1
|
Xu Z, Qiao S, Wang Z, Peng C, Hou Y, Liu B, Cao G, Wang T. PMA1-containing extracellular vesicles of Candida albicans triggers immune responses and colitis progression. Gut Microbes 2025; 17:2455508. [PMID: 39886799 PMCID: PMC11792855 DOI: 10.1080/19490976.2025.2455508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 02/01/2025] Open
Abstract
Candida albicans (C. albicans) exhibits aberrant changes in patients with colitis, and it has been reported to dominate the colonic mucosal immune response. Here, we found that PMA1 expression was significantly increased in C. albicans from patients with IBD compared to that in healthy controls. A Crispr-Cas9-based fungal strain editing system was then used to knock out PMA1 expression in C. albicans. Compared to WT-C.a, ΔPMA1-C.a could not aggravate colitis. Proteomic analysis showed that PMA1 was transported by extracellular vesicles (EVs) of C. albicans. PMA1-containing EVs aggravated colitis, modulated the migration of cDC2 from the lamina propria to mesenteric lymph nodes, and induced TH17 cell differentiation. Moreover, the adaptor protein CARD9 was critical in PMA1-containing EV-induced colitis, and CARD9-deficient DCs did not induce TH17 cell differentiation or IL-17A production. Mechanically, CARD9 combines with the glycolytic protein GAPDH (aa2-146 domain) through its CARD region. CARD9 deficiency led to decreased enzyme activity of GAPDH and decreased glycolysis of DCs. These findings indicate that PMA1 is a potential virulence factor responsible for the pathogenesis of C. albicans colitis.
Collapse
Affiliation(s)
- Zhen Xu
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Shuping Qiao
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Zelin Wang
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Chen Peng
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Yayi Hou
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
| | - Guochun Cao
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Tingting Wang
- Department of Oncology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- The Comprehensive Cancer Centre of Drum Tower Hospital, Medical School of Nanjing University & Clinical Cancer Institute of Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
2
|
Ríos Colombo NS, Paul Ross R, Hill C. Synergistic and off-target effects of bacteriocins in a simplified human intestinal microbiome: implications for Clostridioides difficile infection control. Gut Microbes 2025; 17:2451081. [PMID: 39817466 PMCID: PMC11740676 DOI: 10.1080/19490976.2025.2451081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 12/04/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025] Open
Abstract
Clostridioides difficile is a major cause of nosocomial diarrhea. As current antibiotic treatment failures and recurrence of infections are highly frequent, alternative strategies are needed for the treatment of this disease. This study explores the use of bacteriocins, specifically lacticin 3147 and pediocin PA-1, which have reported inhibitory activity against C. difficile. We engineered Lactococcus lactis strains to produce these bacteriocins individually or in combination, aiming to enhance their activity against C. difficile. Our results show that lacticin 3147 and pediocin PA-1 display synergy, resulting in higher anti-C. difficile activity. We then evaluated the effects of these L. lactis strains in a Simplified Human Intestinal Microbiome (SIHUMI-C) model, a bacterial consortium of eight diverse human gut species that includes C. difficile. After introducing the bacteriocin-producing L. lactis strains into SIHUMI-C, samples were collected over 24 hours, and the genome copies of each species were assessed using qPCR. Contrary to expectations, the combined bacteriocins increased C. difficile levels in the consortium despite showing synergy against C. difficile in agar-based screening. This can be rationally explained by antagonistic inter-species interactions within SIHUMI-C, providing new insights into how broad-spectrum antimicrobials might fail to control targeted species in complex gut microbial communities. These findings highlight the need to mitigate off-target effects in complex gut microbiomes when developing bacteriocin-based therapies with potential clinical implications for infectious disease treatment.
Collapse
Affiliation(s)
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Colin Hill
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| |
Collapse
|
3
|
Kurilovich E, Geva-Zatorsky N. Effects of bacteriophages on gut microbiome functionality. Gut Microbes 2025; 17:2481178. [PMID: 40160174 PMCID: PMC11959909 DOI: 10.1080/19490976.2025.2481178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/28/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
The gut microbiome, composed of bacteria, fungi, and viruses, plays a crucial role in maintaining the delicate balance of human health. Emerging evidence suggests that microbiome disruptions can have far-reaching implications, ranging from the development of inflammatory diseases and cancer to metabolic disorders. Bacteriophages, or "phages", are viruses that specifically infect bacterial cells, and their interactions with the gut microbiome are receiving increased attention. Despite the recently revived interest in the gut phageome, it is still considered the "dark matter" of the gut, with more than 80% of viral genomes remaining uncharacterized. Today, research is focused on understanding the mechanisms by which phages influence the gut microbiota and their potential applications. Bacteriophages may regulate the relative abundance of bacterial communities, affect bacterial functions in various ways, and modulate mammalian host immunity. This review explores how phages can regulate bacterial functionality, particularly in gut commensals and pathogens, emphasizing their role in gut health and disease.
Collapse
Affiliation(s)
- Elena Kurilovich
- Department of Cell Biology and Cancer Science, Rappaport Technion Integrated Cancer Center (RTICC), Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, Israel
| | - Naama Geva-Zatorsky
- Department of Cell Biology and Cancer Science, Rappaport Technion Integrated Cancer Center (RTICC), Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, Israel
- Humans and the Microbiome program, CIFAR, Toronto, ON, Canada
| |
Collapse
|
4
|
Wang L, Chen X, Pollock NR, Villafuerte Gálvez JA, Alonso CD, Wang D, Daugherty K, Xu H, Yao J, Chen Y, Kelly CP, Cao Y. Metagenomic analysis reveals distinct patterns of gut microbiota features with diversified functions in C. difficile infection (CDI), asymptomatic carriage and non-CDI diarrhea. Gut Microbes 2025; 17:2505269. [PMID: 40366862 PMCID: PMC12080279 DOI: 10.1080/19490976.2025.2505269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/12/2025] [Accepted: 05/07/2025] [Indexed: 05/16/2025] Open
Abstract
Clostridioides difficile infection (CDI) has been recognized as a leading cause of healthcare-associated infections and a considerable threat to public health globally. Increasing evidence suggests that the gut microbiota plays a key role in the pathogenesis of CDI. The taxonomic composition and functional capacity of the gut microbiota associated with CDI have not been studied systematically. Here, we performed a comprehensive shotgun metagenomic sequencing in a well-characterized human cohort to reveal distinct patterns of gut microbiota and potential functional features associated with CDI. Fecal samples were collected from 104 inpatients, including : (1) patients with clinically significant diarrhea and positive nucleic acid amplification testing (NAAT) and received CDI treatment (CDI, n = 47); (2) patients with positive stool NAAT but without diarrhea (Carrier, n = 17); (3) patients with negative stool NAAT but with diarrhea (Diarrhea, n = 14); and (4) patients with negative stool NAAT and without diarrhea (Control, n = 26). Downstream statistical analyses (including alpha and beta diversity analysis, differential abundance analysis, correlation network analysis, and potential functional analysis) were then performed. The gut microbiota in the Control group showed higher Chao1 index (p < 0.05), while Shannon index at KEGG module level was higher in CDI than in Carrier and Control (p < 0.05). Beta diversity for species composition differed significantly between CDI vs Carrier/Control cohorts (p < 0.05). Microbial Linear discriminant analysis Effect Size and ANCOM analysis both identified 8 species (unclassified_f_Enterobacteriaceae, Veillonella_parvula, unclassified_g_Klebsiella and etc.) were enriched in CDI, Enterobacter_aerogenes was enriched in Diarrhea, Collinsella_aerofaciens, Collinsella_sp_4_8_47FAA, Collinsella_tanakaei and Collinsella_sp_CAG_166 were enriched in Control (LDA >3.0, adjusted p < 0.05). Correlation network complexity was higher in CDI with more negative correlations than in other three cohorts. Modules involved in iron complex transport system (M00240) was enriched in CDI, ABC-2 type transport system (M00254), aminoacyl-tRNA biosynthesis (M00359), histidine biosynthesis (M00026) and inosine monophosphate biosynthesis (M00048) were enriched in Carrier, ribosome (M00178 and M00179) was enriched in Diarrhea, fluoroquinolone resistance (M00729) and aminoacyl-tRNA biosynthesis (M00360) were enriched in Control (LDA > 2.5, adjusted p < 0.05). Resistance functions of acriflavine and glycylcycline were enriched in CDI, while resistance function of bacitracin was enriched in Carrier (LDA > 3.0, adjusted p < 0.05), and the contributions of phylum and species to resistance functions differed among the four groups. Our results reveal alterations of gut microbiota composition and potential functions among four groups of differential colonization/infection status of Clostridioides difficile. These findings support the potential roles of gut microbiota and their potential functions in the pathogenesis of CDI.
Collapse
Affiliation(s)
- Lamei Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Xinhua Chen
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nira R. Pollock
- Division of Infectious Disease, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Laboratory Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Javier A. Villafuerte Gálvez
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Carolyn D. Alonso
- Division of Infectious Disease, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Dangdang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Kaitlyn Daugherty
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Hua Xu
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yulin Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Ciaran P. Kelly
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yangchun Cao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Mukhopadhya I, Martin JC, Shaw S, Gutierrez-Torrejon M, Boteva N, McKinley AJ, Gratz SW, Scott KP. Novel insights into carbohydrate utilisation, antimicrobial resistance, and sporulation potential in Roseburia intestinalis isolates across diverse geographical locations. Gut Microbes 2025; 17:2473516. [PMID: 40089923 PMCID: PMC11913394 DOI: 10.1080/19490976.2025.2473516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 02/11/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
Roseburia intestinalis is one of the most abundant and important butyrate-producing human gut anaerobic bacteria that plays an important role in maintaining health and is a potential next-generation probiotic. We investigated the pangenome of 16 distinct strains, isolated over several decades, identifying local and time-specific adaptations. More than 50% of the genes in each individual strain were assigned to the core genome, and 77% of the cloud genes were unique to individual strains, revealing the high level of genome conservation. Co-carriage of the same enzymes involved in carbohydrate binding and degradation in all strains highlighted major pathways in carbohydrate utilization and reveal the importance of xylan, starch and mannose as key growth substrates. A single strain had adapted to use rhamnose as a sole growth substrate, the first time this has been reported. The ubiquitous presence of motility and sporulation gene clusters demonstrates the importance of these phenotypes for gut survival and acquisition of this bacterium. More than half the strains contained functional, potentially transferable, tetracycline resistance genes. This study advances our understanding of the importance of R. intestinalis within the gut ecosystem by elucidating conserved metabolic characteristics among different strains, isolated from different locations. This information will help to devise dietary strategies to increase the abundance of this species providing health benefits.
Collapse
Affiliation(s)
- Indrani Mukhopadhya
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
- Microbiology and Immunity, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Jennifer C. Martin
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Sophie Shaw
- Centre for Genome Enabled Biology and Medicine, University of Aberdeen, Aberdeen, UK
- All Wales Medical Genomics Service, Institute of Medical Genetics, University Hospital of Wales, Heath Park, Cardiff, UK
| | | | - Nikoleta Boteva
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Aileen J. McKinley
- Department of Surgery, Aberdeen Royal Infirmary Foresterhill, Aberdeen, UK
| | - Silvia W. Gratz
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Karen P. Scott
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
6
|
Barrios Steed D, Koundakjian D, Harris AD, Rosato AE, Konstantinidis KT, Woodworth MH. Leveraging strain competition to address antimicrobial resistance with microbiota therapies. Gut Microbes 2025; 17:2488046. [PMID: 40195644 PMCID: PMC11988218 DOI: 10.1080/19490976.2025.2488046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 12/28/2024] [Accepted: 03/28/2025] [Indexed: 04/09/2025] Open
Abstract
The enteric microbiota is an established reservoir for multidrug-resistant organisms that present urgent clinical and public health threats. Observational data and small interventional studies suggest that microbiome interventions, such as fecal microbiota products and characterized live biotherapeutic bacterial strains, could be an effective antibiotic-sparing prevention approach to address these threats. However, bacterial colonization is a complex ecological phenomenon that remains understudied in the context of the human gut. Antibiotic resistance is one among many adaptative strategies that impact long-term colonization. Here we review and synthesize evidence of how bacterial competition and differential fitness in the context of the gut present opportunities to improve mechanistic understanding of colonization resistance, therapeutic development, patient care, and ultimately public health.
Collapse
Affiliation(s)
- Danielle Barrios Steed
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Anthony D. Harris
- Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
- Institute for Healthcare Computing, University of Maryland, Baltimore, MD, USA
| | - Adriana E Rosato
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
| | | | - Michael H Woodworth
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
7
|
Jamerlan AM, An SSA, Hulme JP. Microbial diversity and fitness in the gut-brain axis: influences on developmental risk for Alzheimer's disease. Gut Microbes 2025; 17:2486518. [PMID: 40207973 PMCID: PMC11988266 DOI: 10.1080/19490976.2025.2486518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
The gut-brain axis (GBA) denotes the dynamic and bidirectional communication system that connects the gastrointestinal tract and the central nervous system (CNS). This review explored this axis, focusing on the role of microbial diversity and fitness in maintaining gastrointestinal health and preventing neurodegeneration, particularly in Alzheimer's disease (AD). Gut dysbiosis, characterized by the imbalance in populations of beneficial and harmful bacteria, has been associated with increased systemic inflammation, neuroinflammation, and the progression of AD through pathogenic mechanisms involving amyloid deposition, tauopathy, and increased blood-brain barrier (BBB) permeability. Emerging evidence highlighted the therapeutic potential of probiotics, dietary interventions, and intermittent fasting in restoring microbial balance, reducing inflammation, and minimizing neurodegenerative risks. Probiotics and synbiotics are promising in helping improve cognitive function and metabolic health, while dietary patterns like the Mediterranean diet were linked to decreased neuroinflammation and enhanced gut-brain communication. Despite significant advancement, further research is needed to elucidate the specific microbial strains, metabolites, and mechanisms influencing brain health. Future studies employing longitudinal designs and advanced omics technologies are essential to developing targeted microbiome-based therapies for managing AD-related disorders.
