Tao Li, Yongjun Lu, Fangfang Yu, Qiuling Zhong, Yifan Meng, Yiwei Feng, Yi Hu, Xiangyang Tian, Tingting Li, Rengfei Shi
{"title":"Aerobic exercise modalities on gut microbiome and skeletal muscle quality in ovariectomized mice.","authors":"Tao Li, Yongjun Lu, Fangfang Yu, Qiuling Zhong, Yifan Meng, Yiwei Feng, Yi Hu, Xiangyang Tian, Tingting Li, Rengfei Shi","doi":"10.3389/fcimb.2025.1634934","DOIUrl":"10.3389/fcimb.2025.1634934","url":null,"abstract":"<p><strong>Objective: </strong>This study aimed to investigate the effects of aerobic exercise on skeletal muscle quality, gut microbiota composition, and estrogen levels in ovariectomized (OVX) mice, and to elucidate the potential underlying mechanisms, thereby providing experimental evidence for exercise intervention in postmenopausal women.</p><p><strong>Methods: </strong>Adult female C57BL/6J mice were randomly assigned to four groups (n = 6 per group): Sham, OVX, Sham+ET, and OVX+ET. After 6 weeks of recovery, the exercise groups received 8 weeks of treadmill training. Muscle morphology, function, and protein metabolism pathways were assessed using histology, grip tests, and Western blotting. Aromatase and estrogen levels were evaluated by immunofluorescence and ELISA. Gut microbiota composition was analyzed via 16S rRNA sequencing and correlated with muscle function.</p><p><strong>Results: </strong>Eight weeks of aerobic exercise significantly improved skeletal muscle mass, fiber cross-sectional area, and grip strength in OVX mice, and reduced fatigue index compared to OVX controls. Immunofluorescence revealed increased aromatase expression and intramuscular E<sub>2</sub> levels following exercise, with no significant difference in serum estradiol. Western blot analysis indicated activation of the Akt/mTOR/p-S6 pathway and inhibition of FOXO3-mediated protein degradation. 16S rRNA sequencing showed that exercise increased α-diversity (Shannon and Simpson indices) and altered microbial community structure, as shown by distinct clustering in PCoA plots. At the genus level, exercise modulated the relative abundance of several bacterial taxa. Spearman correlation analysis demonstrated that microbial diversity indices were positively associated with lean mass and fatigue resistance.</p><p><strong>Conclusion: </strong>Aerobic exercise significantly improves muscle mass and function in ovariectomized mice, potentially through a combined mechanism involving regulation of protein metabolism, enhancement of local estrogen synthesis, and modulation of gut microbiota composition.</p>","PeriodicalId":12458,"journal":{"name":"Frontiers in Cellular and Infection Microbiology","volume":"15 ","pages":"1634934"},"PeriodicalIF":4.8,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12479491/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145206059","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Liu Xin, Xiaodan Jiao, Xiaowei Gong, Jing Yu, Jing Zhao, Jing Lv, Qixuan Feng, YaDong Yuan, Wensen Pan
{"title":"Diagnostic value of metagenomic next generation sequencing of bronchoalveolar lavage fluid in immunocompromised patients with pneumonia.","authors":"Liu Xin, Xiaodan Jiao, Xiaowei Gong, Jing Yu, Jing Zhao, Jing Lv, Qixuan Feng, YaDong Yuan, Wensen Pan","doi":"10.3389/fcimb.2025.1602636","DOIUrl":"10.3389/fcimb.2025.1602636","url":null,"abstract":"<p><strong>Background: </strong>Metagenomic next-generation sequencing (mNGS) enables simultaneous sequencing of DNA fragments for comprehensive pathogen identification. Pneumonia in immunocompromised patients-characterized by atypical clinical manifestations and rapid progression-poses diagnostic challenges. Conventional microbiological testing (CMT), which relies on pathogen culture and serological assays, is limited by prolonged turnaround times and suboptimal detection rates. This study was performed to evaluate the clinical utility of mNGS through comparative analysis with CMT in detecting pathogens among immunocompromised patients with pneumonia.</p><p><strong>Methods: </strong>We conducted a retrospective cohort study of 146 immunocompromised patients with suspected pneumonia. The mNGS and CMT results were systematically analyzed. Pathogen detection rates and microbial spectrum concordance were visualized using pie and bar charts. Diagnostic performance was compared using McNemar's test and Kappa (κ) statistics for inter-method agreement. The sensitivity, specificity, accuracy, and area under the curve were calculated for pathogen-specific evaluations.