded proteins. OXA-treated ∆ agr /OXAEVs induced stronger Dengue EDIII-specific antibodies in BALB/c mice than did ∆ agrEVs. Overall, this study provided mechanic insights into β-lactam-promoted EV production in S. aureus, and highlighted the potential strategies to prepare EVs for various applications.
    Keywords:  Staphylococcus aureus; extracellular vesicle; lipid metabolism; penicillin binding proteins; reactive oxygen species; β‐lactam antibiotics
    DOI:  https://doi.org/10.1002/jev2.70077
  21. Biomedicines. 2025 Mar 25. pii: 791. [Epub ahead of print]13(4):
      The skin microbiome, a diverse and dynamic ecosystem of microorganisms, plays a pivotal role in maintaining skin health by interacting with skin cells, immune components, and structural barriers. It is essential for skin homeostasis, immune defense, and protection against pathogenic colonization. Dysbiosis in the microbiome has been implicated in numerous dermatological conditions, including acne, eczema, psoriasis, and rosacea. Acne, the most prevalent skin condition, affects up to 85% of individuals at some point in their lives, while eczema and psoriasis impose significant public health and economic burdens. The composition of the skin microbiome varies across skin types and anatomical sites, with sebaceous, moist, and dry areas fostering distinct microbial communities. Emerging therapeutic strategies such as microbiome-targeted treatments offer novel avenues for addressing skin diseases. Among these approaches, postbiotics have gained significant attention for their safety and efficacy. Unlike probiotics, postbiotics are non-viable microbial cells or their metabolites, which reduce safety concerns while providing functional benefits such as UV protection and wound healing. This review consolidates current insights into the role of the skin microbiome in health and disease, emphasizing postbiotics as a promising therapeutic strategy by exploring the clinical and commercial potential of microbiome-based treatments, particularly postbiotics, and their ability to redefine dermatological care and improve patient outcomes.
    Keywords:  acne; dermatology; eczema; postbiotics; psoriasis; skin barrier function; skin diseases; skin health; skin microbiome
    DOI:  https://doi.org/10.3390/biomedicines13040791
  22. Benef Microbes. 2025 Mar 26. 1-15
      Lactic acid bacteria are a group of microorganisms recognised for their health-promoting properties, with several strains being commercially utilised as probiotics. Probiotics offer numerous benefits, including modulation of the immune system, enhancement of nutrient absorption, regulation of intestinal microbiota, protection against intestinal pathogens, and strengthening of the intestinal barrier. However, the precise mechanisms by which probiotics exert their effects remain incompletely understood. In recent years, research into new therapeutic applications for probiotics has intensified, driven by the urgent need for strategies to combat antibiotic-resistant bacteria. Among the newly discovered properties of probiotics is their ability to produce antivirulence compounds. These compounds reduce the virulence of pathogens without inhibiting microbial growth, thereby imposing less selective pressure for the development of resistance compared to traditional antibiotics. Given the potential for these compounds in clinical settings, this study aims to provide a comprehensive review of the antivirulence activities of probiotics, with particular focus on lactic acid bacteria. It discusses their effects on two-component and quorum sensing systems, which regulate the simultaneous expression of various virulence genes, as well as their anti-adhesion, anti-biofilm, anti-toxin, and anti-enzymatic activities against a range of pathogens. Thus, this review offers insight into the novel mechanisms by which lactic acid bacteria contribute to health, potentially broadening their applications.
    DOI:  https://doi.org/10.1163/18762891-bja00063
  23. Front Immunol. 2025 ;16 1549749
      Amphiregulin (AREG), a member of the EGF family, exists as a transmembrane protein anchored to the cell surface. In response to external stimuli, its extracellular domain is released into the extracellular matrix through paracrine or autocrine signaling. However, its role in septic macrophage pyroptosis remains poorly understood. This study aims to investigate the role of extracellular AREG in septic macrophages, mice, and patients. We found that high expression of extracellular AREG was regulated by RPLP1 at the translation level, which increased the expression of IL-6, CCL2, and CCL3 protein, as well as Caspase 1, IL-1β, and Nlrp3 mRNA expression, resulting in macrophage pyroptosis. Mechanistically, macrophage pyroptosis was aggravated by extracellular AREG pretreatment, which was triggered by extracellular AREG and ATP (adenosine 5'-triphosphate). The AREG-neutralizing antibody reduced LPS-induced epidermal growth factor receptor (EGFR) activation, TLR4 expression, and pyroptosis. Extracellular AREG-induced macrophage pyroptosis decreased with EGFR and NF-κB inhibition, as well as TLR4 and Myd88 knockout. Additionally, DTT-pretreated extracellular AREG suppressed macrophage pyroptosis. In vivo, extracellular AREG attenuates systemic inflammation infiltration and delays survival in a septic mouse model. Furthermore, extracellular AREG mediates sepsis in humans, and genes involved in the AREG-mediated pyroptosis signaling pathway were highly expressed in patients with severe sepsis compared with those with general or moderate sepsis. Overall, LPS-induced extracellular AREG aggravated or triggered macrophage pyroptosis through the EGFR/TLR4/Myd88/NF-κB signaling pathway, providing promising treatment strategies for sepsis.