Collapse
Affiliation(s)
- Angelo M. Jamerlan
- Department of Bionanotechnology, Bionano Research Institute, Gachon University, Seongnam-si, Republic of Korea
| | - Seong Soo A. An
- Department of Bionanotechnology, Bionano Research Institute, Gachon University, Seongnam-si, Republic of Korea
| | - John P. Hulme
- Department of Bionanotechnology, Bionano Research Institute, Gachon University, Seongnam-si, Republic of Korea
| |
Collapse
|
8
|
Li XL, Megdadi M, Quadri HS. Interaction between gut virome and microbiota on inflammatory bowel disease. World J Methodol 2025; 15:100332. [DOI: 10.5662/wjm.v15.i3.100332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/31/2024] [Accepted: 01/15/2025] [Indexed: 03/06/2025] Open
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease and ulcerative colitis, is a chronic condition marked by recurring gastrointestinal inflammation. While immune, genetic, and environmental factors are well-studied, the gut virome has received less attention. This editorial highlights the work which investigates the gut virome’s role in IBD and its interactions with the bacterial microbiome and host immune system. The gut virome consists of bacteriophages, eukaryotic viruses, and endogenous retroviruses. Among these, Caudovirales bacteriophages are predominant and influence bacterial communities via lysogenic and lytic cycles. Eukaryotic viruses infect host cells directly, while endogenous retroviruses impact gene regulation and immune responses. In IBD, the virome shows distinct alterations, including an increased abundance of Caudovirales phages and reduced Microviridae diversity, suggesting a pro-inflammatory viral environment. Dysbiosis, chronic inflammation, and aberrant immune responses contribute to these changes by disrupting microbial communities and modifying virome composition. Phages affect bacterial dynamics through lysis, lysogeny, and horizontal gene transfer, shaping microbial adaptability and resilience. Understanding these interactions is crucial for identifying novel therapeutic targets and restoring microbial balance in IBD.
Collapse
Affiliation(s)
- Xiao-Long Li
- Department of Surgery, Ascension St Agnes Hospital, Baltimore, MD 21009, United States
| | - Mueen Megdadi
- Department of Surgery, Ascension St Agnes Hospital, Baltimore, MD 21009, United States
| | - Humair S Quadri
- Department of Surgery, Ascension St Agnes Hospital, Baltimore, MD 21009, United States
| |
Collapse
|
9
|
Kulkarni H, Gaikwad AB. The mitochondria-gut microbiota crosstalk - A novel frontier in cardiovascular diseases. Eur J Pharmacol 2025; 998:177562. [PMID: 40157703 DOI: 10.1016/j.ejphar.2025.177562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/06/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Cardiovascular diseases (CVDs), including hypertension, atherosclerosis, and cardiomyopathy among others, remain the leading cause of global morbidity and mortality. Despite advances in treatment, the complex pathophysiology of CVDs necessitates innovative approaches to improve patient outcomes. Recent research has uncovered a dynamic interplay between mitochondria and gut microbiota, fundamentally altering our understanding of cardiovascular health. However, while existing studies have primarily focused on individual components of this axis, this review examines the bidirectional communication between these biological systems and their collective impact on cardiovascular health. Mitochondria, serving as cellular powerhouses, are crucial for maintaining cardiovascular homeostasis through oxidative phosphorylation (OXPHOS), calcium regulation, and redox balance. Simultaneously, the gut microbiota influences cardiovascular function through metabolite production, barrier integrity maintenance, and immune system modulation. The mitochondria-gut microbiota axis operates through various molecular mechanisms, including microbial metabolites such as trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFA), and secondary bile acids, which directly influence mitochondrial function. Conversely, mitochondrial stress signals and damage-associated molecular patterns (DAMPs) affect gut microbial communities and barrier function. Key signalling pathways, including AMP-activated protein kinase (AMPK), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), and the silent information regulator 1-peroxisome proliferator-activated receptor gamma coactivator 1-alpha (SIRT1-PGC-1α) axis, integrate these interactions, highlighting their role in CVD pathogenesis. Understanding these interactions has revealed promising therapeutic targets, suggesting new therapies aimed at both mitochondrial function and gut microbiota composition. Thus, this review provides a comprehensive framework for leveraging the mitochondria-gut microbiota axis in providing newer therapeutics for CVDs by targeting the AMPK/SIRT-1/PGC-1α/NF-κB signalling.
Collapse
Affiliation(s)
- Hrushikesh Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Vidya Vihar, Pilani, Rajasthan 333031, India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Campus, Vidya Vihar, Pilani, Rajasthan 333031, India.
| |
Collapse
|
10
|
Das M, Kiruthiga C, Shafreen RB, Nachammai K, Selvaraj C, Langeswaran K. Harnessing the human microbiome and its impact on immuno-oncology and nanotechnology for next-generation cancer therapies. Eur J Pharmacol 2025; 996:177436. [PMID: 40023356 DOI: 10.1016/j.ejphar.2025.177436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
The integration of microbiome research and nanotechnology represents a significant advancement in immuno-oncology, potentially improving the effectiveness of cancer immunotherapies. Recent studies highlight the influential role of the human microbiome in modulating immune responses, presenting new opportunities to enhance immune checkpoint inhibitors (ICIs) and other cancer therapies. Nanotechnology offers precise drug delivery and immune modulation capabilities, minimizing off-target effects while maximizing therapeutic outcomes. This review consolidates current knowledge on the interactions between the microbiome and the immune system, emphasizing the microbiome's impact on ICIs, and explores the incorporation of nanotechnology in cancer treatment strategies. Additionally, it provides a forward-looking perspective on the synergistic potential of microbiome modulation and nanotechnology to overcome existing challenges in immuno-oncology. This integrated approach may enhance the personalization and effectiveness of next-generation cancer treatments, paving the way for transformative patient care.
Collapse
Affiliation(s)
- Mamali Das
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India
| | | | - R Beema Shafreen
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India
| | - Kathiresan Nachammai
- Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India
| | - Chandrabose Selvaraj
- CsrDD Lab, Department of Microbiology, Dr. D. Y. Patil Medical College Hospital & Research Centre, Dr. D. Y. Patil Vidyapeeth (Deemed to Be University), Pimpri, Pune, 411018, India.
| | - K Langeswaran
- Department of Biomedical Science, Alagappa University, Karaikudi, 630003, India; Department of Biotechnology, Alagappa University, Science Campus, Karaikudi, Tamil Nadu, India.
| |
Collapse
|
11
|
Chong-Nguyen C, Yilmaz B, Coles B, Sokol H, MacPherson A, Siepe M, Reineke D, Mosbahi S, Tomii D, Nakase M, Atighetchi S, Ferro C, Wingert C, Gräni C, Pilgrim T, Windecker S, Blasco H, Dupuy C, Emond P, Banz Y, Losmanovà T, Döring Y, Siontis GCM. A scoping review evaluating the current state of gut microbiota and its metabolites in valvular heart disease physiopathology. Eur J Clin Invest 2025; 55:e14381. [PMID: 39797472 DOI: 10.1111/eci.14381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND The human microbiome is crucial in regulating intestinal and systemic functions. While its role in cardiovascular disease is better understood, the link between intestinal microbiota and valvular heart diseases (VHD) remains largely unexplored. METHODS Peer-reviewed studies on human, animal or cell models analysing gut microbiota profiles published up to April 2024 were included. Eligible studies used 16S rRNA or shotgun sequencing, metabolite profiling by mass spectrometry, and examined osteogenesis or fibrosis signalling in valve cells. Methods and findings were qualitatively analysed, with data charted to summarize study design, materials and outcomes. RESULTS Thirteen studies were included in the review: five human, three animal and five in vitro. Of the nine studies on calcific aortic stenosis (CAS), elevated trimethylamine N-oxide (TMAO) levels were linked to an increased risk of cardiovascular events in cohort studies, with CAS patients showing higher levels of Bacteroides plebeius, Enterobacteriaceae, Veillonella dispar and Prevotella copri. In vivo, TMAO promoted aortic valve fibrosis, while tryptophan derivatives stimulated osteogenic differentiation and interleukin-6 secretion in valvular interstitial cells. Two studies on rheumatic mitral valve disease found altered microbiota profiles and lower short-chain fatty acid levels, suggesting potential impacts on immune regulation. Two studies on Barlow's mitral valve disease in animal models revealed elevated TMAO levels in dogs with congestive heart failure, reduced Paraprevotellaceae, increased Actinomycetaceae and dysbiosis involving Turicibacter and E. coli. CONCLUSIONS TMAO has been mainly identified as a prognostic marker in VHD. Gut microbiota dysbiosis has been observed in various forms of VHD and deserve further study.
Collapse
Affiliation(s)
| | - Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Bernadette Coles
- Velindre University NHS Trust Library and Knowledge Service, Cardiff, UK
| | - Harry Sokol
- Department of Gastroenterology, Saint Antoine Hospital, Assistance Publique-Hopitaux de Paris (APHP), Paris, France
| | - Andrew MacPherson
- Department of Visceral Surgery and Medicine, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Matthias Siepe
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - David Reineke
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Selim Mosbahi
- Department of Cardiac Surgery, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Daijiro Tomii
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Masaaki Nakase
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Sarah Atighetchi
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Cyril Ferro
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Christoph Wingert
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Christoph Gräni
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Thomas Pilgrim
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Stephan Windecker
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Hélène Blasco
- Faculté de médecine, Equipe neurogénétique et neurométabolomique, INSERM U930, Université François Rabelais, Tours, France
| | - Camille Dupuy
- Faculté de médecine, Equipe neurogénétique et neurométabolomique, INSERM U930, Université François Rabelais, Tours, France
| | - Patrick Emond
- Faculté de médecine, Equipe neurogénétique et neurométabolomique, INSERM U930, Université François Rabelais, Tours, France
| | - Yara Banz
- Institute of Tissue Medicine and Pathology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Tereza Losmanovà
- Institute of Tissue Medicine and Pathology, Bern University Hospital, Inselspital, Bern, Switzerland
| | - Yvonne Döring
- Department of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
| | - George C M Siontis
- Department of Cardiology, Bern University Hospital, Inselspital, Bern, Switzerland
| |
Collapse
|
12
|
Burananat T, Wilantho A, Kulalert P, Nanthapisal S, Tonglim J, Deetienin W, Wangkumhang P, Tongsima S, Thaweekul P. The role of gut microbiota in obesity severity and metabolic risk in pediatric populations. Nutr Metab Cardiovasc Dis 2025; 35:103970. [PMID: 40180829 DOI: 10.1016/j.numecd.2025.103970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/12/2025] [Accepted: 03/04/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND AND AIMS Childhood obesity is a considerable public health issue. Recent research has shown that alterations in gut microbiota can have an impact on developing obesity and other metabolic health problems in children. This study aimed to investigate whether the characteristics of gut microbiota in obese children and adolescents are associated with the severity of obesity and any metabolic complications. METHODS AND RESULTS During May 2022 to May 2023, a total of 56 children and adolescents with obesity, aged 6-18 years, were recruited at Thammasat Hospital, situated in provincial Pathumthani in central Thailand. Participants were allocated into two groups, characterized by the severity of their obesity. Demographic data, body composition, along with resting energy expenditures were determined. Serum samples were collected for the metabolic profile and inflammatory markers. Fecal samples were obtained for gut microbiota analysis via 16S rRNA Illumina. The obese group exhibited notably greater relative abundance of Actinobacteriota in comparison to the severely obese group, along with a lower abundance of Bacteroidota. There were no statistically significant differences in the relative abundance of Firmicutes and the Firmicutes to Bacteroidota ratio between the two cohorts. Bacteroidota positively correlated with FMI, while Actinobacteriota showed a negative correlation with FMI. CONCLUSION The data gathered from this study illustrated that children and adolescents with obesity and severe obesity in Thailand showed differences in the relative abundance of Actinobacteriota and Bacteroidota. Certain microbiome taxa showed correlations with various body and metabolic parameters.
Collapse
Affiliation(s)
- Thanyamas Burananat
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Alisa Wilantho
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Prapasri Kulalert
- Department of Clinical Epidemiology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Sira Nanthapisal
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | | | | | - Pongsakorn Wangkumhang
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Sissades Tongsima
- National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Patcharapa Thaweekul
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand.
| |
Collapse
|
13
|
Garcia-Morena D, Fernandez-Cantos MV, Escalera SL, Lok J, Iannone V, Cancellieri P, Maathuis W, Panagiotou G, Aranzamendi C, Aidy SE, Kolehmainen M, El-Nezami H, Wellejus A, Kuipers OP. In Vitro Influence of Specific Bacteroidales Strains on Gut and Liver Health Related to Metabolic Dysfunction-Associated Fatty Liver Disease. Probiotics Antimicrob Proteins 2025; 17:1498-1512. [PMID: 38319537 PMCID: PMC12055940 DOI: 10.1007/s12602-024-10219-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/07/2024]
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) has become a major health risk and a serious worldwide issue. MAFLD typically arises from aberrant lipid metabolism, insulin resistance, oxidative stress, and inflammation. However, subjacent causes are multifactorial. The gut has been proposed as a major factor in health and disease, and over the last decade, bacterial strains with potentially beneficial effects on the host have been identified. In vitro cell models have been commonly used as an early step before in vivo drug assessment and can confer complementary advantages in gut and liver health research. In this study, several selected strains of the order Bacteroidales were used in a three-cell line in vitro analysis (HT-29, Caco-2, and HepG2 cell lines) to investigate their potential as new-generation probiotics and microbiota therapeutics. Antimicrobial activity, a potentially useful trait, was studied, and the results showed that Bacteroidales can be a source of either wide- or narrow-spectrum antimicrobials targeting other closely related strains. Moreover, Bacteroides sp. 4_1_36 induced a significant decrease in gut permeability, as evidenced by the high TEER values in the Caco-2 monolayer assay, as well as a reduction in free fatty acid accumulation and improved fatty acid clearance in a steatosis HepG2 model. These results suggest that Bacteroidales may spearhead the next generation of probiotics to prevent or diminish MAFLD.