</p><p><strong>Results: </strong>mNGS demonstrated superior detection efficacy, identifying pathogens in 98 cases versus 50 by CMT, with 48 overlapping positives. The microbial spectrum showed substantial differences: mNGS detected 73 bacterial, 46 fungal, and 45 viral pathogens, whereas CMT identified 38 bacterial, 27 fungal, and 21 viral agents. mNGS outperformed CMT across all infection types, including single-pathogen infections (bacterial, fungal, or viral only) and mixed infections (bacterial + fungal, bacterial + viral, fungal + viral, or bacterial + fungal + viral). Bacterial and fungal detections showed low inter-method concordance, while viral detection exhibited moderate agreement (κ = 0.510, <i>p</i> < 0.001). Notably, mNGS achieved significantly higher detection rates for <i>Enterococcus faecalis</i> and <i>Pneumocystis jirovecii</i> in intensive care unit (ICU)-admitted patients with severe pneumonia (<i>p</i> < 0.05). Clinical outcomes improved in 45 patients following mNGS-guided therapeutic adjustments.</p><p><strong>Conclusions: </strong>mNGS and CMT demonstrate complementary strengths in bacterial and fungal detection in immunocompromised patients with pneumonia. mNGS provides enhanced diagnostic accuracy for key pathogens such as <i>E. faecalis</i> and <i>P. jirovecii</i>, particularly in severe and ICU-admitted cases. As a high-throughput diagnostic tool, mNGS may improve pathogen detection and clinical management in immunocompromised populations.</p>","PeriodicalId":12458,"journal":{"name":"Frontiers in Cellular and Infection Microbiology","volume":"15 ","pages":"1602636"},"PeriodicalIF":4.8,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12479419/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145206026","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Joachim Müller, Javier Regidor-Cerrillo, David Arranz-Solís, Sophie Braga-Lagache, Anne-Christine Uldry, Manfred Heller, Rafael Calero-Bernal, Andrew Hemphill, Luis Miguel Ortega-Mora
{"title":"Proteome changes during <i>in vitro</i> culture adaptation of <i>Toxoplasma gondii</i> archetypal II and III field isolates.","authors":"Joachim Müller, Javier Regidor-Cerrillo, David Arranz-Solís, Sophie Braga-Lagache, Anne-Christine Uldry, Manfred Heller, Rafael Calero-Bernal, Andrew Hemphill, Luis Miguel Ortega-Mora","doi":"10.3389/fcimb.2025.1633384","DOIUrl":"10.3389/fcimb.2025.1633384","url":null,"abstract":"<p><strong>Introduction: </strong>Rapid <i>in vitro</i> culture adaptation of recently obtained <i>Toxoplasma gondii</i> isolates leading to deep changes in relevant phenotypic traits has been demonstrated earlier. Few reports exist on the molecular bases that govern this adaptation. Herein, we analyzed the <i>T. gondii</i> proteomes of different isolates at two timepoints during cell culture adaptation.</p><p><strong>Methods: </strong>The differential proteomes of six recently obtained archetypal European <i>T. gondii</i> Type II (TgShSp1 (Genotype ToxoDB#3), TgShSp2 (#1), TgShSp3 (#3) and TgShSp16 (#3)) and Type III (TgShSp24 (#2) and TgPigSp1(#2)) isolates maintained at low (10-16) and high (50-53) passage numbers in Vero cells were determined by label free liquid chromatography-mass spectrometry.</p><p><strong>Results: </strong>Among these isolates, 2.3% and 10.2% of proteins were differentially or constantly abundant when comparing low and high passage numbers. Constant proteins included components involved in essential cellular processes such as energy metabolism or protein synthesis, many of them identified as drug and vaccine targets. Interestingly, differentially abundant proteins were clearly linked to phenotypic changes associated to <i>in vitro</i> adaptation: loss of ability to spontaneously form cysts at high passages and decreased expression of cyst and bradyzoite markers (BAG1, Enolase 1, and SRS35A), while culture adaptation was associated with increased abundance of recognized virulence factors such as GRA15, GRA16, TEEGR and NSM.