    Keywords:  EGFR/TLR4; amphiregulin; macrophage; pyroptosis; sepsis
    DOI:  https://doi.org/10.3389/fimmu.2025.1549749
  24. Sci Immunol. 2025 May 02. 10(107): eadl3852
      Resolution of lung injuries is vital to maintain gas exchange, but there is an increased risk of secondary bacterial infections during this stage. Alveolar macrophages (AMs) are crucial to clear bacteria and control the resolution of inflammation, but environmental cues that switch functional phenotypes of AMs remain incompletely understood. Here, we found that AMs lack the capacity to mount an effective immune response against bacteria during resolution of inflammation. Neutrophil (PMN)-derived myeloperoxidase (MPO) fueled canonical glutaminolysis via the mitochondrial membrane transporter uncoupling protein-2 (UCP2), resulting in decreased mtROS-dependent killing of bacteria and secretion of pro-inflammatory cytokines. MPO-enhanced UCP2 expression inhibited mitochondrial hyperpolarization and boosted efferocytosis irrespective of the presence of bacterial pathogens. Conversely, efferocytosis of other cell types resulted in a distinct anti-inflammatory AM phenotype while maintaining antibacterial phenotypic plasticity. Overall, our findings indicate that the uptake of apoptotic PMNs or MPO switches AMs to prioritize resolution of inflammation over antibacterial responses, a feature that is conserved in murine extrapulmonary macrophages and human AMs.
    DOI:  https://doi.org/10.1126/sciimmunol.adl3852
  25. J Mol Histol. 2025 Apr 28. 56(3): 147
       BACKGROUND: S. aureus pneumonia, one of the most common S. aureus-induced diseases, is characterized by infectious inflammation in alveoli, distal airway, and lung interstitial. Forsythiaside A possesses anti-inflammatory, anti-infective, and other pharmacological properties in several diseases. The role of forsythiaside A remains unclear in S. aureus pneumonia.
    AIM OF THE STUDY: We aimed to figure out the role of forsythiaside A in S. aureus pneumonia.
    METHODS: RAW264.7 cells and C57BL6 mice were infected with S. aureus to construct S. aureus pneumonia cell model and animal model, respectively. A series of experiments including MTT, ELISA, Western blot, H&E staining and EBD staining were operated to figure out the role of forsythiaside A in S. aureus pneumonia.
    RESULTS: In RAW264.7 cells, forsythiaside A did not induce cell toxicity but triggered cytokines (TNF-α, IL-6 and IL-1β) release in a dose-dependent manner. Moreover, forsythiaside A inhibited p38 JNK/MAPK/ERK and NF-κB pathways by repressing phosphorylation of p38, JNK, ERK and p65 proteins. For in vivo study, forsythiaside A improved survival rate of S. aureus pneumonia mice by alleviating lung injury. In addition, forsythiaside A protected from air-blood barrier destruction and pulmonary edema. At last, forsythiaside A inhibited neutrophils infiltration and inflammatory response in bronchoalveolar lavage fluid.
    CONCLUSIONS: Forsythoside A inhibited inflammatory response by inhibiting p38 JNK/MAPK/ERK and NF-κB signaling in S. aureus pneumonia, which provided a novel insight for S. aureus pneumonia treatment.
    Keywords:   Staphylococcus aureus pneumonia; Forsythoside A; Inflammatory response; NF-κB pathways; p38 JNK/MAPK/ERK
    DOI:  https://doi.org/10.1007/s10735-025-10418-2
  26. PLoS Biol. 2025 Apr;23(4): e3003088
      Innate immunity serves as a crucial surveillance framework, but can be exploited to facilitate tumor progression. Two new PLOS Biology studies independently show how premalignant cells can exploit Toll-NF-κB signaling, in concert with oncogenic Ras, to enable unchecked growth.