Collapse
Affiliation(s)
- Diego Garcia-Morena
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Maria Victoria Fernandez-Cantos
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Silvia Lopez Escalera
- Chr. Hansen A/S, Bøge Allé 10-12, 2970, Hørsholm, Denmark
- Friedrich-Schiller Universität Jena, Fakultät für Biowissenschaften, 18K, 07743, Bachstraβe, Germany
| | - Johnson Lok
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200, Kuopio, Finland
| | - Valeria Iannone
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200, Kuopio, Finland
| | - Pierluca Cancellieri
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Willem Maathuis
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology (Leibniz-HKI), 07745, Jena, Germany
- Department of Medicine and State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China
- Faculty of Biological Sciences, Friedrich Schiller University, 07745, Jena, Germany
| | - Carmen Aranzamendi
- Groningen Biomolecular Sciences and Biotechnology Institute, Host-Microbe Metabolic Interactions, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands
| | - Sahar El Aidy
- Groningen Biomolecular Sciences and Biotechnology Institute, Host-Microbe Metabolic Interactions, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands
| | - Marjukka Kolehmainen
- School of Medicine, Institute of Public Health and Clinical Nutrition, University of Eastern Finland, 70200, Kuopio, Finland
| | - Hani El-Nezami
- Molecular and Cell Biology Division, School of Biological Sciences, University of Hong Kong, Pok Fu Lam, Hong Kong SAR
| | - Anja Wellejus
- Chr. Hansen A/S, Bøge Allé 10-12, 2970, Hørsholm, Denmark
| | - Oscar P Kuipers
- Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, The Netherlands.
| |
Collapse
|
14
|
He F, Jin X, Sun K, Zhao L, Yang W, Zhang X, Dong X, Zhao Y, Pan L, Bao N, Sun H. Bacillus subtilis JATP-3 Improves Nitrogen Metabolism by Regulating Intestinal Flora and AKG in Weaned Piglets. Probiotics Antimicrob Proteins 2025; 17:1265-1276. [PMID: 38079031 DOI: 10.1007/s12602-023-10196-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2023] [Indexed: 05/07/2025]
Abstract
Recently, it has been reported that oral probiotics improve the apparent digestibility of nitrogen in weaned piglets; however, the underlying mechanism is unclear. A total of 12 crossbred piglets (Yorkshire × Landrace; 28 days old) were randomly divided into two groups. The control (Con) group was fed with a basic diet + Luria-Bertani (LB; sterile; 10 mL), whereas the subject (Sub) group was fed with a basic diet + B. subtilis JATP-3 (1 × 109 CFU/mL; 10 mL). The results showed that feeding B. subtilis JATP-3 increased the final body weight and nitrogen deposition rate of weaned piglets (P < 0.05); while the daily weight gain showed an upward trend (P < 0.1). The abundance of Pedicoccus, Collinella, Turiciator, Veillonella, Clostridium, and Escherichia were significantly increased in the jejunum (P < 0.05). The abundance of Olsenella and Pediococcus were significantly increased in the ileum (P < 0.05). The metabolomics analysis showed that the levels of l-lactic acid and Alpha-ketoglutaric acid (AKG) in portal vein plasma were significantly increased (P < 0.05). In addition, the content of AKG in muscle and liver increased significantly (P < 0.01). The metagenomics analysis showed that Veillonella encoded the functional genes of 2-oxoglutarate synthase and promoted AKG production. The protein expression of eIF4E-binding protein 1 (4EBP1) phosphorylated in the skeletal muscle increased (P < 0.05). In summary, B. subtilis JATP-3 promotes dietary nitrogen metabolism and skeletal muscle synthesis by modulating the intestinal microbiota and its metabolites, in which AKG may be one of the main mediators of the therapeutic effects of B. subtilis JATP-3.
Collapse
Affiliation(s)
- Feng He
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun, 130118, People's Republic of China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Xueying Jin
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun, 130118, People's Republic of China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Kecheng Sun
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun, 130118, People's Republic of China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Lei Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Wenyan Yang
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun, 130118, People's Republic of China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Xuefeng Zhang
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun, 130118, People's Republic of China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Xiaoqing Dong
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun, 130118, People's Republic of China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Yuan Zhao
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun, 130118, People's Republic of China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Li Pan
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun, 130118, People's Republic of China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Nan Bao
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun, 130118, People's Republic of China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
| | - Hui Sun
- College of Animal Science and Technology, Jilin Agricultural University, No. 2888 Xincheng Street, Changchun, 130118, People's Republic of China.
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, China.
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China.
| |
Collapse
|
15
|
Kumar M, Muthurayar T, Karthika S, Gayathri S, Varalakshmi P, Ashokkumar B. Anti-Diabetic Potentials of Lactobacillus Strains by Modulating Gut Microbiota Structure and β-Cells Regeneration in the Pancreatic Islets of Alloxan-Induced Diabetic Rats. Probiotics Antimicrob Proteins 2025; 17:1096-1116. [PMID: 38329697 DOI: 10.1007/s12602-024-10221-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/09/2024]
Abstract
Diabetes mellitus, a most common endocrine disorder of glucose metabolism, has become a global epidemic and poses a serious public health threat with an increased socio-economic burden. Escalating incidence of diabetes is correlated with changes in lifestyle and food habits that cause gut microbiome dysbiosis and β-cells damage, which can be addressed with dietary interventions containing probiotics. Hence, the search for probiotics of human origin with anti-diabetic, anti-AGE, and anti-ACE potentials has gained renewed interest for the effective management of diabetes and its associated complications. The present study used an alloxan (AXN)-induced diabetic rat model to investigate the effects of potential probiotic Lacticaseibacillus casei MKU1, Lactiplantibacillus pentosus MKU3, and Lactiplantibacillus plantarum MKU7 administration individually on physiochemical parameters related to diabetic pathogenesis. Experimental animals were randomly allotted into six groups viz. NCG (control), DCG (AXN), DGM (metformin), DGP1 (MKU1), DGP2 (MKU3), and DGP3 (MKU7), and biochemical data like serum glucose, insulin, AngII, ACE, HbA1c, and TNF-α levels were measured until 90 days. Our results suggest that oral administration with MKU1, MKU3, or MKU7 significantly improved serum insulin levels, glycemic control, glucose tolerance, and body weight. Additionally, β-cell mass was increased by preserving islet integrity in Lactobacillus-treated diabetic rats, whereas TNF-α (~40%), AngII (~30%), and ACE levels (~50%) were strongly inhibited and enhanced sIgA production (5.8 folds) abundantly. Furthermore, Lactobacillus administration positively influenced the gut microbiome with a significant increase in the abundance of Lactobacillus species and the beneficial Bacteroides uniformis and Bacteroides fragilis, while decreased the pathogenic Proteus vulgaris and Parabacteroides distasonis. Among the probiotic treatment groups, L. pentosus MKU3 performed greatly in almost all parameters, indicating its potential use for alleviating diabetes-associated complications.
Collapse
Affiliation(s)
- Manoj Kumar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Tharmar Muthurayar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Sukumaran Karthika
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Santhalingam Gayathri
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India
| | - Perumal Varalakshmi
- Department of Molecular Microbiology, School of Biotechnology, Madurai Kamaraj University, Madurai, India
| | - Balasubramaniem Ashokkumar
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, 625 021, India.
| |
Collapse
|
16
|
Hunjan G, Shah SS, Kosey S, Aran KR. Gut microbiota and the tryptophan-kynurenine pathway in anxiety: new insights and treatment strategies. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02938-8. [PMID: 40369368 DOI: 10.1007/s00702-025-02938-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
Anxiety disorders are mental health disorders characterized by long-lasting fear, worry, nervousness, and alterations in gut microbiota (GM). The GM is a vital modulator of brain function through the gut-brain axis, which acts as the neural pathway between the central and peripheral nervous systems. Dysbiosis of GM plays an essential role in anxiety development because of alterations in the vagus nerve, increased intestinal permeability, and altered breakdown of tryptophan (TRP). The Kynurenine (KYN) pathway plays a crucial role in the pathogenesis of anxiety disorders, primarily through its neuroprotective (KYNA) and neurotoxic (QUIN) metabolites. Higher ratios of KYNA/QUIN result in neuroprotection, whereas higher KYN/TRP ratios indicate increased QUIN production causing neuroinflammation. Studies on germ-free models exhibit higher plasma TRP levels, which interrupt the metabolic balance of TRP-derived compounds, thus causing brain impairment. A key issue in anxiety disorders is the dysregulation of GM, which disrupts TRP metabolism and neuroinflammatory pathways, however, remains poorly understood. Hence, the proper understanding of these mechanisms is crucial for future therapeutic advancements. Here, we highlight the significance of the TRP-KYN pathway and the potential of modulating KYN pathway enzymes, such as kynurenine aminotransferases (KATs), to adjust KYNA levels and restore neurotransmitter balance. It further discusses new therapeutic methods with a particular focus on probiotics that may restore GM and modulate TRP metabolism. Advancing our understanding of the intricate relationship between GM and anxiety disorders may facilitate novel, microbiota-targeted interventions. This ultimately contributes to precision medicine approaches in mental health care, thereby enhancing treatment efficacy and patient outcomes.
Collapse
Affiliation(s)
- Garry Hunjan
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Shiv Shankar Shah
- Krupanidhi College of Pharmacy, Carmelaram Gunjur Road, Hobli, off Sarjapur Road, Varthur, Bengaluru, 560035, Karnataka, India
| | - Sourabh Kosey
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
17
|
Chen Y, Xie Y, Yu X. Progress of research on the gut microbiome and its metabolite short-chain fatty acids in postmenopausal osteoporosis: a literature review. Front Med 2025:10.1007/s11684-025-1129-3. [PMID: 40347368 DOI: 10.1007/s11684-025-1129-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 12/16/2024] [Indexed: 05/12/2025]
Abstract
Postmenopausal osteoporosis (PMOP) is a systemic metabolic bone disease caused by the decrease in estrogen levels after menopause. It leads to bone loss, microstructural damage, and an increased risk of fractures. Studies have found that the gut microbiota and its metabolites can regulate bone metabolism through the gut-bone axis and the gut-brain axis. As research progresses, PMOP has been found to be associated with gut microbiota dysbiosis and Th17/Treg imbalance. The gut microbiota is closely related to the development and differentiation of Treg and Th17 cells. Among them, the metabolites of the gut microbiota such as short-chain fatty acids (SCFAs) can regulate the differentiation of effector T cells by acting on molecular receptors on immune cells, thereby regulating the bone immune process. The multifaceted relationship among the gut microbiota, SCFAs, Th17/Treg cell-mediated bone immunity, and bone metabolism is eliciting attention from researchers. Through a review of existing literature, we have comprehensively summarized the effects of the gut microbiota and SCFAs on PMOP, especially from the perspective of Th17/Treg balance. Regulating this balance may provide new opportunities for PMOP treatment.
Collapse
Affiliation(s)
- Yao Chen
- Department of Internal medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Ying Xie
- Department of Internal medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China
| | - Xijie Yu
- Department of Internal medicine, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
18
|
Zhao Z, Xiang L, Hong JS, Wang Y, Feng J. Mechanisms of Acetate in Alleviating SETDB1-Linked Neuroinflammation and Cognitive Impairment in a Mouse Model of OSA. J Inflamm Res 2025; 18:5931-5950. [PMID: 40357375 PMCID: PMC12067661 DOI: 10.2147/jir.s510690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
Background Microglia-mediated neuroinflammation is crucial for obstructive sleep apnea (OSA)-induced cognitive impairment. We aimed to investigate roles of acetate (ACE) and SET domain bifurcated histone lysine methyltransferase 1 (SETDB1) in neuroinflammation of OSA. Methods After C57BL/6J mice were exposed to OSA-associated intermittent hypoxia (IH) or normoxia for four weeks, the composition of the gut microbiota (GM) and the levels of serum short-chain fatty acids (SCFAs) were measured by 16S rRNA and GC-MS methods, respectively. To assess the effect of ACE on IH mice, glyceryl triacetate (GTA) was gavaged in IH-exposed mice and the cognitive function, microglial activation, and hippocampal neuronal death were examined. Moreover, ACE-treated BV2 microglia cells were also utilized for further mechanistic studies. Results IH disrupts the gut microbiome, reduces microbiota-SCFAs, and impairs cognitive function. Gavage with GTA significantly mitigated these cognitive deficits. Following IH exposure, we observed substantial increases in SETDB1 both in vivo and in vitro, along with elevated levels of histone H3 lysine 9 trimethylation (H3K9me3). Genetic or pharmacological inhibition of SETDB1 in microglia led to decreased induction of proinflammatory factors, as well as reduced reactive oxygen species (ROS) generation. Mechanistically, SETDB1 was found to upregulate the transcription factors p-signal transducer and activator of transcription 3 (p-STAT3) and p-NF-κB. In vitro, ACE supplementation effectively repressed high SETDB1 and H3K9me3 levels, thereby inhibiting microglial pro-inflammatory responses induced by IH. In vivo, ACE supplementation significantly reduced hippocampal levels of p-STAT3, p-NF-κB, and pro-inflammatory cytokines while also protecting neuronal integrity. Conclusion This study provides the first evidence that H3K9 methyltransferase SETDB1 promotes microglial pro-inflammatory response distinct from its previously shown role in macrophages. Our findings also identify ACE supplementation as a promising dietary intervention for OSA-related cognitive impairment with SETDB1 serving as both a mechanistic biomarker and potential therapeutic target.
Collapse
Affiliation(s)
- Zhan Zhao
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| | - Li Xiang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA
| | - Yubao Wang
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| | - Jing Feng
- Respiratory Department, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
| |
Collapse
|
19
|
Cuthill S, Muroke V, Dubois A, Dubé MP, Guertin MC, Millette M, Tardif JC. Effect of probiotic supplementation on glycemic control in patients with type 2 diabetes: A randomized controlled trial. Clin Nutr ESPEN 2025; 68:148-152. [PMID: 40345656 DOI: 10.1016/j.clnesp.2025.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
BACKGROUND Type 2 diabetes presents significant public health challenges. The gut microbiome has emerged as a potential factor influencing glucose metabolism. METHODS We performed a randomized, double-blind, single-center trial involving patients with type 2 diabetes and glycated hemoglobin (HbA1c) concentration of 7 % or greater. Patients were randomly assigned to receive 100 billion colony-forming units (CFUs) of probiotic supplementation daily or placebo. The primary efficacy endpoint was the change in HbA1c from baseline to 12 weeks, and secondary endpoints included lipid and inflammatory markers. RESULTS A total of 130 patients were included. HbA1c was 7.63 ± 0.54 % at baseline and 7.63 ± 0.63 % at 12 weeks in the probiotic group and 7.71 ± 0.74 % and 7.81 ± 0.84 % in the placebo group (p = 0.29 between treatment groups). There were also no significant differences between treatment groups in plasma glucose (p = 0.60) and insulin (p = 0.41), as well as in LDL-cholesterol (p = 0.90) and triglycerides (p = 0.32). The adjusted geometric mean percent change (95 % confidence interval) in high-sensitivity C-reactive protein was 1.59 % (-15.71, 22.44) in the probiotic group and -1.37 % (-18.04, 18.70) in the placebo group (p = 0.82). Gastrointestinal adverse events occurred in 38.5 % and 46.2 % of patients in the probiotic group and placebo group respectively (p = 0.48). CONCLUSIONS Probiotic supplementation for 12 weeks did not improve glycemic control, lipid or inflammatory markers in patients with type 2 diabetes. Further research is needed to determine the potential benefits and underlying mechanisms of probiotics in subsets of patients. CLINICALTRIALS gov Identifier no. NCT03239366.