</p><p><strong>Conclusion: </strong>Our results highlight the changes at the proteomic level that take place in recently obtained isolates after <i>in vitro</i> culture adaptation, an important feature that should be considered during <i>T. gondii</i> investigations.</p>","PeriodicalId":12458,"journal":{"name":"Frontiers in Cellular and Infection Microbiology","volume":"15 ","pages":"1633384"},"PeriodicalIF":4.8,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12479500/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145206065","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Meng Wang, Xiaopeng Yang, Yimin Li, Mingxin Jiang, Bairu Chen, Wei Yang, Nan Ma, Shili He, Chengyue Wang
{"title":"Exploring the influence of psychological factors on the comorbidity of dental caries and obesity in adolescents from the perspective of the oral-gut-brain axis.","authors":"Meng Wang, Xiaopeng Yang, Yimin Li, Mingxin Jiang, Bairu Chen, Wei Yang, Nan Ma, Shili He, Chengyue Wang","doi":"10.3389/fcimb.2025.1659042","DOIUrl":"10.3389/fcimb.2025.1659042","url":null,"abstract":"<p><strong>Introduction: </strong>This study investigates how psychological factors influence the comorbidity of dental caries and obesity in adolescents through the oral-gut-brain axis. Adolescence is a critical period for both physical and psychological development, yet dental caries and obesity are prevalent issues that can negatively impact mental health. The study aims to provide insights into the underlying mechanisms and potential prevention and treatment strategies.</p><p><strong>Methods: </strong>An epidemiological survey was conducted on 1,024 students aged 12-15 from Beizhen No. 1 Junior High School. A total of 90 adolescents were selected for biosample research. The methods used included 16S rRNA gene sequencing, untargeted metabolomics, and SourceTracker analysis to examine oral and gut microbiota and metabolite concentrations.</p><p><strong>Results: </strong>Significant differences in oral and gut microbiota and metabolite concentrations were found among adolescents with different health statuses. Adolescents with caries and obesity showed distinct microbial profiles compared to healthy controls. The study also identified potential oral and gut microbial biomarkers associated with psychological disorders. SourceTracker analysis revealed a higher rate of ectopic colonization of oral microbiota in the intestines of adolescents with caries and obesity.</p><p><strong>Discussion: </strong>The study highlights the roles of the oral-gut and oral-brain axes in the comorbidity of dental caries and obesity among adolescents. The findings suggest that oral and gut microbiota play crucial roles in disease progression, and their imbalances may affect mental health through the oral-gut-brain axis. The results provide a theoretical foundation for developing integrated intervention strategies targeting both oral and systemic health.</p>","PeriodicalId":12458,"journal":{"name":"Frontiers in Cellular and Infection Microbiology","volume":"15 ","pages":"1659042"},"PeriodicalIF":4.8,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12479512/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145206073","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yaqin He, Furui Zhang, Zhiqiang Tian, Ruyi Li, Ming Su, Liping Hong, Jun Wen, Cao Zhang, Jinhai Tian, Le Guo
{"title":"Immune microenvironment-dependent effects of age-associated <i>Bifidobacterium</i> strains on gut immunity and microbial diversity.","authors":"Yaqin He, Furui Zhang, Zhiqiang Tian, Ruyi Li, Ming Su, Liping Hong, Jun Wen, Cao Zhang, Jinhai Tian, Le Guo","doi":"10.3389/fcimb.2025.1639178","DOIUrl":"10.3389/fcimb.2025.1639178","url":null,"abstract":"<p><strong>Background: </strong>The applications of probiotics in food and infant formula are greatly increased. <i>Bifidobacterium</i>, a genus of beneficial bacteria, plays a crucial role in the human gut microbiota. Despite extensive research on probiotics, how age-associated <i>Bifidobacteria</i> strains modulate gut immunity and microbial diversity remains unclear.</p><p><strong>Methods: </strong>Our present study investigates the immunomodulatory effects of two <i>Bifidobacterium</i> strains, <i>Bifidobacterium adolescentis</i> (BA) and <i>Bifidobacterium longum subsp. infantis</i> (BI), on gut immunity and microbial diversity using three models: a DSS-induced chronic colitis mouse model, germ-free mouse model, and in vitro human intestinal γδ T cell co-culture system.