    DOI:  https://doi.org/10.1371/journal.pbio.3003088
  27. Respir Res. 2025 Apr 25. 26(1): 160
       BACKGROUND: The composition and characteristics of airway microbiota offer critical insights for clinical decision-making. Current research on chronic lung diseases shows differences in the composition and characteristics of upper and lower respiratory tract microbiota compared with healthy individuals. However, the temporal changes of these microbial communities in lung transplant recipients remain poorly characterized.
    METHODS: This is a longitudinal prospective study. Respiratory specimens were collected regularly from lung transplant recipients for testing and analysis. A total of 150 bronchoalveolar lavage fluid (BALF) samples, 150 throat swab samples, 51 sputum samples, and 36 lung tissue samples were collected from the recipients, at 7 days, 14 days, 1 month, 2 months, 3 months, and 6 months post-transplant for 16S rRNA gene sequencing and analysis.
    RESULTS: Our study showed that there were significant differences in α-diversity and β-diversity among lung tissue, throat swab, and sputum samples, although α-diversity did not show a significant difference between lung tissue and BALF. Most amplicon sequence variants (ASVs) belonged to the families Enterobacteriaceae, Pseudomonadaceae, and Stenotrophomonas in BALF, while most ASVs belonged to the genera Streptococcus, Pseudomonadaceae, and Stenotrophomonas in sputum samples. Regarding dynamic changes, Corynebacterium and Staphylococcus were more prevalent in the early post-operative period but gradually decreased by 7 days post-operatively, while the common microbiota found in healthy populations based on literature became the most abundant ASVs at 6 months post-operatively in our study participants. Pseudomonadaceae and Stenotrophomonas contributed to the similarity in the composition of upper and lower respiratory microbiota.
    CONCLUSIONS: This study demonstrates that lung transplant recipients exhibit unique characteristics in their upper and lower respiratory tract microbiota, which are distinct ecological profiles, and both undergo significant changes within 6 months post-operatively. The similarity between upper and lower respiratory tract microbiota is associated with microbial diversity and taxonomic dominance.
    CLINICAL TRIAL: The clinical trial was registered at Chinese Clinical Trial Registry (ChiCTR2200056908) in February 2022.
    Keywords:  16S rRNA; Lung transplant; Upper versus lower airway Microbiome
    DOI:  https://doi.org/10.1186/s12931-025-03235-4
  28. Elife. 2025 Apr 30. pii: RP90668. [Epub ahead of print]12
      The Staphylococcus aureus clonal complex 8 (CC8) is made up of several subtypes with varying levels of clinical burden; from community-associated methicillin-resistant S. aureus USA300 strains to hospital-associated (HA-MRSA) USA500 strains and ancestral methicillin-susceptible (MSSA) strains. This phenotypic distribution within a single clonal complex makes CC8 an ideal clade to study the emergence of mutations important for antibiotic resistance and community spread. Gene-level analysis comparing USA300 against MSSA and HA-MRSA strains have revealed key horizontally acquired genes important for its rapid spread in the community. However, efforts to define the contributions of point mutations and indels have been confounded by strong linkage disequilibrium resulting from clonal propagation. To break down this confounding effect, we combined genetic association testing with a model of the transcriptional regulatory network (TRN) to find candidate mutations that may have led to changes in gene regulation. First, we used a De Bruijn graph genome-wide association study to enrich mutations unique to the USA300 lineages within CC8. Next, we reconstructed the TRN by using independent component analysis on 670 RNA-sequencing samples from USA300 and non-USA300 CC8 strains which predicted several genes with strain-specific altered expression patterns. Examination of the regulatory region of one of the genes enriched by both approaches, isdH, revealed a 38-bp deletion containing a Fur-binding site and a conserved single-nucleotide polymorphism which likely led to the altered expression levels in USA300 strains. Taken together, our results demonstrate the utility of reconstructed TRNs to address the limits of genetic approaches when studying emerging pathogenic strains.