Collapse
Affiliation(s)
- Sabine Cuthill
- Montreal Heart Institute, Montreal, Canada; Queens University, Ontario, Canada
| | - Valtteri Muroke
- Montreal Heart Institute, Montreal, Canada; Université de Montréal, Montreal, Canada
| | | | - Marie-Pierre Dubé
- Montreal Heart Institute, Montreal, Canada; Université de Montréal, Montreal, Canada; Beaulieu-Saucier Pharmacogenomics Centre, Montreal, Canada
| | | | | | - Jean-Claude Tardif
- Montreal Heart Institute, Montreal, Canada; Université de Montréal, Montreal, Canada.
| |
Collapse
|
20
|
Laaraj J, Lachance G, Bergeron A, Fradet Y, Robitaille K, Fradet V. New insights into gut microbiota-prostate cancer crosstalk. Trends Mol Med 2025:S1471-4914(25)00087-5. [PMID: 40374457 DOI: 10.1016/j.molmed.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/24/2025] [Accepted: 03/28/2025] [Indexed: 05/17/2025]
Abstract
Recent evidence underscores a reciprocal relationship between the gut microbiota and prostate cancer (PCa). Dysbiosis, often driven by Western dietary habits and antibiotic use, can heighten systemic inflammation and hinder antitumor immunity, thereby fostering PCa onset and progression. Conversely, certain gut microbes and their metabolites may protect against tumor growth by modulating immune and hormonal pathways that impact therapeutic responses, including androgen deprivation therapy (ADT). Emerging evidence links gut microbial shifts to PCa aggressiveness, potentially sustaining local androgen production and promoting resistance. In this review, we explore current understanding of the gut-PCa interplay, highlighting key knowledge gaps and the need for further research to clarify how targeting the microbiome might influence PCa outcomes.
Collapse
Affiliation(s)
- Jalal Laaraj
- Oncology Research program, CHU de Québec-Université Laval Research center and Cancer Research Center of Université Laval, Québec, QC, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada; Institute of Nutrition and Functional Foods (INAF) and NUTRISS Center - Nutrition, Health and Society of Université Laval, Québec, QC, Canada
| | - Gabriel Lachance
- Oncology Research program, CHU de Québec-Université Laval Research center and Cancer Research Center of Université Laval, Québec, QC, Canada; Institute of Nutrition and Functional Foods (INAF) and NUTRISS Center - Nutrition, Health and Society of Université Laval, Québec, QC, Canada
| | - Alain Bergeron
- Oncology Research program, CHU de Québec-Université Laval Research center and Cancer Research Center of Université Laval, Québec, QC, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Yves Fradet
- Oncology Research program, CHU de Québec-Université Laval Research center and Cancer Research Center of Université Laval, Québec, QC, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Karine Robitaille
- Oncology Research program, CHU de Québec-Université Laval Research center and Cancer Research Center of Université Laval, Québec, QC, Canada; Institute of Nutrition and Functional Foods (INAF) and NUTRISS Center - Nutrition, Health and Society of Université Laval, Québec, QC, Canada
| | - Vincent Fradet
- Oncology Research program, CHU de Québec-Université Laval Research center and Cancer Research Center of Université Laval, Québec, QC, Canada; Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada; Institute of Nutrition and Functional Foods (INAF) and NUTRISS Center - Nutrition, Health and Society of Université Laval, Québec, QC, Canada.
| |
Collapse
|
21
|
Dissayabutra T, Chuaypen N, Somnark P, Boonkaew B, Udomkarnjananun S, Kittiskulnam P, Charoenchittang P, Prombutara P, Tangkijvanich P. Characterization of gut dysbiosis and intestinal barrier dysfunction in patients with metabolic dysfunction-associated steatotic liver disease and chronic kidney disease: a comparative study. Sci Rep 2025; 15:15481. [PMID: 40319096 PMCID: PMC12049563 DOI: 10.1038/s41598-025-00237-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 04/25/2025] [Indexed: 05/07/2025] Open
Abstract
The mechanistic role of gut microbiota in metabolic dysfunction-associated steatotic liver disease (MASLD) and chronic kidney disease (CKD) is increasingly recognized. Despite their close association, comparative data regarding gut dysbiosis in these disorders are limited. This study included 22 healthy controls and 180 patients (90 MASLD, 60 CKD, and 30 both diseases with sex- and age-matched). Fecal bacterial 16 S ribosomal RNA sequencing and butyryl-CoA: acetate CoA transferase (BCoAT) gene expression were analyzed. Plasma intestinal fatty acid binding protein (I-FABP), representing intestinal barrier dysfunction, was assessed using the ELISA method. Our data showed that alpha and beta diversities of gut microbiota differed between MASLD and healthy controls. However, only beta diversities were different between CKD and healthy individuals. The MASLD and CKD groups displayed fewer SCFA-producing genera, particularly Bifidobacterium, than healthy controls. Fecal BCoAT levels were inversely correlated with eGFR and I-FABP levels. Patients with CKD had significantly enriched pathogenic bacteria, reduced BCoAT, and increased I-FABP levels versus MASLD. Combining significant bacterial genera discriminated MASLD from CKD with high diagnostic accuracy (AUC of 0.90). Among patients with both diseases, gut microbial alterations showed mixed characteristics of MASLD and CKD. These data highlighted the shared and distinct gut dysbiosis and related biomarkers, which could provide a better understanding of MASLD and CKD pathogenesis.
Collapse
Affiliation(s)
- Thasinas Dissayabutra
- Metabolic Diseases in Gut and Urinary System Research Unit (MeDGURU), Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Natthaya Chuaypen
- Metabolic Diseases in Gut and Urinary System Research Unit (MeDGURU), Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pornjira Somnark
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Bootsakorn Boonkaew
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Suwasin Udomkarnjananun
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Piyawan Kittiskulnam
- Division of Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Division of Internal Medicine-Nephrology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, Thailand
| | - Pimpisa Charoenchittang
- Department of Computer Science, Faculty of Science, Kasetsart University, Bangkok, Thailand
- Mod Gut Co., Ltd., Bangkok, Thailand
| | - Pinidphon Prombutara
- Mod Gut Co., Ltd., Bangkok, Thailand
- Omics Sciences and Bioinformatics Center, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Pisit Tangkijvanich
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
22
|
Shen Y, Fan N, Ma S, Cheng X, Yang X, Wang G. Gut Microbiota Dysbiosis: Pathogenesis, Diseases, Prevention, and Therapy. MedComm (Beijing) 2025; 6:e70168. [PMID: 40255918 PMCID: PMC12006732 DOI: 10.1002/mco2.70168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 04/22/2025] Open
Abstract
Dysbiosis refers to the disruption of the gut microbiota balance and is the pathological basis of various diseases. The main pathogenic mechanisms include impaired intestinal mucosal barrier function, inflammation activation, immune dysregulation, and metabolic abnormalities. These mechanisms involve dysfunctions in the gut-brain axis, gut-liver axis, and others to cause broader effects. Although the association between diseases caused by dysbiosis has been extensively studied, many questions remain regarding the specific pathogenic mechanisms and treatment strategies. This review begins by examining the causes of gut microbiota dysbiosis and summarizes the potential mechanisms of representative diseases caused by microbiota imbalance. It integrates clinical evidence to explore preventive and therapeutic strategies targeting gut microbiota dysregulation, emphasizing the importance of understanding gut microbiota dysbiosis. Finally, we summarized the development of artificial intelligence (AI) in the gut microbiota research and suggested that it will play a critical role in future studies on gut dysbiosis. The research combining multiomics technologies and AI will further uncover the complex mechanisms of gut microbiota dysbiosis. It will drive the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Yao Shen
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
| | - Nairui Fan
- Basic Medical College of Jiamusi UniversityHeilongjiangChina
| | - Shu‐xia Ma
- Basic Medical College of Jiamusi UniversityHeilongjiangChina
| | - Xin Cheng
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
- International SchoolGuangzhou Huali College, ZengchengGuangzhouChina
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal MedicineDivision of Histology and EmbryologySchool of MedicineJinan UniversityGuangzhouChina
- Key Laboratory for Regenerative Medicine of the Ministry of EducationJinan UniversityGuangzhouChina
- Guangdong‐Hong Kong Metabolism & Reproduction Joint LaboratoryGuangdong Second Provincial General HospitalSchool of MedicineJinan UniversityGuangzhouChina
| |
Collapse
|
23
|
Zhou W, He Y, Lv JM, Wang R, He H, Wu M, Zhang R, He J. Preparation technologies, structural characteristics and biological activities of polysaccharides from bee pollen: A review. Int J Biol Macromol 2025; 306:141545. [PMID: 40020838 DOI: 10.1016/j.ijbiomac.2025.141545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 02/13/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Bee pollen, a natural honeybee product, is hailed as a treasure trove of human nutrition. Among the nourishing substances of bee pollen, the constituents with a low molecular weight (such as phenolic acids and flavonoid glycosides) have been extensively studied in the past decades, whereas the polysaccharides with a relatively high molecular weight have received much less attention. To deepen our understanding of bee pollen polysaccharides, this review summarizes the published findings related to their preparation technologies, structural characteristics and biological activities. Among the preparation technologies, ultrasonic-assisted extraction is currently the most effective technology for the recovery of polysaccharides from bee pollen, because ultrasound can crack the pollen exine into fragments and facilitate the release of polysaccharides present in the pollen intine. The preliminary structures, including the molecular weight and monosaccharide composition, of bee pollen polysaccharides have been widely reported, but their fine structures have not fully elucidated. Moreover, bee pollen polysaccharides have antioxidant, immunomodulatory, and antitumor activities, exhibiting potential application in functional foods. Furthermore, bee pollen polysaccharides can modulate the composition of gut microbiota and promote the production of short-chain fatty acids. It is expected that this review can provide inspiration for the development and utilization of bee pollen polysaccharides.
Collapse
Affiliation(s)
- Wangting Zhou
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China
| | - Yuzhen He
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China
| | - Ji-Min Lv
- The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou 310058, PR China; Xianghu Laboratory, Hangzhou 311231, PR China
| | - Runqi Wang
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China
| | - Huaiye He
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China
| | - Muci Wu
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, PR China
| | - Rui Zhang
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, PR China.
| | - Jingren He
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, PR China.
| |
Collapse
|
24
|
Capatina TF, Oatu A, Babasan C, Trifu S. Translating Molecular Psychiatry: From Biomarkers to Personalized Therapies-A Narrative Review. Int J Mol Sci 2025; 26:4285. [PMID: 40362522 PMCID: PMC12072283 DOI: 10.3390/ijms26094285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/10/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
In this review, we explore the biomarkers of different psychiatric disorders, such as major depressive disorder, generalized anxiety disorder, schizophrenia, and bipolar disorder. Moreover, we show the interplay between genetic and environmental factors. Novel techniques such as genome-wide association studies (GWASs) have identified numerous risk loci and single-nucleotide polymorphisms (SNPs) implicated in these conditions, contributing to a better understanding of their mechanisms. Moreover, the impact of genetic variations on drug metabolisms, particularly through cytochrome P450 (CYP450) enzymes, highlights the importance of pharmacogenomics in optimizing psychiatric treatment. This review also explores the role of neurotransmitter regulation, immune system interactions, and metabolic pathways in psychiatric disorders. As the technology advances, integrating genetic markers into clinical practice will be crucial in advancing precision psychiatry, improving diagnostic accuracy and therapeutic interventions for individual patients.
Collapse
Affiliation(s)
| | - Anamaria Oatu
- Department of Psychiatry, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.O.); (C.B.)
| | - Casandra Babasan
- Department of Psychiatry, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.O.); (C.B.)
| | - Simona Trifu
- Department of Neurosciences, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
25
|
Wang T, Ahmad S, Cruz-Lebrón A, Ernst SE, Olivos Caicedo KY, Jeong Y, Binion B, Mbuvi P, Dutta D, Fernandez-Materan FV, Breister AM, Tang E, Lee JW, Kang JD, Harris SC, Ikegawa S, Gaskins HR, Erdman JW, Yang G, Cann I, Daniel SL, Hylemon PB, Anantharaman K, Bernardi RC, Alves JMP, Sfanos KS, Irudayaraj J, Ridlon JM. An expanded metabolic pathway for androgen production by commensal bacteria. Nat Microbiol 2025; 10:1084-1098. [PMID: 40259019 DOI: 10.1038/s41564-025-01979-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 03/06/2025] [Indexed: 04/23/2025]
Abstract
Commensal bacteria have been implicated in the modulation of steroid hormones, including circulating androgen levels in the host. However, the microbial genetic pathways involved in androgen production have not been fully characterized. Here we identify a microbial gene encoding an enzyme that catalyses the conversion of androstenedione to epitestosterone in the gut microbiome member Clostridium scindens and named this gene desF. We demonstrate that epitestosterone impacts androgen receptor-dependent prostate cancer cell proliferation in vitro. We also demonstrate that stool desF levels are elevated in patients with prostate cancer who are unresponsive to abiraterone/prednisone therapy. Bacterial isolates from urine or prostatectomy tissue produced androgens, and 17β-hydroxysteroid dehydrogenase activity encoded by the desG gene was detected in strains of the urinary tract bacterium Propionimicrobium lymphophilum. Furthermore, we demonstrate that urinary androgen-producing bacterial strains can promote prostate cancer cell growth through metabolism of cortisol and prednisone. Abiraterone, which targets host desmolase (CYP17A1), a rate-limiting enzyme in adrenal steroidogenesis, does not inhibit bacterial desmolase (DesAB), whereas the conversion of prednisone to androgens by DesAB, DesF and DesG drives androgen-receptor-dependent prostate cancer cell line proliferation in vitro. Our results are a significant advance in steroid microbiology and highlight a potentially important role for gut and urinary tract bacteria in host endocrine function and drug metabolism.
Collapse
Affiliation(s)
- Taojun Wang
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
| | - Saeed Ahmad
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Biomedical Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
| | - Angélica Cruz-Lebrón
- Departments of Pathology, Oncology, and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sarah E Ernst
- Departments of Pathology, Oncology, and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Yoon Jeong
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Biomedical Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
| | - Briawna Binion
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
| | - Pauline Mbuvi
- Biomedical Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
- Department of Urology, Carle Foundation Hospital, Urbana, IL, USA
| | - Debapriya Dutta
- Biomedical Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA
- Department of Urology, Carle Foundation Hospital, Urbana, IL, USA
| | - Francelys V Fernandez-Materan
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
| | - Adam M Breister
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth Tang
- Department of Physics, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jae Won Lee
- Department of Biotechnology, Sungshin Women's University, Seoul, South Korea
| | - Jason D Kang
- Stravitz-Sanyal Institute for Liver Disease & Metabolic Health, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | - Spencer C Harris
- Stravitz-Sanyal Institute for Liver Disease & Metabolic Health, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
| | | | - H Rex Gaskins
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - John W Erdman
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Glen Yang
- Department of Urology, Carle Foundation Hospital, Urbana, IL, USA
| | - Isaac Cann
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Steven L Daniel
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Phillip B Hylemon
- Stravitz-Sanyal Institute for Liver Disease & Metabolic Health, Virginia Commonwealth University, School of Medicine, Richmond, VA, USA
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | | | | | - João M P Alves
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Karen S Sfanos
- Departments of Pathology, Oncology, and Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Joseph Irudayaraj
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA.
- Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Biomedical Research Center, Mills Breast Cancer Institute, Carle Foundation Hospital, Urbana, IL, USA.
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Carle-Illinois College of Medicine, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| | - Jason M Ridlon
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, Urbana, IL, USA.
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
- Center for Advanced Study, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
26
|
Ajith TA, Anita B. Impact of Gut Microbiota and Probiotics on Rheumatoid Arthritis: A Potential Treatment Challenge. Int J Rheum Dis 2025; 28:e70266. [PMID: 40329613 DOI: 10.1111/1756-185x.70266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/02/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025]
Abstract
Over the past few decades, there has been a surge in global study on the relationship between gut microbiota and human health. Numerous human illnesses have been linked to dysbiosis. Gram-positive firmicutes and Gram-negative bacteroidetes are the two leading bacterial phyla that make up 90% of the gut microbiome. Many symbionts in the gut environment establish intricate relationships with host defense to stop both local and non-native dangerous bacteria from colonizing and invading. Dysbiosis alters the paracellular route and damages the epithelium, enabling them to penetrate the epithelium and come into contact with the immune cells. Impaired intestinal barrier function, immune regulation mediated by metabolites derived from the gut microbiota, posttranslational modification of host proteins such as increased citrullination, regulation of the gut microbiota's effect on immune cells, intestinal epithelial cell autophagy, interaction between the microbiome and human leukocyte antigen alleles, and interaction with microRNAs are some of the mechanisms involved in rheumatoid arthritis (RA). The gut microbiota, Prevotella copri, and Collinsella spp. were shown to be higher in the early/preclinical phases of RA, while Bacteroidetes, Bifidobacteria, and Eubacterium rectale were found to be lower. Probiotic-based early dietary intervention may reduce inflammation and slow the rate of joint deterioration, and such intervention can also aid in the restoration of gut microbiota equilibrium. This review article describes the gut microbial dysbiosis and role of probiotics in RA.
Collapse
Affiliation(s)
| | - Bejoy Anita
- Department of General Medicine, Amala Institute of Medical Sciences, Thrissur, Kerala, India
| |
Collapse
|
27
|
Metris A, Walker AW, Showering A, Doolan A, McBain AJ, Ampatzoglou A, Murphy B, O'Neill C, Shortt C, Darby EM, Aldis G, Hillebrand GG, Brown HL, Browne HP, Tiesman JP, Leng J, Lahti L, Jakubovics NS, Hasselwander O, Finn RD, Klamert S, Korcsmaros T, Hall LJ. Assessing the safety of microbiome perturbations. Microb Genom 2025; 11. [PMID: 40371892 DOI: 10.1099/mgen.0.001405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025] Open
Abstract
Everyday actions such as eating, tooth brushing or applying cosmetics inherently modulate our microbiome. Advances in sequencing technologies now facilitate detailed microbial profiling, driving intentional microbiome-targeted product development. Inspired by an academic-industry workshop held in January 2024, this review explores the oral, skin and gut microbiomes, focussing on the potential long-term implications of perturbations. Key challenges in microbiome safety assessment include confounding factors (ecological variability, host influences and external conditions like geography and diet) and biases from experimental measurements and bioinformatics analyses. The taxonomic composition of the microbiome has been associated with both health and disease, and perturbations like regular disruption of the dental biofilm are essential for preventing caries and inflammatory gum disease. However, further research is required to understand the potential long-term impacts of microbiome disturbances, particularly in vulnerable populations including infants. We propose that emerging technologies, such as omics technologies to characterize microbiome functions rather than taxa, leveraging artificial intelligence to interpret clinical study data and in vitro models to characterize and measure host-microbiome interaction endpoints, could all enhance the risk assessments. The workshop emphasized the importance of detailed documentation, transparency and openness in computational models to reduce uncertainties. Harmonisation of methods could help bridge regulatory gaps and streamline safety assessments but should remain flexible enough to allow innovation and technological advancements. Continued scientific collaboration and public engagement are critical for long-term microbiome monitoring, which is essential to advancing safety assessments of microbiome perturbations.
Collapse
Affiliation(s)
- Aline Metris
- Unilever, Safety, Environmental and Regulatory Sciences (SERS), Sharnbrook, UK
| | - Alan W Walker
- Microbiome, Food Innovation and Food Security Theme, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | | | | | - Andrew J McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Antonis Ampatzoglou
- Unilever, Safety, Environmental and Regulatory Sciences (SERS), Sharnbrook, UK
| | - Barry Murphy
- Unilever R&D Port Sunlight, Bebington, Wirral, UK
| | - Catherine O'Neill
- Division of Dermatology and Musculoskeletal Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | | | - Elizabeth M Darby
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | | | - Greg G Hillebrand
- University of Cincinnati, James L. Winkle College of Pharmacy, Cincinnati, OH, USA
| | - Helen L Brown
- School of Biosciences, Sir Martin Evans Building, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Hilary P Browne
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College, Cork, Ireland
| | | | - Joy Leng
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Leo Lahti
- Department of Computing, University of Turku, Turku FI-20014, Finland
| | - Nicholas S Jakubovics
- School of Dental Sciences, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | | | - Robert D Finn
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Silvia Klamert
- Unilever, Safety, Environmental and Regulatory Sciences (SERS), Sharnbrook, UK
| | - Tamas Korcsmaros
- Food, Microbiomes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Division of Digestive Diseases, Imperial College London, London, UK
- NIHR Imperial BRC Organoid Facility, Imperial College London, London, UK
| | - Lindsay J Hall
- Food, Microbiomes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Department of Microbes, Infection and Microbiomes, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, UK
| |
Collapse
|
28
|
Wang N, Wu M, Gu W, Dai C, Shao Z, Subbalakshmi KP. MSFT-transformer: a multistage fusion tabular transformer for disease prediction using metagenomic data. Brief Bioinform 2025; 26:bbaf217. [PMID: 40370098 PMCID: PMC12078939 DOI: 10.1093/bib/bbaf217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/05/2025] [Accepted: 04/21/2025] [Indexed: 05/16/2025] Open
Abstract
More and more recent studies highlight the crucial role of the human microbiome in maintaining health, while modern advancements in metagenomic sequencing technologies have been accumulating data that are associated with human diseases. Although metagenomic data offer rich, multifaceted information, including taxonomic and functional abundance profiles, their full potential remains underutilized, as most approaches rely only on one type of information to discover and understand their related correlations with respect to disease occurrences. To address this limitation, we propose a multistage fusion tabular transformer architecture (MSFT-Transformer), aiming to effectively integrate various types of high-dimensional tabular information extracted from metagenomic data. Its multistage fusion strategy consists of three modules: a fusion-aware feature extraction module in the early stage to improve the extracted information from inputs, an alignment-enhanced fusion module in the mid stage to enforce the retainment of desired information in cross-modal learning, and an integrated feature decision layer in the late stage to incorporate desired cross-modal information. We conduct extensive experiments to evaluate the performance of MSFT-Transformer over state-of-the-art models on five standard datasets. Our results indicate that MSFT-Transformer provides stable performance gains with reduced computational costs. An ablation study illustrates the contributions of all three models compared with a reference multistage fusion transformer without these novel strategies. The result analysis implies the significant potential of the proposed model in future disease prediction with metagenomic data.
Collapse
Affiliation(s)
- Ning Wang
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214121, Jiangsu, China
| | - Minghui Wu
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214121, Jiangsu, China
| | - Wenchao Gu
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214121, Jiangsu, China
| | - Chenglong Dai
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214121, Jiangsu, China
| | | | - K P Subbalakshmi
- Department of Electrical and Computer Engineering, Stevens Institute of Technology, Castle Point Terrace, Hoboken, NJ 07030, United States
| |
Collapse
|
29
|
Husain N, Kumar A, Anbazhagan AN, Gill RK, Dudeja PK. Intestinal luminal anion transporters and their interplay with gut microbiome and inflammation. Am J Physiol Cell Physiol 2025; 328:C1455-C1472. [PMID: 40047092 PMCID: PMC12023768 DOI: 10.1152/ajpcell.00026.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 01/29/2025] [Accepted: 02/28/2025] [Indexed: 04/16/2025]
Abstract
The intestine, as a critical interface between the external environment and the internal body, plays a central role in nutrient absorption, immune regulation, and maintaining homeostasis. The intestinal epithelium, composed of specialized epithelial cells, harbors apical anion transporters that primarily mediate the transport of chloride and bicarbonate ions, essential for maintaining electrolyte balance, pH homeostasis, and fluid absorption/secretion. In addition, the intestine hosts a diverse population of gut microbiota that plays a pivotal role in various physiological processes including nutrient metabolism, immune regulation, and maintenance of intestinal barrier integrity, all of which are critical for host gut homeostasis and health. The anion transporters and gut microbiome are intricately interconnected, where alterations in one can trigger changes in the other, leading to compromised barrier integrity and increasing susceptibility to pathophysiological states including gut inflammation. This review focuses on the interplay of key apical anion transporters including Down-Regulated in Adenoma (DRA, SLC26A3), Putative Anion Transporter-1 (PAT1, SLC26A6), and Cystic Fibrosis Transmembrane Conductance Regulator [CFTR, ATP-binding cassette subfamily C member 7 (ABCC7)] with the gut microbiome, barrier integrity, and their relationship to gut inflammation.
Collapse
Affiliation(s)
- Nazim Husain
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
| | - Anoop Kumar
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Arivarasu N. Anbazhagan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
| | - Ravinder K Gill
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Pradeep. K. Dudeja
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, IL, USA
- Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
30
|
Liang J, Ren Y, Zheng Y, Lin X, Song W, Zhu J, Zhang X, Zhou H, Wu Q, He Y, Yin J. Functional Outcome Prediction of Acute Ischemic Stroke Based on the Oral and Gut Microbiota. Mol Neurobiol 2025; 62:5413-5431. [PMID: 39546118 PMCID: PMC11953115 DOI: 10.1007/s12035-024-04618-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Although several studies have identified a distinct gut microbiota in individuals with acute ischemic stroke (AIS), there is a limited amount of research that has simultaneously investigated alterations in the oral and intestinal microbiota in AIS patients and their correlation with clinical prognosis. This was a prospective and observational single-center cohort study in which we included 160 AIS patients who were admitted within 24 h after a stroke event. We collected oral and rectal swab samples for analysis using 16S rRNA high-throughput sequencing. Our study revealed that patients with unfavorable outcomes after AIS showed early disruptions in their oral and intestinal microbiota. Rectal swabs showed increased levels of facultatively anaerobic bacteria in patients with a poor prognosis, while the oral cavity exhibited higher levels of anaerobic and opportunistic pathogenic bacteria. By employing machine learning analysis, we found that the microbiota composition at both rectal and oral sites could predict early and long-term outcomes. Moreover, patients with a poor prognosis displayed increased oral bacterial colonization in the rectal microbiota and altered interactions between the oral and gut microbiota. This study reveals distinct rectal and oral bacteria that could predict unfavorable outcomes for AIS patients. Monitoring the microbiota of various body sites during the early stages after admission may hold prognostic value and inform personalized treatment strategies. The presence of oral bacteria colonizing the intestines during the acute phase of stroke could serve as an early indication of poor outcomes for AIS patients.
Collapse
Affiliation(s)
- Jingru Liang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yueran Ren
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yifeng Zheng
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaofei Lin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Song
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiajia Zhu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaomei Zhang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Guangdong Provincial Clinical Research Center for Laboratory Medicine, Guangzhou, Guangdong, China
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong, China
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou, Guangdong, China
| | - Qiheng Wu
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| | - Yan He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
- Guangdong Provincial Clinical Research Center for Laboratory Medicine, Guangzhou, Guangdong, China.
- State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong, China.
- Key Laboratory of Mental Health of the Ministry of Education, Guangzhou, Guangdong, China.
| | - Jia Yin
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
31
|
Zou M, Li X, Li C, Pei H, Kang R, Liu L, Gao L. Comparative Analysis of Gut Bacteria of Four Waterbirds Species in Taolimiao-Alashan Nur (T-A Nur) in Erdos Relic Gull National Nature Reserve, Inner Mongolia, China. Ecol Evol 2025; 15:e71432. [PMID: 40370353 PMCID: PMC12074897 DOI: 10.1002/ece3.71432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/15/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
Taolimiao-Alashan Nur (T-A Nur) is an important breeding site for the Relict Gulls (Larus relictus) and many other waterbirds. To understand the gut health status of rare bird species living there and to protect these bird species, this study analyzed the gut microbiota of four waterbird species, including Relict Gull (L. relictus), Black-necked Grebe (Podiceps nigricollis), Greylag Goose (Anser anser), and Ruddy Shelduck (Tadorna ferruginea), using 16S rRNA high-throughput sequencing. Results showed that the gut microbiota of Ruddy Shelduck had the highest α-diversity, while Greylag Goose had the lowest. The composition of gut microbiota varied significantly among the bird species. The dominant bacterial phylum in the guts of Black-necked Grebe, Greylag Goose, and Ruddy Shelduck was Firmicutes, while it was Pseudomonadota in Relict Gull. At the genus level, the dominant bacteria were Halomonas in Black-necked Grebe, Escherichia-Shigella in Relict Gull, Ligilactobacillus in Greylag Goose, and Enterococcus in Ruddy Shelduck. Correlation analysis revealed significant relationships among gut bacterial communities, suggesting that gut bacteria can regulate host metabolism and physiological state by their interactions. KEGG functional predictions indicated that gut microbiota were primarily involved in metabolism. The abundance of metabolism-related microorganisms in Relict Gull was significantly lower than in Greylag Goose and Ruddy Shelduck, indicating that the gut microbiota of Greylag Goose and Ruddy Shelduck can provide stronger metabolic functions for the hosts. Additionally, microorganisms related to human diseases were more abundant in the gut of Relict Gull compared to Ruddy Shelduck and Black-necked Grebe, and in Greylag Goose compared to Ruddy Shelduck. These findings suggested that the gut microbiota of birds in this area harbor some human pathogens, which warrants attention and preventive measures.