</p><p><strong>Results: </strong>Transcriptomic analysis in the DSS-induced colitis model revealed differential gene expression, particularly in cytokine signaling pathways and γ-chain-related cytokines crucial for γδ T cell function. Both BA and BI reduced γδ T cell infiltration in colorectal tissues, and modulated immune activation markers, with distinct effects on peripheral blood γδ T cell levels. RNA-seq analysis post-probiotic treatment highlighted strain-specific changes, with BA activating NOD2-like receptor signaling and BI enhancing IL-17 and TNF signaling pathways. Direct co-culture experiments demonstrated BI's robust activation of γδ T cells, while BA showed minimal direct effects. Multi-omics correlation analysis suggested that BA and BI modulated immune responses through microenvironment-dependent mechanisms, offering potential therapeutic insights for gut-related inflammatory diseases.</p><p><strong>Conclusions: </strong>Our findings provide a theoretical basis for the development of age-associated probiotic intervention strategies, offering new insights into personalized microbiota modulation to enhance immune health and gut homeostasis across different life stages.</p>","PeriodicalId":12458,"journal":{"name":"Frontiers in Cellular and Infection Microbiology","volume":"15 ","pages":"1639178"},"PeriodicalIF":4.8,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12479543/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145206090","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Pingwei Powder alleviates high-fat diet-induced colonic inflammation by modulating microbial metabolites SCFAs.","authors":"Tangjuan Liu, Guosen Ou, Jialin Wu, Shiqi Wang, Hao Wang, Ziqi Wu, Yawen Jiang, Yaokang Chen, Huachong Xu, Li Deng, Xiaoyin Chen, Lu Xu","doi":"10.3389/fcimb.2025.1628488","DOIUrl":"10.3389/fcimb.2025.1628488","url":null,"abstract":"<p><strong>Background: </strong>Pingwei Powder (PWP), a renowned traditional Chinese medicine (TCM) formula, it has demonstrated excellent therapeutic effects in ulcerative colitis (UC), yet its underlying pharmacological mechanisms remain unclear. This study aims to investigate the therapeutic effect of PWP on the aggravation of colonic inflammation induced by a high-fat diet and particularly focuses on its regulatory mechanisms on gut microbiota, which are closely related to UC.</p><p><strong>Methods: </strong>Network pharmacology analysis was employed to screen potential pharmacological targets of PWP for UC. Histological changes in colonic tissue were observed using hematoxylin and eosin (H&E) staining, and immunofluorescence staining was performed to evaluate the expression of tight junction proteins (ZO1 and Occludin). Western blotting was used to detect the expression levels of proteins related to the PI3K/AKT/mTOR pathway, ZO1, and Occludin. qRT-PCR was conducted to measure the relative expression of inflammatory cytokines (IL-1β, IL-17, IL-6, and TNF-α) in colonic tissue. Additionally, 16S rDNA sequencing was performed to analyze gut microbiota alterations, and GC/MS was used to quantify short-chain fatty acids (SCFAs) in gut contents. The gutMgene database was utilized to validate the mediating roles of gut microbiota metabolites in the pharmacological effects of PWP. And their mediating role in PWP efficacy was verified by fecal microbiota transplantation (FMT) and butyrate supplementation.</p><p><strong>Results: </strong>Network pharmacology analysis predicted that PWP may regulate the PI3K/AKT pathway to exert therapeutic effects in UC. Experimental validation showed that PWP significantly downregulated the levels of PI3K, pAKT/AKT, and pmTOR/mTOR in colonic tissue, thereby enhancing autophagy in colonic epithelial cells, as evidenced by decreased levels of P62 and increased LC3B-II/LC3B-I ratios. Furthermore, 16S rDNA sequencing combined with targeted SCFAs analysis of gut contents revealed that the pharmacological effects of PWP may be mediated by increasing the abundance of SCFAs-producing gut microbiota (<i>Alistipes</i> and <i>Parabacteroides</i>) and elevating the levels of SCFAs in the gut.