1
|
O’Sullivan J, Patel S, Leventhal GE, Fitzgerald RS, Laserna-Mendieta EJ, Huseyin CE, Konstantinidou N, Rutherford E, Lavelle A, Dabbagh K, DeSantis TZ, Shanahan F, Temko A, Iwai S, Claesson MJ. Host-microbe multi-omics and succinotype profiling have prognostic value for future relapse in patients with inflammatory bowel disease. Gut Microbes 2025; 17:2450207. [PMID: 39812341 PMCID: PMC11740686 DOI: 10.1080/19490976.2025.2450207] [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: 07/16/2024] [Revised: 11/07/2024] [Accepted: 01/02/2025] [Indexed: 01/16/2025] Open
Abstract
Crohn's disease (CD) and ulcerative colitis (UC) are chronic relapsing inflammatory bowel disorders (IBD), the pathogenesis of which is uncertain but includes genetic susceptibility factors, immune-mediated tissue injury and environmental influences, most of which appear to act via the gut microbiome. We hypothesized that host-microbe alterations could be used to prognostically stratify patients experiencing relapses up to four years after endoscopy. We therefore examined multiple omics data, including published and new datasets, generated from paired inflamed and non-inflamed mucosal biopsies from 142 patients with IBD (54 CD; 88 UC) and from 34 control (non-diseased) biopsies. The relapse-predictive potential of 16S rRNA gene and transcript amplicons (standing and active microbiota) were investigated along with host transcriptomics, epigenomics and genetics. While standard single-omics analysis could not distinguish between patients who relapsed and those that remained in remission within four years of colonoscopy, we did find an association between the number of flares and a patient's succinotype. Our multi-omics machine learning approach was also able to predict relapse when combining features from the microbiome and human host. Therefore multi-omics, rather than single omics, better predicts relapse within 4 years of colonoscopy, while a patient's succinotype is associated with a higher frequency of relapses.
Collapse
Affiliation(s)
- Jill O’Sullivan
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- SFI Centre for Research Training in Genomics Data Science, University of Galway, Galway, Ireland
| | - Shriram Patel
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- SeqBiome Ltd, Cork, Ireland
| | | | - Rachel S. Fitzgerald
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Emilio J. Laserna-Mendieta
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Chloe E. Huseyin
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Nina Konstantinidou
- School of Microbiology, University College Cork, Cork, Ireland
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Erica Rutherford
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Aonghus Lavelle
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, County Cork, Ireland
| | - Karim Dabbagh
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Todd Z. DeSantis
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Fergus Shanahan
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| | - Andriy Temko
- Department of Electrical and Electronic Engineering, University College Cork, Cork, Ireland
| | - Shoko Iwai
- Department of Informatics, Second Genome Inc, South San Francisco, California, USA
| | - Marcus J. Claesson
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| |
Collapse
|
2
|
Nepal S, Shi N, Hoyd R, Spakowicz DJ, Orwoll E, Shikany JM, Napoli N, Tabung FK. Role of insulinemic and inflammatory dietary patterns on gut microbial composition and circulating biomarkers of metabolic health among older American men. Gut Microbes 2025; 17:2497400. [PMID: 40296253 PMCID: PMC12045561 DOI: 10.1080/19490976.2025.2497400] [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: 01/28/2025] [Revised: 03/25/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
Chronic low-grade inflammation and hyperinsulinemia are linked with metabolic dysfunction and dysbiosis. This study investigated the role of dietary inflammatory and insulinemic potential on gut microbiome and circulating health biomarkers in older men. Data from the Osteoporotic Fractures in Men (MrOS) study were analyzed. Reversed Empirical Dietary Inflammatory Pattern (rEDIP), Empirical Dietary Index for Hyperinsulinemia (rEDIH), and Healthy Eating Index (HEI)-2020 scores were computed from food frequency questionnaire data. Stool samples were profiled using 16S rRNA sequencing. Elastic net regression identified diet-associated microbial profiles and multivariable-adjusted linear regression assessed diet-biomarker associations. Higher rEDIP, rEDIH, and HEI-2020 scores were positively associated with gut microbiota alpha diversity. Specific genera, including Intestinibacter and Lachnospira, associated positively, while Dielma, Peptococcus, Feacalitalea, and Negativibaccilus associated inversely with healthier dietary patterns. When evaluating changes in dietary patterns between baseline and visit 4 ( ~ 14 years), these genera tended to define rEDIP, rEDIH more than HEI-2020. In addition, higher dietary quality was linked to better biomarker profiles, including lower creatinine, sodium, triglycerides, and insulin resistance. Beneficial effects of higher dietary quality on health may be mediated by the ability of diet to regulate gut microbial composition and metabolic biomarker profiles.