Collapse
Affiliation(s)
- Mingxin Zou
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| | - Xuanyu Li
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| | - Chunyu Li
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| | - Hongda Pei
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| | - Ruobing Kang
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| | - Li Liu
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| | - Li Gao
- College of Ecology and EnvironmentBaotou Teacher's CollegeBaotouChina
| |
Collapse
|
32
|
Piłot M, Dzięgielewska-Gęsiak S, Walkiewicz KW, Bednarczyk M, Waniczek D, Muc-Wierzgoń M. Gut Microbiota and Metabolic Dysregulation in Elderly Diabetic Patients: Is There a Gender-Specific Effect. J Clin Med 2025; 14:3103. [PMID: 40364140 PMCID: PMC12073094 DOI: 10.3390/jcm14093103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/14/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Background/Objectives: The aim of this study was to qualitatively and quantitatively assess the bacterial domain of the gut microbiome in elderly patients with type 2 diabetes (T2D), with a focus on sex differences, glycemic control, and lipid disorders. Methods: This study included 60 older adults with T2D (38 women and 22 men) treated with metformin or a combination of metformin and insulin. The gut microbiota was profiled using 16S rRNA gene sequencing. Statistical analyses, including correlation analysis and multiple regression, were performed to identify the associations between microbial taxa, sex, and metabolic parameters. Results: No statistically significant differences in alpha or beta diversity were observed between the sexes. Multiple regression analysis indicated a positive relationship between Tenericutes and HbA1c in male participants (β = 2.22931, CI [0.75, 3.70], R = 0.67; R2 = 0.36; unadjusted p = 0.0052; adjusted p = 0.0496). In female participants, G0' (β = -2.24107, CI [-3.19, -1.30], R = 0.78; R2 = 0.58; unadjusted p = 0.00003; adjusted p = 0.0005) and HbA1c (β = -1.86670, CI [-2.61, -1.12], R = 0.78; R2 = 0.58; unadjusted p = 0.00001; adjusted p = 0.0003) correlated negatively with Verrucomicrobia as well G0' (β = -1.90427, CI [-2.95, -0.85], R = 0.46; R2 = 0.17; unadjusted p = 0.0008; adjusted p = 0.007) and HbA1c (β = -1.69561, CI [-2.52, -0.87], R = 0.46; R2 = 0.17; unadjusted p = 0.0002; adjusted p = 0.002) correlated negatively with OD1 bacteria, known as Parcubacteria. Conclusions: In this elderly population with type 2 diabetes, biological sex did not significantly affect the gut microbiota diversity. However, several exploratory associations between microbial taxa and metabolic parameters differed between men and women, suggesting that sex may influence specific aspects of microbiota-metabolism interactions. These preliminary findings underscore the importance of considering both age- and sex-related factors when investigating the gut microbiome in the context of type 2 diabetes.
Collapse
Affiliation(s)
- Magdalena Piłot
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Piekarska 18, 44-902 Bytom, Poland; (M.P.); (S.D.-G.); (K.W.W.)
| | - Sylwia Dzięgielewska-Gęsiak
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Piekarska 18, 44-902 Bytom, Poland; (M.P.); (S.D.-G.); (K.W.W.)
| | - Katarzyna Weronika Walkiewicz
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Piekarska 18, 44-902 Bytom, Poland; (M.P.); (S.D.-G.); (K.W.W.)
| | - Martyna Bednarczyk
- Department of Cancer Prevention, Faculty of Public Health, Medical University of Silesia in Katowice, 40-752 Katowice, Poland;
| | - Dariusz Waniczek
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-808 Katowice, Poland;
| | - Małgorzata Muc-Wierzgoń
- Department of Internal Diseases Propaedeutics and Emergency Medicine, Faculty of Public Health in Bytom, Medical University of Silesia in Katowice, Piekarska 18, 44-902 Bytom, Poland; (M.P.); (S.D.-G.); (K.W.W.)
| |
Collapse
|
33
|
Sessa L, Malavolta E, Sodero G, Cipolla C, Rigante D. The conspiring role of gut microbiota as primer of autoimmune thyroid diseases: A scoping focus. Autoimmun Rev 2025; 24:103780. [PMID: 39971108 DOI: 10.1016/j.autrev.2025.103780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/31/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
The thyroid gland is the body's largest single organ specialized for endocrine hormone production, and still unraveled mechanisms regulate its interaction between the hypothalamic-pituitary-thyroid axis and composition of the gut microbiota: in particular, a disrupted integrity of the intestinal barrier, causing dysbiosis and increasing detrimental substances or reducing beneficial metabolites, such as short-chain fatty acids (SCFAs) with proinflammatory effects, may be crucial for the induction of an autoimmune thyroid disease. More specifically, Lactobacilli and Bifidobacteria have a role in this partnership through a "molecular mimicry" mechanism, as their protein sequences share structural similarity with thyroid peroxidase and thyroglobulin. Lactobacilli can also increase T helper 17 cells, modifying the number of colonic regulatory T cells, largely implicated in the maintenance of immunological tolerance at the gut barrier. Additionally, Blautia and Anaerostipes work beneficially with butyric acid, one of the SCFAs, promoting antimicrobial peptide synthesis from the intestinal cells and bolstering the innate immune system's ability to struggle against pathogens, which can also influence thyroid hormone levels by regulating iodine uptake and metabolism. This review aims to summarize the current knowledge about the contribution of gut microbiota changes in triggering immune abnormalities leading to autoimmune thyroid diseases.
Collapse
Affiliation(s)
- Linda Sessa
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elena Malavolta
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giorgio Sodero
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Clelia Cipolla
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Donato Rigante
- Department of Life Sciences and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Università Cattolica Sacro Cuore, Rome, Italy.
| |
Collapse
|
34
|
Cui X, Li C, Zhong J, Liu Y, Xiao P, Liu C, Zhao M, Yang W. Gut microbiota - bidirectional modulator: role in inflammatory bowel disease and colorectal cancer. Front Immunol 2025; 16:1523584. [PMID: 40370465 PMCID: PMC12075242 DOI: 10.3389/fimmu.2025.1523584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
The gut microbiota is a diverse ecosystem that significantly impacts human health and disease. This article focuses on how the gut microbiota interacts with inflammatory bowel diseases and colorectal tumors, especially through immune regulation. The gut microbiota plays a role in immune system development and regulation, while the body's immune status can also affect the composition of the microbiota. These microorganisms exert pathogenic effects or correct disease states in gastrointestinal diseases through the actions of toxins and secretions, inhibition of immune responses, DNA damage, regulation of gene expression, and protein synthesis. The microbiota and its metabolites are essential in the development and progression of inflammatory bowel diseases and colorectal tumors. The complexity and bidirectionality of this connection with tumors and inflammation might render it a new therapeutic target. Hence, we explore therapeutic strategies for the gut microbiota, highlighting the potential of probiotics and fecal microbiota transplantation to restore or adjust the microbial community. Additionally, we address the challenges and future research directions in this area concerning inflammatory bowel diseases and colorectal tumors.
Collapse
Affiliation(s)
- Xilun Cui
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Changfeng Li
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jing Zhong
- Department of Medical Imaging, The Third Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, China
| | - Yuanda Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Pengtuo Xiao
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chang Liu
- Department of Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Mengwei Zhao
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Wei Yang
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
35
|
Yao C, Zhang Y, You L, E J, Wang J. Comparative analysis of three experimental methods for revealing human fecal microbial diversity. BMC Microbiol 2025; 25:258. [PMID: 40301726 PMCID: PMC12039119 DOI: 10.1186/s12866-025-03985-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 04/22/2025] [Indexed: 05/01/2025] Open
Abstract
Due to the heterogeneity of the human gut environment, the gut microbiota is complex and diverse, and has been insufficiently explored. In this study, one fresh fecal sample was cultured using 12 commercial or modified media and incubation of culture plates anaerobically and aerobically, the conventional experienced colony picking (ECP) was first used to isolate the colonies and obtain pure culture strains. On this basis, all the colonies grown on the culture plates were collected for culture-enriched metagenomic sequencing (CEMS), and the original sample was also subjected to direct culture-independent metagenomic sequencing (CIMS), the study compared the effects of three methods for analyzing the microbiota contained in the sample. It was found that compared with CEMS, conventional ECP failed to detect a large proportion of strains grown in culture media, resulting in missed detection of culturable microorganisms in the gut. Microbes identified by CEMS and CIMS showed a low degree of overlap (18% of species), whereas species identified by CEMS and CIMS alone accounted for 36.5% and 45.5%, respectively. It suggests that both culture-dependent and culture-independent approaches are essential in revealing gut microbial diversity. Moreover, based on the CEMS results, growth rate index (GRiD) values for various strains on different media were calculated to predict the optimal medium for bacterial growth; this method can be used to design new media for intestinal microbial isolation, promote the recovery of specific microbiota, and obtain new insights into the human microbiome diversity. This is among the first studies on CEMS of the human gut microbiota.
Collapse
Affiliation(s)
- Caiqing Yao
- College of Food Science, Shanxi Normal University, Taiyuan, 030031, China
| | - Yu Zhang
- College of Food Science, Shanxi Normal University, Taiyuan, 030031, China
| | - Lijun You
- School of Food Science and Engineering, Bohai University, Liaoning, 121013, China
| | - Jingjing E
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, School of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Junguo Wang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, School of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| |
Collapse
|
36
|
He AQ, Xiao WY, Zheng T, Li KY, Li BS, Wang S, Yu QX, Liu G. Efficacy of curcumin supplementation for the treatment and prevention of pouchitis after ileal pouch-anal anastomosis: a randomized controlled trial. Eur J Nutr 2025; 64:167. [PMID: 40295333 DOI: 10.1007/s00394-025-03676-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 04/02/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Pouchitis is the most common complication after ileal pouch-anal anastomosis (IPAA) for ulcerative colitis. Induction and maintenance of remission is a crucial therapeutic goal. We investigated curcumin's efficacy in treatment of pouchitis. METHODS The double-blind trial included an induction cohort of refractory pouchitis patients and a maintenance cohort of patients without pouchitis after IPAA. Patients received either placebo or curcumin for 8 weeks. The pouchitis activity were assessed before and after and was compared between cohorts or groups. Laboratory inflammation indicators, nutritional status and quality of life were also appraised. RESULTS 52 patients were included, with 39 and 13 patients entering the maintenance cohort and induction cohort, respectively. In maintenance cohort, the proportion of clinical remission elevated from 11 to 89% in curcumin group (p = 0.005), whereas there was no significant difference in placebo group (10% vs 5%, p = 1).In induction cohort, 67% (4/6) patients achieved clinical response after 8 weeks' intervention of curcumin, whereas none treated with placebo (p = 0.021). Patients treated with curcumin appeared less inflammation and there was no significant difference in indicators changes between two cohorts. CONCLUSIONS Curcumin has preventive and therapeutic effects on pouchitis. Curcumin supplementation can reduce the disease activity and improve the nutritional status of patients with after IPAA. TRIAL REGISTRATION ChiCTR, ChiCTR1900022243. Registered 31 March 2019, https://www.chictr.org.cn/historyversionpub.aspx?regno=ChiCTR1900022243.
Collapse
Affiliation(s)
- An-Qi He
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China
| | - Wan-Yi Xiao
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China
| | - Ting Zheng
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China
| | - Kai-Yu Li
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China
| | - Bao-Song Li
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China
| | - Song Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China
| | - Qing-Xiang Yu
- Department of Gastroenterology, Tianjin Medical University General Hospital, Tianjin, 300052, People's Republic of China.
| | - Gang Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Anshan Road NO.154, Heping District, Tianjin, People's Republic of China.
| |
Collapse
|
37
|
Romani FE, Luvira V, Chancharoenthana W, Albanese M, Maddaloni L, Branda F, D'Amelio S, Gabrielli S, Scagnolari C, Mastroianni CM, Ceccarelli G, d'Ettorre G. Human microbiota in dengue infection: A narrative review. Microb Pathog 2025; 205:107643. [PMID: 40306589 DOI: 10.1016/j.micpath.2025.107643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/02/2025]
Abstract
Dengue fever, a widespread mosquito-borne viral infection in tropical regions, typically manifests fever and gastrointestinal symptoms, including nausea, vomiting, and diarrhea. However, the human gut microbiota's role in dengue pathogenesis remains incompletely understood. Studies have demonstrated dysbiosis during dengue virus infection, characterized by increased abundance of potentially pathogenic bacteria like Bacteroidaceae and Proteobacteria, particularly during the critical phase. Furthermore, microbial translocation and leaky gut syndrome, characterized by the translocation of intestinal microbial products, have been observed in dengue patients and are associated with hypercytokinemia, plasma leakage, and disease severity. These findings underscore the necessity for an in-depth investigation into the role of human intestinal microbiota as a potential contributing factor in the pathogenesis and progression of dengue. Further research focusing on human intestinal microbiota, leaky gut syndrome, and the potential implications of treatment with oral bacteriotherapy, as previously observed in other viral diseases, is essential to clarify dengue pathology and evaluate new therapeutic strategies.