</p><p><strong>Conclusion: </strong>PWP enhances the abundance of SCFAs-producing bacteria (<i>Alistipes</i> and <i>Parabacteroides</i>) in the gut, increases the levels of butyrate, and inhibits the PI3K/AKT/mTOR pathway in the colon. These effects promote colonic autophagy and contribute to the resolution of colonic inflammation.</p>","PeriodicalId":12458,"journal":{"name":"Frontiers in Cellular and Infection Microbiology","volume":"15 ","pages":"1628488"},"PeriodicalIF":4.8,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12479493/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145206044","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Bowen Dong, Haoran Zhong, Danlin Zhu, Hao Li, Ke Lu, Zhiqiang Fu, Jinming Liu, Yamei Jin
{"title":"Membrane proteomics and transcriptomic profiling analysis of hepatic stellate cells co-incubated with <i>Schistosoma japonicum</i> eggs.","authors":"Bowen Dong, Haoran Zhong, Danlin Zhu, Hao Li, Ke Lu, Zhiqiang Fu, Jinming Liu, Yamei Jin","doi":"10.3389/fcimb.2025.1674880","DOIUrl":"10.3389/fcimb.2025.1674880","url":null,"abstract":"<p><p>Schistosomiasis has been recognized as the second most prevalent parasitic disease worldwide, following malaria. Schistosome eggs can persist for extended periods within host hepatic tissues, leading to hepatic fibrosis primarily through the activation of hepatic stellate cells (HSCs). However, the mechanisms by which egg-secreted products modulate the activation of HSCs remain incompletely elucidated. In this study, purified <i>Schistosoma japonicum</i> (<i>S. japonicum</i>) eggs (Egg group) or corresponding unused medium (Control group) were placed in the upper chamber of a Transwell system, with HSCs cultured in the lower chamber. Following co-culture, HSCs surface proteins were eluted and subsequently analyzed by mass spectrometry. Protein identities were determined by matching spectral data against both human and schistosome protein databases. A total of 88 schistosome proteins, including both <i>S. japonicum</i>-specific and non-specific proteins, were identified in the Egg group. Bioinformatic analyses suggested that HSCs were exposed to egg-derived secretory proteins, indicating potential molecular interactions between schistosome eggs and host cells. Furthermore, RNA-sequencing was performed on HSCs following co-culture, resulting in the identification of 634 differentially expressed genes (DEGs), of which 454 were upregulated and 180 were downregulated. Functional enrichment analyses revealed significant involvement of these DEGs in fibrosis- and inflammation-related pathways. Collectively, this study provides novel evidence that <i>S. japonicum</i> eggs may remotely modulate the transcriptional profiles of HSCs via secreted bioactive molecules, thus offering a theoretical foundation for identifying potential therapeutic targets in hepatic fibrosis.</p>","PeriodicalId":12458,"journal":{"name":"Frontiers in Cellular and Infection Microbiology","volume":"15 ","pages":"1674880"},"PeriodicalIF":4.8,"publicationDate":"2025-09-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12479499/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145206081","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Editorial: Cytokine signaling and innate host defense in modulation of viral infections and the viral evasion.","authors":"M Victoria Delpino, Jorge Quarleri","doi":"10.3389/fcimb.2025.1694793","DOIUrl":"10.3389/fcimb.2025.1694793","url":null,"abstract":"","PeriodicalId":12458,"journal":{"name":"Frontiers in Cellular and Infection Microbiology","volume":"15 ","pages":"1694793"},"PeriodicalIF":4.8,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12477162/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145198880","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ning Li, Fan Yi, Yujun Wang, Feng Geng, Yanli Liu, Qiushuang Liu, Yanan Guo, Ding Long