Collapse
Affiliation(s)
- Sushma Nepal
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Ni Shi
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Rebecca Hoyd
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Daniel J. Spakowicz
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Eric Orwoll
- Department of Medicine, Oregon Health & Sciences University, Portland, OR, USA
| | - James M. Shikany
- Division of General Internal Medicine and Population Science, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nicola Napoli
- Unit of Endocrinology and Diabetes, Department of Medicine, Campus Bio-Medico University of Rome, Rome, Italy
- Division of Bone and Mineral Diseases, Washington University, St Louis, MO, USA
| | - Fred K. Tabung
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, OH, USA
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Division of Epidemiology, College of Public Health, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
3
|
Masaadeh AH, Eletrebi M, Parajuli B, De Jager N, Bosch DE. Human colitis-associated colorectal carcinoma progression is accompanied by dysbiosis with enriched pathobionts. Gut Microbes 2025; 17:2479774. [PMID: 40094201 PMCID: PMC11917176 DOI: 10.1080/19490976.2025.2479774] [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: 01/06/2025] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
Dysbiosis and pathobionts contribute to inflammation and the risk of colitis-associated carcinoma (CAC) in animal models, but their roles in humans with this uncommon disease are unknown. We identified microbiome differences in human CAC compared with longstanding inflammatory bowel disease (IBD) and sporadic colorectal carcinoma (CRC). Twenty-four CAC resections were matched with CRC and IBD controls. Methods included histopathology, 16S rDNA metagenomics, and pathobiont-specific qPCR. Beta diversity differed by diagnosis (PERMANOVA p = 0.007). The distinguishing taxa included Akkermansia enriched in CRC, and Bacteroides spp. enriched in IBD. The non-neoplastic mucosae presented distinct beta diversity (p = 0.005), but the CAC/CRC tumor microbiomes were similar (p = 0.7). Within metastases and margins, Enterobacteriaceae were enriched in CAC, and Bacteroidales in CRC. Pathobiont-specific qPCR confirmed a greater frequency of pks+ E. coli and enterotoxigenic Bacteroides fragilis in CAC than IBD. High alpha diversity was associated with active inflammation, advanced cancer stage, and shorter overall survival (log-rank p = 0.008). Mucosal microbiomes distinguish CAC from longstanding IBD, implicating pathobionts as markers for disease progression. Integrating our findings with prior animal model research, pathobionts promote carcinogenesis in IBD patients through genotoxicity and host cell signaling.