Collapse
Affiliation(s)
- Francesco Eugenio Romani
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy; Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Viravarn Luvira
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Wiwat Chancharoenthana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Mattia Albanese
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy; Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Luca Maddaloni
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy
| | - Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, 00128, Rome, Italy
| | - Stefano D'Amelio
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy
| | - Simona Gabrielli
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy
| | - Carolina Scagnolari
- Department of Molecular Medicine, Laboratory of Virology, University of Rome Sapienza, Rome, Italy
| | | | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy; Azienda Ospedaliero Universitaria Policlinico Umberto I, Rome, Italy; Migrant and Global Health Research Organization (Mi-HeRO), Rome, Italy.
| | - Gabriella d'Ettorre
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy
| |
Collapse
|
38
|
Lee SH, Han C, Shin C. IUPHAR Review: Microbiota-Gut-Brain Axis and its role in Neuropsychiatric Disorders. Pharmacol Res 2025; 216:107749. [PMID: 40306604 DOI: 10.1016/j.phrs.2025.107749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 04/20/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
The human gut microbiome, composed of a vast array of microorganisms that have co-evolved with humans, is crucial for the development and function of brain systems. Research has consistently shown bidirectional communication between the gut and the brain through neuronal, endocrine, and immunological, and chemical pathways. Recent neuroscience studies have linked changes in the microbiome and microbial metabolites to various neuropsychiatric disorders such as autism, depression, anxiety, schizophrenia, eating disorders, and neurocognitive disorders. Novel metagenome-wide association studies have confirmed these microbiome variations in large samples and expanded our understanding of the interactions between human genes and the gut microbiome. The causal relationship between gut microbiota and neuropsychiatric disorders is being elucidated through the establishment of large cohort studies incorporating microbiome data and advanced statistical techniques. Ongoing animal and human studies focused on the microbiota-gut-brain axis are promising for developing new prevention and treatment strategies for neuropsychiatric conditions. The scope of these studies has broadened from microbiome-modulating therapies including prebiotics, probiotics, synbiotics and postbiotics to more extensive approaches such as fecal microbiota transplantation. Recent systematic reviews and meta-analyses have strengthened the evidence base for these innovative treatments. Despite extensive research over the past decade, many intriguing aspects still need to be elucidated regarding the role and therapeutic interventions of the microbiota-gut-brain axis in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Seung-Hoon Lee
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Changsu Han
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Cheolmin Shin
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
39
|
Lu J, Tao K. Urinary microbiota in patients with kidney stones: a systematic review and meta-analysis. Microb Pathog 2025; 205:107641. [PMID: 40306588 DOI: 10.1016/j.micpath.2025.107641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 03/14/2025] [Accepted: 04/26/2025] [Indexed: 05/02/2025]
Abstract
PURPOSE The global prevalence of kidney stones (KS) has been on the rise in recent years. Numerous studies have suggested an association between urine microbes and KS, although the precise underlying mechanisms remain unclear. This meta-analysis and systematic review aim to investigate the urine microbiota composition in patients with KS and delineate the disparities in urine microbiota between healthy individuals and KS patients. METHODS A systematic search was conducted on PubMed, Embase, and Web of Science databases to identify relevant studies. Seven studies were selected for analysis to compare the urinary microbiota profiles of KS patients with controls. This research adhered to the PRISMA guidelines, with data extraction performed independently by two researchers. The study outcomes focused on assessing differences in the α-diversity index of urinary microbiota between KS patients and healthy controls, as well as discrepancies in microbiota abundance at the phylum, genus, and species levels. Statistical analyses were carried out using Review Manager 5.4.1 and Stata 17.0 software. The I2 statistic was utilized to evaluate result heterogeneity, and a sensitivity analysis was conducted to explore potential sources of heterogeneity. Publication bias was assessed through funnel plots and Egger's tests. This study is registered with PROSPERO (No. CRD42024506599). RESULTS We have searched PubMed, Embase and Web of Science and analyzed statistics by using Review Manager 5.4.1 and Stata 17.0 software. Our research has included seven articles with a total of 162 kidney stone patients and 139 healthy controls. Based on our results, compared to healthy controls, the α-diversity of KS patients including Shannon index is significantly lower (SMD -0.55, 95 % CI, -1.10∼ -0.01). However, the relative abundances of Acinetobacter, Pseudomonadota and Corynebacterium of patients are higher than healthy controls, while the abundances of Bacilliota and Bacteroides are lower. CONCLUSION The meta-analysis and systematic review has indicated that the urinary microbiota is not only different between patients with KS and healthy people, but also relates to the pathogenesis of kidney stones. The results suggests that diet modification or appropriate use of antibiotics may effectively prevent the development of KS.
Collapse
Affiliation(s)
- Junyu Lu
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Kai Tao
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
40
|
Zhou Y, Zhu H, Zhao L, Zhao G, Sun J. Bidirectional Mendelian randomization and potential mechanistic insights into the causal relationship between gut microbiota and malignant mesothelioma. Medicine (Baltimore) 2025; 104:e42245. [PMID: 40295238 PMCID: PMC12040020 DOI: 10.1097/md.0000000000042245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Malignant mesothelioma (MM) is a rare but aggressive cancer originating from mesothelial cells, which presents significant challenges to patients' physical and psychological well-being. The gut-lung axis underscores the connection between gut microbiota and respiratory diseases, with emerging evidence suggesting a strong association between gut microbiota and the development of MM. In this study, we conducted a two-sample Mendelian randomization (MR) analysis to investigate the potential causal relationship between gut microbiota and MM, while also exploring the underlying mechanisms through bioinformatics approaches. Gut microbiota summary data were obtained from the MiBioGen consortium, while MM data were sourced from the FinnGen R11 dataset. Causality was examined using the inverse variance weighted method as the primary analysis. Additional methods, including the weighted median, simple mode, MR-Egger, and weighted mode, were also employed. The robustness of the findings was validated through sensitivity analyses, and reverse causality was considered to further strengthen the MR results. Moreover, bioinformatics analyses were conducted on genetic loci associated with both gut microbiota and MM to explore potential underlying mechanisms. Our study suggests that genetically predicted increases in class.Bacilli, family.Rikenellaceae, genus.Clostridium innocuum group, and order.Lactobacillales were suggestively associated with a higher risk of MM, whereas increases in genus.Ruminococcaceae UCG004, genus.Flavonifractor, phylum.Firmicutes, genus.Anaerofilum, genus.Clostridium sensu stricto 1, and genus.Lactobacillus appeared to confer protective effects. Bioinformatics analysis indicated that differentially expressed genes near loci associated with gut microbiota might affect MM by modulating pathways and the tumor microenvironment. The results of this study point to a potential genetic predisposition linking gut microbiota to MM. Further experimental validation is crucial to confirm these candidate microbes, establish causality, and elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Yinjie Zhou
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Huangkai Zhu
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Long Zhao
- Department of Cardiovascular Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Guofang Zhao
- Department of Thoracic Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| | - Jiaen Sun
- Department of Cardiovascular Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
| |
Collapse
|
41
|
Li S, Zhang Y, Liu C, Li X. Where Do Milk Microbes Originate? Traceability of Microbial Community Structure in Raw Milk. Foods 2025; 14:1490. [PMID: 40361573 PMCID: PMC12072076 DOI: 10.3390/foods14091490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Variations in ecological environments (including milk collection equipment and milk storage tanks in the pasture) and seasonal changes may contribute to raw milk contamination, thereby affecting food safety. The composition, structure, and relationships between raw milk and microbial communities in these environments are not well understood. In this study, 84 samples from spring and autumn in Luxian County, Yunnan Province, China, were collected for high-throughput sequencing technology. The results showed that the skin on the nipple surface and the environment (including the wiping samples of the automatic milking machine and the inner cover of the milk tank) had the greatest impact on microbial community composition in raw milk, followed by dung. In addition, microbial diversity in autumn samples was significantly higher, likely due to seasonal factors, including increased rainfall and reduced ultraviolet radiation. By analyzing the microbial community of raw milk and its environmental source, this study traced the origin of microorganisms in milk, providing insights for further exploration of the interaction between the pasture environment and raw milk microorganisms.
Collapse
Affiliation(s)
| | | | - Chenjian Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (S.L.); (Y.Z.)
| | - Xiaoran Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (S.L.); (Y.Z.)
| |
Collapse
|
42
|
Verspohl V, van Egmond M, Kneisel L, Reese F, Thelen AC, Korten N, Neumann M, Schaack L, Voelz C, Käver L, Herpertz-Dahlmann B, Beyer C, Seitz J, Trinh S. Chronic starvation induces microglial cell depletion in an activity-based anorexia model. Sci Rep 2025; 15:14132. [PMID: 40269196 PMCID: PMC12019532 DOI: 10.1038/s41598-025-98237-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/10/2025] [Indexed: 04/25/2025] Open
Abstract
Anorexia nervosa (AN) is a severe psychiatric disease with a largely unknown pathophysiology. AN leads to reduced brain volume and a disbalance of the gut microbiome suggesting the involvement of the gut-brain-axis. Also, in the activity-based anorexia (ABA) animal model mimicking AN brain volume loss is observed. This study investigated the impact of chronic starvation on brain cell populations and evaluated the potential protective effects of omega-3 fatty acids (FA) and probiotics in rats. We used a chronic ABA model and provided daily oral supplementation of omega-3 FA and probiotics. Immunohistochemistry and qPCR were used to analyze GFAP-positive astrocytes, IBA1-positive microglia, OLIG1/2-positive oligodendrocytes, MAP2-positive neurons and Ki-67-positive proliferating cells in the cerebral cortex and corpus callosum. We found a significant reduction of astrocytes and microglia in all ABA groups, likely due to reduced proliferating cells. Reduced running wheel activity and reduced amount of food needed to sustain body weight were observed in animals with supplementation with omega-3 FA and probiotics but we did not observe alterations in brain cells that could be attributed to these supplementations. Our results indicate that glial cell depletion potentially underlies the diminished brain volume found in ABA rats. Omega-3 FA and probiotics show potential for reducing AN-related symptoms and merit further study as a therapeutic approach.
Collapse
Affiliation(s)
- Valerie Verspohl
- Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Miranda van Egmond
- Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Lilly Kneisel
- Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Friederike Reese
- Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Anna C Thelen
- Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Nele Korten
- Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Maren Neumann
- Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Lena Schaack
- Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Clara Voelz
- Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Larissa Käver
- Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Beate Herpertz-Dahlmann
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, Neuenhofer Weg 21, 52074, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, RWTH Aachen University, Neuenhofer Weg 21, 52074, Aachen, Germany
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, LVR-University Hospital, University of Duisburg-Essen, Wickenburgstraße 21, 45147, Essen, Germany
| | - Stefanie Trinh
- Institute of Neuroanatomy, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| |
Collapse
|
43
|
Yang X, Liu R, An Z, Li B, Lin Y, Li Y, Song B, Yuan J, Meng W, Waydhas C. Probiotic mitigates gut hypoperfusion-associated acute gastrointestinal injury in patients undergoing cardiopulmonary bypass: a randomized controlled trial. BMC Med 2025; 23:238. [PMID: 40264088 PMCID: PMC12016265 DOI: 10.1186/s12916-025-04082-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 04/15/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Acute gastrointestinal injury (AGI) after cardiopulmonary bypass (CPB) is associated with poor prognosis. This study aimed to evaluate the effect of preoperative probiotic supplementation on the incidence of AGI in patients undergoing CPB procedure. METHODS This was a double-blind, randomized controlled trial conducted in a single center. The patients undergoing HVR with CPB between September 2022 and February 2023 were randomly assigned to receive either probiotic (Lac group) or placebo (Placebo group). The probiotic was administered daily for seven days prior to surgery.Univariate and multivariate logistic regression analysis was performed to identify independent risk factors for AGI. A P-value < 0.05 was considered statistically significant. Gut microbiota composition was assessed using 16 s rRNA analysis. RESULTS A total of 52 patients were randomly assigned to two groups (26 in the Lac group, 26 in the Placebo group). Patients were followed for at least 30 days after surgery. During the follow-up period, 15 of the 52 patients (28.85%) developed AGI. The incidence of AGI was significantly lower in the Lac group (15.38%) compared to the Placebo group (42.31%), with a difference of 26.93% (P = 0.032). Moreover, patients in the Lac group had a significantly shorter ICU stay (6 [5, 36] vs. 5 [4, 5.5] days, P = 0.041) and a lower incidence of nosocomial infections (11.54% vs. 34.62%, P = 0.048). Multivariate analysis identified a higher Cardiac Surgery Score (CASUS) and CPB duration ≥ 132 min as independent risk factors for AGI, whereas probiotic supplementation was the only protective factor. Furthermore, 16S rRNA sequencing revealed significant differences in gut microbiota composition between the Lac and Placebo groups. CONCLUSIONS Preoperative probiotic supplementation may be an effective strategy to reduce the incidence of AGI and AGI-related complications in CPB patients. These findings suggest that probiotics could be considered a preventive intervention for AGI in this patient population. TRIAL REGISTRATION ClinicalTrials.gov: NCT05498948.
Collapse
Affiliation(s)
- Xiaofang Yang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730030, Gansu, China
- Department of Cardiovascular Surgery, The First Hospital of Lanzhou University, Lanzhou, , Gansu, China
| | - Ruisheng Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730030, Gansu, China
- Department of Cardiovascular Surgery, The First Hospital of Lanzhou University, Lanzhou, , Gansu, China
| | - Zhijing An
- Department of Cardiovascular Surgery, The First Hospital of Lanzhou University, Lanzhou, , Gansu, China
| | - Boxia Li
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yanyan Lin
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yuanmin Li
- Department of Cardiovascular Surgery, The First Hospital of Lanzhou University, Lanzhou, , Gansu, China
| | - Bing Song
- Department of Cardiovascular Surgery, The First Hospital of Lanzhou University, Lanzhou, , Gansu, China
| | - Jinqiu Yuan
- Clinical Research Center, Big Data Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong, China.
| | - Wenbo Meng
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730030, Gansu, China.
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, Gansu, China.
- Gansu Province Key Laboratory of Biological Therapy and Regenerative Medicine Transformation, Lanzhou, Gansu, China.
| | - Christian Waydhas
- Trauma Intensive Care, Department of Trauma Surgery, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
44
|
Zeng H, Yang H, Fu Z, Ma L, Lu L, Zeng T, Xiao Y, Lyu W. Integrated 16S rRNA and metagenomic sequencing reveals the distribution of key antibiotic resistance genes in duck gut microbiota. Poult Sci 2025; 104:105206. [PMID: 40294554 PMCID: PMC12056789 DOI: 10.1016/j.psj.2025.105206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 04/20/2025] [Accepted: 04/22/2025] [Indexed: 04/30/2025] Open
Abstract
The duck gut microbiota is essential for host health and is considered a potential reservoir for antibiotic resistance genes (ARGs). However, research on ARGs in the duck gut microbiota is limited. This study collected 120 intestinal content samples from five segments (duodenum, jejunum, ileum, cecum, and colorectum) of ducks raised under two rearing conditions (with or without an open-air swimming pool). We compiled a comprehensive inventory of microbial genes in the duck gut and conducted an analysis of microbial composition and function across all intestinal segments using 16S rRNA gene sequencing combined with metagenomics. The findings revealed that Firmicutes were the most prevalent microbes in all intestinal segments. In the foregut (duodenum, jejunum, and ileum), microbial functions were mainly related to genetic information processing such as transcription, translation, replication, and glycosynthesis/gluconeogenesis. Conversely, in the hindgut (cecum and colorectum), microbial functions were primarily associated with the biosynthesis of secondary metabolites and various metabolic pathways. The analysis of ARGs indicated a higher relative abundance of ARGs in the cecum and colorectum (P < 0.05) of ducks in the presence of an open-air swimming pool compared to the absence of one. Furthermore, through co-occurrence network analysis, we identified Bacteroides, Roseburia, Ruminococcus, and Blautia as potential hosts of ARGs such as tetQ, tet32, tet37, vanR, vanG, and acrB in the hindgut. This study provides new insights into the complex relationship between ARGs and the microbial community in duck intestines, laying a theoretical groundwork for understanding the transmission dynamics of ARGs in these ecosystems.