{"title":"Effects of Shenfu injection on intestinal microbiota and inflammation in sepsis mice.","authors":"Ning Li, Fan Yi, Yujun Wang, Feng Geng, Yanli Liu, Qiushuang Liu, Yanan Guo, Ding Long","doi":"10.3389/fcimb.2025.1599903","DOIUrl":"10.3389/fcimb.2025.1599903","url":null,"abstract":"<p><strong>Introduction: </strong>Sepsis remains a critical challenge in intensive care medicine, necessitating novel therapeutic approaches.</p><p><strong>Methods: </strong>In this study, healthy 8-week-old male C57BL/6J mice were treated with cecal ligation and puncture (CLP) to induce a sepsis model. After successful model establishment, mice in the sham and CLP groups were injected with 200 μL of normal saline, while mice in the SFI group were injected with 200 μL of SFI. Changes in intestinal mucosal barrier function, inflammation, and intestinal microbiota were assessed in septic mice after SFI treatment.</p><p><strong>Results: </strong>SFI treatment significantly ameliorated intestinal inflammation and reduced serum levels of pro-inflammatory cytokines (IL-1β, IL-6) and renal injury markers (SCr, BUN). 16S rRNA sequencing revealed SFI-mediated gut microbial remodeling, characterized by a marked reduction in pathogenic Escherichia-Shigella abundance and concurrent enrichment of beneficial probiotics, including Akkermansia and Lactobacillus. Mechanistically, SFI exhibited dual regulatory effects on both systemic inflammation and gut microbiota homeostasis.</p><p><strong>Discussion: </strong>These findings not only validate SFI's efficacy in sepsis treatment but also propose a novel mechanism involving gut microbiome modulation. This study provides critical experimental evidence for repurposing traditional Chinese medicine in sepsis therapy and establishes a foundation for future research on microbiota-targeted interventions in critical care.</p>","PeriodicalId":12458,"journal":{"name":"Frontiers in Cellular and Infection Microbiology","volume":"15 ","pages":"1599903"},"PeriodicalIF":4.8,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12477228/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145198939","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Transcriptomic profile of the zoonotic parasite <i>Anisakis pegreffii</i> upon <i>in vitro</i> exposure to human dendritic cells.","authors":"Marialetizia Palomba, Aurelia Rughetti, Tiziana Castrignanò, Chiara Napoletano, Xavier Roca-Geronès, Valentina Pinna, Franco Liberati, Daniele Canestrelli, Simonetta Mattiucci","doi":"10.3389/fcimb.2025.1646537","DOIUrl":"10.3389/fcimb.2025.1646537","url":null,"abstract":"<p><p><i>Anisakis pegreffii</i> is a zoonotic marine nematode whose life-cycle involves marine organisms-small crustaceans, fish and squids as intermediate/paratenic hosts, and marine mammals, mainly cetaceans-as definitive ones. When its third-stage larvae (L3) are accidentally ingested by humans with the consumption of raw or undercooked parasitized fish and/or squids, the parasite fails to complete its life cycle, leading to human anisakiasis. Despite increasing interest in understanding the molecular basis of pathogenic effects in human anisakiasis, the transcriptomic response of <i>A. pegreffii</i> L3 to interaction with human immune cells, remains poorly understood. Thus, in this study, the transcriptomic profile of <i>A. pegreffii</i> L3 larvae under <i>in vitro</i> exposure to human dendritic cells (DCs) was performed for the first time. A total of 3914 differentially expressed genes (DEGs) were identified in <i>A. pegreffii</i> L3 after exposure to immature DCs (iDCs), by RNA-seq, allowing to detect 1868 upregulated and 2046 downregulated transcripts. Upregulated genes were significantly enriched in pathways related to energy metabolism, oxidative stress response and structural maintenance, suggesting active metabolic and structural adaptation to immune-induced stress. Conversely, genes involved in cytoskeletal organization and intracellular trafficking were downregulated, potentially reflecting the parasite's developmental arrest in an unsuitable host such as humans. These findings provide novel insights into the molecular response pathways activated by this zoonotic parasite during the early stages of interaction with the human immune system.</p>","PeriodicalId":12458,"journal":{"name":"Frontiers in Cellular and Infection Microbiology","volume":"15 ","pages":"1646537"},"PeriodicalIF":4.8,"publicationDate":"2025-09-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12477248/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145198870","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}