    Keywords:  GWAS; Staphylococcus aureus; computational biology; independent component analysis; infectious disease; microbiology; systems biology; transcriptional regulatory network
    DOI:  https://doi.org/10.7554/eLife.90668
  29. FEBS J. 2025 Apr 28.
      Aberrant activation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome increases the release of mature pro-inflammatory cytokines interleukin (IL)-1β and IL-18, and enhances pyroptosis; thereby necessitating tight regulation of the NLRP3 inflammasome. Dysfunctional glutamine metabolism contributes to the pathogenesis of multiple inflammatory disorders, and the precise mechanism remains to be elucidated. Here, we provide evidence that glutamine deprivation enhances NLRP3 inflammasome activation in macrophages. Indeed, the absence of exogenous glutamine specifically enhanced NLRP3 inflammasome assembly, thereby accelerating pyroptosis and promoting the maturation of IL-1β and IL-18. Inhibition of glutaminolysis exhibited a similar effect to glutamine deprivation, whereas this effect was reversed by α-ketoglutarate (α-KG), a tricarboxylic acid (TCA)-cycle intermediate that can be replenished by glutamine supply. We further observed reduced generation of endogenous itaconate by glutamine deprivation and verified that both exogenous supplementation of itaconate derivative and increased endogenous itaconate production by overexpressing immune-responsive gene 1 [IRG1; also known as aconitate decarboxylase 1 (ACOD1)] could replace glutamine to inhibit the NLRP3 inflammasome. Mechanistically, glutamine deprivation decreased the source of substrate and inhibited transcription factor EB (TFEB)-dependent transcriptional upregulation of IRG1, thereby impairing the IRG1/itaconate axis that suppresses the NLRP3 inflammasome. Furthermore, glutamine deficiency was detected in a murine sepsis model, whereas extrinsic glutamine supplementation conferred protection against intestinal inflammation and tissue damage in septic mice. Taken together, our findings provide a novel insight into the link between glutamine metabolism and NLRP3 inflammasome activation, highlighting the target of glutamine metabolism, which holds as a potential therapeutic strategy for inflammatory diseases.
    Keywords:  IRG1/itaconate axis; NLRP3 inflammasome; glutamine metabolism; itaconate; pyroptosis
    DOI:  https://doi.org/10.1111/febs.70119
  30. ACS Nano. 2025 Apr 30.
      Celiac disease (CeD), triggered by gliadin exposure, necessitates therapeutic strategies that establish an antigen-specific immune tolerance. This study explores the therapeutic efficacy and mechanism of rapamycin-gliadin composite nanoparticles (PLN-GR) for CeD treatment. In vivo analyses demonstrated the efficient uptake of PLN-GR by antigen-presenting cells (APCs), particularly Kupffer cells and splenic dendritic cells (DCs), driving their tolerogenic phenotypic transformation. In a murine CeD model, PLN-GR administration significantly enhanced gluten tolerance and mitigated intestinal inflammation, as indicated by reduced paw edema and improved histopathological parameters. Mechanistically, PLN-GR induced macrophage metabolic reprogramming from glycolysis to oxidative phosphorylation, concomitant with elevated serum itaconate levels. This metabolic shift potentiated interorgan immunoregulatory crosstalk, expanding PD-L1+ tolerogenic splenic DCs while suppressing pathogenic Th1 cell populations. Bone marrow-derived macrophages (BMDMs) from Acod1-/- mice (deficient in itaconate synthesis) failed to induce DC tolerance upon PLN-GR treatment. However, supplementation with the itaconate derivative 4-octyl itaconate (4-OI) restored PD-L1 expression in DC2.4 cells in vitro, revealing that itaconate induces and stabilizes the tolerant DC phenotype. These findings underscore PLN-GR as a novel nanotherapeutic platform for CeD, achieving gliadin-specific tolerance through hepatic-splenic immunometabolic reprogramming and itaconate-dependent PD-L1 regulation, thereby offering a translatable strategy for autoimmune disease management.
    Keywords:  gluten sensitivity; immune tolerance; immunometabolic reprogramming; immunomodulation; itaconate metabolism
    DOI:  https://doi.org/10.1021/acsnano.4c18354
  31. bioRxiv. 2025 Apr 10. pii: 2025.04.10.648192. [Epub ahead of print]
      Studies of antimicrobial therapeutics have traditionally neglected the contribution of the host in determining the course of treatment and its outcome. One critical host element, which shapes the dynamics of treatment is the innate immune system. Studies of chemotherapeutics and complementary therapies such as bacteriophage (phage), are commonly performed with mice that purposely have an ablated innate immune system. Here, we generate a mathematical and computer-simulation model of the joint action of antibiotics, phage, and phagocytes. Our analysis of this model highlights the need for future studies to consider the role of the innate immune system of a host in determining treatment outcomes. Critically, our model predicts that the conditions under which resistance to the treatment agent(s) will emerge are much narrower than commonly anticipate. We also generate a second model to predict the dynamics of treatment when multiple phages are used. This model provides support for the application of cocktails to treat infections rather than individual phages. Overall, this study provides hypotheses that can readily be tested experimentally with both in vitro and in vivo experiments.