Collapse
Affiliation(s)
- Hongbo Zeng
- State Key Laboratory for Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Hua Yang
- State Key Laboratory for Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Zixian Fu
- College of Animal Science, Zhejiang A&F University, Hangzhou, China
| | - Lingyan Ma
- State Key Laboratory for Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Lizhi Lu
- State Key Laboratory for Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Tao Zeng
- State Key Laboratory for Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yingping Xiao
- State Key Laboratory for Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Wentao Lyu
- State Key Laboratory for Quality and Safety of Agro-Products, Institute of Agro-Product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China.
| |
Collapse
|
45
|
Hu M, Zhu X, Huang X, Hua L, Lin X, Zhang H, Hu Y, Tong T, Li L, Xuan B, Zhao Y, Zhou Y, Ding J, Ma Y, Jiang Y, Ning L, Zhang Y, Wang Z, Fang JY, Zhang Y, Xiao X, Hong J, Chen H, Li J, Chen H. Optimizing anti-PD-1/PD-L1 therapy efficacy and fecal microbiota transplantation donor selection through gut mycobiome-based enterotype. Cell Rep 2025; 44:115589. [PMID: 40257861 DOI: 10.1016/j.celrep.2025.115589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/06/2025] [Accepted: 03/28/2025] [Indexed: 04/23/2025] Open
Abstract
Immunotherapy has revolutionized cancer treatment, but response variability remains a challenge. The gut microbiome's role in therapeutic efficacy is well established, but the impact of the gut mycobiome is less understood. Using unsupervised clustering, we identify two gut mycobiome-based enterotypes, favorable type and unfavorable type, characterized by distinct microbial compositions linked to immunotherapy outcomes. Favorable-type enterotypes exhibit higher fungal and bacterial alpha diversity, enriched butyrate-producing bacteria, and metabolic pathways related to butyric acid and sugar/starch metabolism. External validation confirms their predictive value in assessing immunotherapy efficacy. Multi-omics analysis reveals increased CD8+ T cell infiltration in the tumor microenvironment of favorable-type patients. Fecal microbiota transplantation (FMT) from favorable-type donors enhances anti-PD-1 sensitivity, promotes CD8+ T cell infiltration, and boosts butyrate production in vivo. These findings highlight the gut mycobiome's role in immunotherapy response and support FMT from favorable-type donors as a potential strategy for improving treatment outcomes and patient stratification.
Collapse
Affiliation(s)
- Muni Hu
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Xiaoqiang Zhu
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China; Baoshan Branch, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200444, China
| | - Xiaowen Huang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Li Hua
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaolin Lin
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Hangyu Zhang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ye Hu
- Department of Gastroenterology, Shanghai Jiaotong University School of Medicine Xinhua Hospital, Shanghai, China
| | - Tianying Tong
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Lingxi Li
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Baoqin Xuan
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Ying Zhao
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Yilu Zhou
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Jinmei Ding
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Yanru Ma
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Yi Jiang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Lijun Ning
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Yue Zhang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Zhenyu Wang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Jing-Yuan Fang
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Youwei Zhang
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou 221009, China
| | - Xiuying Xiao
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jie Hong
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China
| | - Huimin Chen
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China.
| | - Jiantao Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Haoyan Chen
- State Key Laboratory of Systems Medicine for Cancer, NHC Key Laboratory of Digestive Diseases, Division of Gastroenterology and Hepatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai Cancer Institute, Shanghai 200001, China.
| |
Collapse
|
46
|
Fuggle R, Matias MG, Mayer-Pinto M, Marzinelli EM. Multiple stressors affect function rather than taxonomic structure of freshwater microbial communities. NPJ Biofilms Microbiomes 2025; 11:60. [PMID: 40251215 PMCID: PMC12008304 DOI: 10.1038/s41522-025-00700-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
Microbial community responses to environmental stressors are often characterised by assessing changes in taxonomic structure, but such changes, or lack thereof, may not reflect functional changes that are critical to ecosystem processes. We investigated the individual and combined effects of nutrient enrichment ( + 10 mg/L N, + 1 mg/L P) and salinisation ( + 15 g/L NaCl)-key stressors in freshwater systems-on the taxonomic structure and metabolic function of benthic microbial communities using 1000 L open freshwater ponds established >10 years ago in the field. Combined stressors drove strong decreases in maximum and mean total carbon metabolic rates and shifted carbon metabolic profiles compared to either stressor individually and compared to ambient conditions. These metabolic functional changes did not recover through time and occurred without significant alterations in bacterial community taxonomic structure. These results imply that critical functions, including organic carbon release, are likely to be impaired under multiple stressors, even when taxonomic structure remains stable.
Collapse
Affiliation(s)
- Rose Fuggle
- The University of Sydney, School of Life and Environmental Sciences, Sydney, NSW, 2006, Australia.
| | | | - Mariana Mayer-Pinto
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, UNSW, Sydney, NSW, 2052, Australia
| | - Ezequiel M Marzinelli
- The University of Sydney, School of Life and Environmental Sciences, Sydney, NSW, 2006, Australia
| |
Collapse
|
47
|
Schille TB, Sprague JL, Naglik JR, Brunke S, Hube B. Commensalism and pathogenesis of Candida albicans at the mucosal interface. Nat Rev Microbiol 2025:10.1038/s41579-025-01174-x. [PMID: 40247134 DOI: 10.1038/s41579-025-01174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2025] [Indexed: 04/19/2025]
Abstract
Fungi are important and often underestimated human pathogens. Infections with fungi mostly originate from the environment, from soil or airborne spores. By contrast, Candida albicans, one of the most common and clinically important fungal pathogens, permanently exists in the vast majority of healthy individuals as a member of the human mucosal microbiota. Only under certain circumstances will these commensals cause infections. However, although the pathogenic behaviour and disease manifestation of C. albicans have been at the centre of research for many years, its asymptomatic colonization of mucosal surfaces remains surprisingly understudied. In this Review, we discuss the interplay of the fungus, the host and the microbiome on the dualism of commensal and pathogenic life of C. albicans, and how commensal growth is controlled and permitted. We explore hypotheses that could explain how the mucosal environment shapes C. albicans adaptations to its commensal lifestyle, while still maintaining or even increasing its pathogenic potential.
Collapse
Affiliation(s)
- Tim B Schille
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Jakob L Sprague
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany.
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
48
|
Beaudoin CA, Norget S, Omran Z, Hala S, Daqeeq AH, Burnet PWJ, Blundell TL, van Tonder AJ. Similarity of drug targets to human microbiome metaproteome promotes pharmacological promiscuity. THE PHARMACOGENOMICS JOURNAL 2025; 25:9. [PMID: 40246834 PMCID: PMC12006021 DOI: 10.1038/s41397-025-00367-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 02/27/2025] [Accepted: 03/24/2025] [Indexed: 04/19/2025]
Abstract
Similarity between candidate drug targets and human proteins is commonly assessed to minimize the occurrence of side effects. Although numerous drugs have been found to disrupt the health of the human microbiome, no comprehensive comparison between established drug targets and the human microbiome metaproteome has yet been conducted. Therefore, herein, sequence and structure alignments between human and pathogen drug targets and representative human gut, oral, and vaginal microbiome metaproteomes were performed. Both human and pathogen drug targets were found to be similar in sequence, function, structure, and drug binding capacity to proteins in diverse pathogenic and non-pathogenic bacteria from all three microbiomes. The gut metaproteome was identified as particularly susceptible overall to off-target effects. Certain symptoms, such as infections and immune disorders, may be more common among drugs that non-selectively target host microbiota. These findings suggest that similarities between human microbiome metaproteomes and drug target candidates should be routinely checked.
Collapse
Affiliation(s)
| | - Shannon Norget
- Department of Psychology, Health & Technology, University of Twente, Enschede, the Netherlands
| | - Ziad Omran
- King Abdullah International Medical Research Center, King Saud Bin Abdelaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Sharif Hala
- Biothreat Department, Public Health Laboratory, Public Health Authority, Riyadh, Saudi Arabia
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Abdullah H Daqeeq
- Department of Anesthesia, International Medical Center, Jeddah, Kingdom of Saudi Arabia
| | | | - Tom L Blundell
- Victor Phillip Dahdaleh Heart and Lung Research Institute, Biomedical Campus, Trumpington, Cambridge, UK
| | | |
Collapse
|
49
|
Zhang Z, Sun J, Wang D, Lin T, Yin Y, Wang W, Wang Y, Wang Z, Fan L, Jiao X. Effects of rotation corn on potato yield, quality, and soil microbial communities. Front Microbiol 2025; 16:1493333. [PMID: 40309109 PMCID: PMC12040919 DOI: 10.3389/fmicb.2025.1493333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 03/19/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction Potato is an important crop that can be used both as grain and vegetable in northern China. However, the continuous cropping system of potato has led to a sharp decline in its yield and quality. As one of the effective strategies to alleviate the continuous cropping obstacle, crop rotation has received extensive attention in agricultural practices. On this basis, we have conducted an in-depth exploration of the effects of the potato-maize rotation system on the structure and diversity of the soil microbial community, aiming to analyze the internal correlation mechanism between the structure of the soil microbial community and the yield and quality of crops. Methods This study was based on fields that had been under potato monoculture for five years and established six experimental treatments: potato-potato-potato (IR-A), potato-maize-potato (IR-B), potato-maize-maize (IR-C), potato-potato-potato (RF-A), potato-maize-potato (RF-B), and potato-maize-maize (RF-C). Results The results showed that under the IR planting model, IR-B significantly increased potato yield and vitamin C content while reducing reducing sugar content compared to IR-A (p < 0.05). In the RF planting model, RF-B significantly increased potato yield, starch content, and vitamin C content compared to RF-A (p < 0.05). Microbial community structure results indicated that crop rotation significantly enhanced the relative abundance of microorganisms such as Bradyrhizobium, Pseudomonas, Sphingomonas, Purpureocillium, Streptomyces, and Halovivax (p < 0.05). These microorganisms are involved in the cycling of carbon, phosphorus, and other nutrients in the soil, playing an important role in promoting root growth, organic matter decomposition, and alleviating soil salinization. The LEfSe and RDA indicated significant differences in microbial communities between monoculture and crop rotation (p < 0.05), with soil slow-growing rhizobia, Burkholderia, and actinomycetes positively correlated with potato yield and quality. Additionally, KEGG functional annotation of different treatments revealed that K00239, K00626, K01681, and K01915 were involved in three key metabolic pathways related to carbon and nitrogen. A total of 20 significantly enriched pathways were identified (p < 0.05), among which K01681 is involved in the tricarboxylic acid cycle and is a differential gene in the RF-B treatment, suggesting that the efficient expression of K01681 during crop rotation contributes to the material cycling of the soil ecosystem. LEfSe analysis of the bins revealed that under the RF-C treatment, the relative abundance of Hyphomicrobiales was significantly higher than in other treatments (p < 0.05). Hyphomicrobiales are involved in the nitrogen fixation process and play an important role in soil nutrient cycling and plant nutrition. In summary, the potato-maize rotation significantly altered the composition of soil microbial communities (p < 0.05), increasing the relative abundance of beneficial microorganisms. This change helps maintain the health of the soil ecosystem, promotes nutrient cycling, reduces the incidence of diseases, and effectively improves both the yield and quality of potatoes. Discussion The potato-maize rotation significantly altered the composition of soil microbial communities (p < 0.05), increasing the relative abundance of beneficial microorganisms. This change helps maintain the health of the soil ecosystem, promotes nutrient cycling, reduces the incidence of diseases, and effectively improves both the yield and quality of potatoes.
Collapse
Affiliation(s)
- Zhicheng Zhang
- Inner Mongolia Agricultural University, College of Agriculture, Hohhot, China
- Ulanqab Institute of Agricultural and Forestry Sciences, Potato Research Laboratory, Ulanqab, China
| | - Jiying Sun
- Inner Mongolia Agricultural University, College of Agriculture, Hohhot, China
| | - Dan Wang
- Jining Normal University, College of Life Science and Technology, Ulanqab, China
| | - Tuanrong Lin
- Ulanqab Institute of Agricultural and Forestry Sciences, Potato Research Laboratory, Ulanqab, China
| | - Yuhe Yin
- Ulanqab Institute of Agricultural and Forestry Sciences, Potato Research Laboratory, Ulanqab, China
| | - Wei Wang
- Ulanqab Institute of Agricultural and Forestry Sciences, Potato Research Laboratory, Ulanqab, China
| | - Yufeng Wang
- Ulanqab Institute of Agricultural and Forestry Sciences, Potato Research Laboratory, Ulanqab, China
| | - Zhen Wang
- Ulanqab Institute of Agricultural and Forestry Sciences, Potato Research Laboratory, Ulanqab, China
| | - Longqiu Fan
- Ulanqab Institute of Agricultural and Forestry Sciences, Potato Research Laboratory, Ulanqab, China
| | - Xinlei Jiao
- Ulanqab Institute of Agricultural and Forestry Sciences, Potato Research Laboratory, Ulanqab, China
| |
Collapse
|
50
|
Liu W, Wang L, Ou J, Peng D, Zhang Y, Chen W, Wang Y. Gut Microbiota Metabolites and Chronic Diseases: Interactions, Mechanisms, and Therapeutic Strategies. Int J Mol Sci 2025; 26:3752. [PMID: 40332366 PMCID: PMC12027615 DOI: 10.3390/ijms26083752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/05/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
The gut microbiota, shaped by factors such as diet, lifestyle, and genetics, plays a pivotal role in regulating host metabolism, immune function, and overall health. The diversity and balance of the gut microbiota are closely linked to the onset and progression of various chronic diseases. A growing body of evidence has demonstrated that alterations in the composition, function, and metabolites of the gut microbiota are significantly associated with cardiovascular diseases, including hypertension, atherosclerosis, and heart failure; metabolic disorders such as obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease; and gastrointestinal conditions like inflammatory bowel disease and colorectal cancer. Despite substantial advances in microbiome research, challenges remain in fully elucidating the causal relationships between the gut microbiota and disease, as well as in translating these insights into clinical applications. This review aims to investigate the regulatory pathways via which the gut microbiota affects cardiovascular health, metabolic function, and gastrointestinal disease. Additionally, it highlights emerging strategies for the prevention and treatment of these chronic conditions, focusing on microbiota-targeted therapies and personalized dietary interventions as promising approaches for improving health outcomes.
Collapse
Affiliation(s)
- Wenwen Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
| | - Lei Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
- Ministry of Education-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Institute of Traditional Chinese Medicine Resources Protection and Development, Hefei 230012, China
| | - Jinmei Ou
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
- Anhui Key Laboratory of New Manufacturing Technology of Chinese Medicine Pieces, Hefei 230012, China
| | - Daiyin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
- Ministry of Education-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Institute of Traditional Chinese Medicine Resources Protection and Development, Hefei 230012, China
| | - Yue Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
- Ministry of Education-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
| | - Weidong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
- Ministry of Education-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Institute of Traditional Chinese Medicine Resources Protection and Development, Hefei 230012, China
- Anhui Key Laboratory of New Manufacturing Technology of Chinese Medicine Pieces, Hefei 230012, China
| | - Yanyan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; (W.L.); (L.W.); (J.O.); (D.P.); (W.C.)
- Ministry of Education-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei 230012, China
- Institute of Traditional Chinese Medicine Resources Protection and Development, Hefei 230012, China
| |
Collapse
|