Collapse
Affiliation(s)
- Amr H. Masaadeh
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Mohamed Eletrebi
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Bishal Parajuli
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Nicola De Jager
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Dustin E. Bosch
- Department of Pathology, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, Iowa City, IA, USA
| |
Collapse
|
4
|
Rodrigues CS, Gaifem J, Pereira MS, Alves MF, Silva M, Padrão N, Cavadas B, Moreira-Barbosa C, Alves I, Marcos-Pinto R, Torres J, Lavelle A, Colombel JF, Sokol H, Pinho SS. Alterations in mucosa branched N-glycans lead to dysbiosis and downregulation of ILC3: a key driver of intestinal inflammation. Gut Microbes 2025; 17:2461210. [PMID: 39918275 PMCID: PMC11810091 DOI: 10.1080/19490976.2025.2461210] [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: 08/29/2024] [Revised: 11/29/2024] [Accepted: 01/13/2025] [Indexed: 02/12/2025] Open
Abstract
The perturbation of the symbiotic relationship between microbes and intestinal immune system contributes to gut inflammation and Inflammatory Bowel Disease (IBD) development. The host mucosa glycans (glycocalyx) creates a major biological interface between gut microorganisms and host immunity that remains ill-defined. Glycans are essential players in IBD immunopathogenesis, even years before disease onset. However, how changes in mucosa glycosylation shape microbiome and how this impact gut immune response and inflammation remains to be clarified. Here, we revealed that alterations in the expression of complex branched N-glycans at gut mucosa surface, modeled in glycoengineered mice, resulted in dysbiosis, with a deficiency in Firmicutes bacteria. Concomitantly, this mucosa N-glycan switch was associated with a downregulation of type 3 innate lymphoid cells (ILC3)-mediated immune response, leading to the transition of ILC3 toward an ILC1 proinflammatory phenotype and increased TNFα production. In addition, we demonstrated that the mucosa glycosylation remodeling through prophylactic supplementation with glycans at steady state was able to restore microbial-derived short-chain fatty acids and microbial sensing (by NOD2 expression) alongside the rescue of the expression of ILC3 module, suppressing intestinal inflammation and controlling disease onset. In a complementary approach, we further showed that IBD patients, often displaying dysbiosis, exhibited a tendency of decreased MGAT5 expression at epithelial cells that was accompanied by reduced ILC3 expression in gut mucosa. Altogether, these results unlock the effects of alterations in mucosa glycome composition in the regulation of the bidirectional crosstalk between microbiota and gut immune response, revealing host branched N-glycans/microbiota/ILC3 axis as an essential pathway in gut homeostasis and in preventing health to intestinal inflammation transition.
Collapse
Affiliation(s)
- Cláudia S. Rodrigues
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Joana Gaifem
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
| | - Márcia S. Pereira
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Maria Francisca Alves
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Faculty of Sciences, University of Porto, Porto, Portugal
| | - Mariana Silva
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
| | - Nuno Padrão
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| | - Bruno Cavadas
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
| | | | - Inês Alves
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
| | - Ricardo Marcos-Pinto
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Department of Gastroenterology, Centro Hospitalar do Porto, Porto, Portugal
- Centro de Investigação em Tecnologias e Serviços de Saúde, University of Porto, Porto, Portugal
| | - Joana Torres
- Division of Gastroenterology, Hospital Beatriz Ângelo, Loures, Portugal
- Faculty of Medicine, University of Lisbon, Lisbon, Portugal
- Division of Gastroenterology, Hospital da Luz, Lisbon, Portugal
| | - Aonghus Lavelle
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Gastroenterology Department, Sorbonne Université, INSERM, Paris, France
| | - Jean-Frederic Colombel
- Henry D. Janowitz Division of Gastroenterology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Harry Sokol
- Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint-Antoine Hospital, Gastroenterology Department, Sorbonne Université, INSERM, Paris, France
- INRAE, AgroParisTech, Micalis Institute, Université Paris-Saclay, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Salomé S. Pinho
- Institute for Research and Innovation in Health (i3S), Immunology, Cancer & Glycomedicine Group, University of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
5
|
Jiao Y, Ren J, Xie S, Yuan N, Shen J, Yin H, Wang J, Guo H, Cao J, Wang X, Wu D, Zhou Z, Qi X. Raffinose-metabolizing bacteria impair radiation-associated hematopoietic recovery via the bile acid/FXR/NF-κB signaling pathway. Gut Microbes 2025; 17:2488105. [PMID: 40192235 PMCID: PMC11980471 DOI: 10.1080/19490976.2025.2488105] [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: 12/20/2024] [Revised: 03/23/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
Radiation-associated hematopoietic recovery (RAHR) is critical for mitigating lethal complications of acute radiation syndrome (ARS), yet therapeutic strategies remain limited. Through integrated multi-omics analysis of a total body irradiation (TBI) mouse model, we identify Bacteroides acidifaciens-dominated gut microbiota as key mediators of RAHR impairment. 16S ribosomal rRNA sequencing revealed TBI-induced dysbiosis characterized by Bacteroidaceae enrichment, while functional metagenomics identified raffinose metabolism as the most significantly perturbed pathway. Notably, raffinose supplementation (10% w/v) recapitulated radiation-induced microbiota shifts and delayed bone marrow recovery. Fecal microbiota transplantation (FMT) revealed a causative role for raffinose-metabolizing microbiota, particularly Bacteroides acidifaciens, in delaying RAHR progression. Mechanistically, B. acidifaciens-mediated bile acid deconjugation activated FXR, subsequently suppressing NF-κB-dependent hematopoietic recovery. Therapeutic FXR inhibition via ursodeoxycholic acid (UDCA) had been shown to be a viable method for rescuing RAHR. Our results delineated a microbiome-bile acid-FXR axis as a master regulator of post-irradiation hematopoiesis. Targeting B. acidifaciens or its metabolic derivatives could represent a translatable strategy to mitigate radiation-induced hematopoietic injury.