    DOI:  https://doi.org/10.1101/2025.04.10.648192
  32. Immunobiology. 2025 Apr 24. pii: S0171-2985(25)00042-7. [Epub ahead of print]230(3): 152908
       OBJECTIVE: Chronic obstructive pulmonary disease (COPD) is intricately linked to pulmonary fibrosis, yet the underlying mechanisms remain unclear. This study investigates whether CH25H/25-hydroxycholesterol (25-HC) promotes pulmonary fibrosis in COPD by modulating AMPK/STAT6-dependent M2 macrophage polarization.
    METHODS: Using GEO datasets and a cigarette smoke-induced COPD mouse model, we analyzed CH25H expression and fibrotic pathology. CH25H was silenced via adeno-associated virus (AAV)-delivered shRNA. Histopathology, flow cytometry, qPCR, and Western blotting assessed fibrosis, macrophage polarization (M1/M2), and AMPK/STAT6 pathway activity. Bone marrow-derived macrophages (BMDMs) were employed to validate polarization mechanisms. The role of the AMPK/STAT6 pathway was investigated using the specific activator.
    RESULTS: Analysis of the GEO database and experimental verification showed significantly increased CH25H expression in both lung tissues and macrophages from COPD mice. CH25H knockdown alleviated alveolar damage, airway remodeling, and pulmonary fibrosis in COPD mice, evidenced by reduced expression of fibrosis-related proteins, improved lung function, and attenuated inflammatory responses. Moreover, CH25H knockdown inhibited M2 macrophage polarization and decreased the proportion of M2-type macrophages. Importantly, decreased levels of 25-HC following CH25H knockdown were asso ciated with suppressed activation of the AMPK/STAT6 pathway. Rescue experiments demonstrated that the protective effects of CH25H knockdown could be reversed by adding the AMPKα activator GSK621.
    CONCLUSION: Our findings demonstrate that CH25H/25-HC exacerbates COPD-associated pulmonary fibrosis by promoting AMPK/STAT6-dependent M2 macrophage polarization. Targeting CH25H may represent a novel therapeutic strategy for mitigating fibrosis in COPD.
    Keywords:  AMPK/STAT6; CH25H/25-HC; Chronic Obstructive Pulmonary Disease; M2 Macrophages; Pulmonary Fibrosis
    DOI:  https://doi.org/10.1016/j.imbio.2025.152908
  33. Front Biosci (Landmark Ed). 2025 Apr 23. 30(4): 37430
       BACKGROUND: As pivotal immunoregulatory sentinels in pulmonary defense systems, alveolar macrophages (AMs) play dual roles in mediating inflammatory responses and tissue repair processes during various phases of inflammatory cascades. The present investigation focuses on elucidating the regulatory influence of Notch pathway activation within AM populations on the pathophysiological mechanisms underlying acute lung injury (ALI) development.
    METHODS: To investigate the regulatory roles of Notch intracellular domain (NICD) and C-C chemokine receptor type 5 (CCR5) in pulmonary inflammation, an ALI model was established through lipopolysaccharide (LPS) administration. Complementary studies used macrophage-specific Notch1 knockout mice and immortalized bone marrow-derived macrophages (iBMDMs). Molecular profiling of CCR5 and inflammatory mediators was performed through real-time quantitative reverse transcription PCR (qRT-PCR) and immunofluorescence staining. Functional assessments of macrophage migration were carried out using scratch wound healing assays and transwell migration assays.
    RESULTS: In the LPS-induced ALI model, pulmonary tissues exhibited elevated expression of both NICD and CCR5. Conversely, Notch1 knockout mice attenuated CCR5 expression, reduced macrophage infiltration and downregulated transcription of pro-inflammatory mediators compared to wild-type controls (p < 0.05). Lung injury was milder in the Notch1-deficient mice model compared to wild mice (p < 0.05). In vitro experiments demonstrated that inhibiting the Notch pathway in macrophages reduced CCR5 expression and attenuated CCL5-induced macrophage migration.
    CONCLUSION: Notch signaling regulates macrophage infiltration and the inflammatory response by modulating CCR5 expression in ALI induced by LPS.
    Keywords:  CCR5; Notch; acute lung injury; alveolar macrophages; cell migration
    DOI:  https://doi.org/10.31083/FBL37430