Collapse
Affiliation(s)
- Yang Jiao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Suzhou, China
| | - Jiawei Ren
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Shichang Xie
- Key Laboratory of Alkene-Carbon Fibers-Based Technology & Application for Detection of Major Infectious Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Cancer Institute, Suzhou Medical College, Soochow University, Suzhou, China
| | - Nan Yuan
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Jiaqi Shen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China
| | - Huafang Yin
- The Affiliated Jiangyin People’s Hospital of Nantong University, Jiangyin, China
| | - Jian Wang
- The Affiliated Jiangyin People’s Hospital of Nantong University, Jiangyin, China
| | - Hongjuan Guo
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Jianping Cao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
- Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Xin Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products & Institute of Food Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Depei Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Suzhou, China
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China
| | - Zhemin Zhou
- Key Laboratory of Alkene-Carbon Fibers-Based Technology & Application for Detection of Major Infectious Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Cancer Institute, Suzhou Medical College, Soochow University, Suzhou, China
- Cancer Institute, Suzhou Medical College, The Second Affiliated Hospital of Soochow University, Suzhou, China
- National Center of Technology Innovation for Biopharmaceuticals, Suzhou Biomedical Industry Innovation Center, Suzhou, China
| | - Xiaofei Qi
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, Collaborative Innovation Center of Hematology, Suzhou, China
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Cyrus Tang Hematology Center, Soochow University, Suzhou, China
| |
Collapse
|
6
|
Alizadeh M, Wong U, Siaton BC, France MT, Patil SA, George L, Hudhud D, Motwani K, Scott WH, Raufman JP, von Rosenvinge EC, Cross RK, Ravel J. The intestinal mucosa-associated microbiota in IBD-associated arthritis displays lower relative abundance of Roseburia intestinalis. Gut Microbes 2025; 17:2505114. [PMID: 40382763 PMCID: PMC12087651 DOI: 10.1080/19490976.2025.2505114] [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: 02/07/2025] [Revised: 03/26/2025] [Accepted: 05/07/2025] [Indexed: 05/20/2025] Open
Abstract
The most common extra-intestinal manifestation (EIM) of inflammatory bowel disease (IBD), IBD-associated arthritis (IAA), occurs in 25-40% of patients and can be debilitating. In IBD, mucosal and stool microbiota richness is decreased, and compositional changes can precede or accompany disease onset. Likewise, spondyloarthritides are associated with altered gut microbiota, with overlapping bacterial signatures observed in IBD, suggesting key shared microbial factors are involved in both conditions. Much has been learned about the role of the intestinal microbiome in IBD, but less is known regarding its role in IAA. To address this knowledge gap, we analyzed the mucosa-associated intestinal microbiota of participants enrolled in the LOCATION-IBD cohort. Microbiota composition was established using 16S rRNA gene amplicon sequencing of intestinal biopsy samples taken from participants with IBD, with or without arthropathy. Microbiota samples clustered predominantly by participant, and similar taxa were present across the colon. The mucosal intestinal microbiota of females with IAA displayed a lower relative abundance of R. intestinalis, while males with IAA had a higher relative abundance of Corynebacterium, even when controlling for IBD-type, whether samples were taken from a site of inflammation and intestinal location. These findings indicate the mucosa-associated intestinal microbiota is associated with IAA in a sex-specific manner.
Collapse
Affiliation(s)
- Madeline Alizadeh
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Uni Wong
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Bernadette C. Siaton
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
- Division of Rheumatology and Clinical Immunology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael T. France
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Seema A. Patil
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Lauren George
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Dania Hudhud
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Kiran Motwani
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - William H. Scott
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Jean-Pierre Raufman
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Erik C. von Rosenvinge
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Veterans Affairs, Veterans Affairs Maryland Health Care System, Baltimore, MD, USA
| | - Raymond K. Cross
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jacques Ravel
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
7
|
He Y, Zhao G, Ouyang X, Wang S, Chen Y, Li C, He Y, Gao J, Han S, Zhao J, Wang J, Wang C. Creatine-mediated ferroptosis inhibition is involved in the intestinal radioprotection of daytime-restricted feeding. Gut Microbes 2025; 17:2489072. [PMID: 40205678 PMCID: PMC11988229 DOI: 10.1080/19490976.2025.2489072] [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: 10/26/2024] [Revised: 03/11/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
Ionizing radiation-induced intestinal injury (IRIII) is a catastrophic disease lack of sufficient medical countermeasures currently. Regulation of the gut microbiota through dietary adjustments is a potential strategy to mitigate IRIII. Time-restricted feeding (TRF) is an emerging behavioral nutrition intervention with pleiotropic health benefits. Whether this dietary pattern influences the pathogenesis of IRIII remains vague. We evaluated the impact of TRF on intestinal radiosensitivity in this study and discovered that only daytime TRF (DTRF), not nighttime TRF, could ameliorate intestinal damage in mice that received a high dose of IR. Faecal metagenomic and metabolomic studies revealed that the intestinal creatine level was increased by approximate 9 times by DTRF, to which the Bifidobacterium pseudolongum enrichment contribute. Further investigations showed that creatine could activate the energy sensor AMP-activated protein kinase in irradiated enterocytes and induce phosphorylation of acetyl-CoA carboxylase, resulting in reduced production of polyunsaturated fatty acids and reduced ferroptosis after IR. The administration of creatine mitigated IRIII and reduced bacteremia and proinflammatory responses. Blockade of creatine import compromised the ferroptosis inhibition and mitigation of DTRF on IRIII. Our study demonstrates a radioprotective dietary mode that can reshape the gut microbiota and increase intestinal creatine, which can suppress IR-induced ferroptosis, thereby providing effective countermeasures for IRIII prevention.
Collapse
Affiliation(s)
- Yingjuan He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Gaomei Zhao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Xue Ouyang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Shaobo Wang
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Yin Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Chenwenya Li
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Yongwu He
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jining Gao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Songling Han
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jinghong Zhao
- Department of Nephrology, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Cheng Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury of PLA, College of Preventive Medicine, Army Medical University, Chongqing, China
| |
Collapse
|
8
|
Driuchina A, Isola V, Hulmi JJ, Salmi VM, Hintikka J, Ahtiainen JP, Pekkala S. Unveiling the impact of competition weight loss on gut microbiota: alterations in diversity, composition, and predicted metabolic functions. J Int Soc Sports Nutr 2025; 22:2474561. [PMID: 40033182 PMCID: PMC11881659 DOI: 10.1080/15502783.2025.2474561] [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: 03/26/2024] [Accepted: 02/26/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Competitive sports and sports nutrition, popular among amateur athletes aiming for a lean physique, have limited research on gut microbiota. METHODS We conducted a 46-week study to analyze the consequences of fat loss and diet restrictions in 23 fitness athletes who prepared for a physique competition. Body composition, dietary intakes, serum cytokines and chemokines, and fecal samples were analyzed. RESULTS Fat loss through caloric restriction and aerobic exercise led to an increased phylogenetic diversity of gut microbiota and changes in the composition of gut microbiota, with Faecalibacterium, Lachnospiraceae, Bacteroides, and Intestinimonas showing altered abundances. Fat loss also changed the predicted microbial functions responsible for the metabolism of carbohydrates and amino acids. Consumption of energy, carbohydrates, fiber, vitamins and minerals, and various fatty acids decreased during the preparation for the competition, which was partly associated with changes in gut microbiota. Several cytokine levels decreased (IL1a, IL1b, IL10, and TFNα), and certain chemokine levels increased (GROa and RANTES). During the 23-week regain period after the competition, gut microbiota showed signs of recovery, with increased diversity compared to pre- and post-competition measurements. Most taxonomic changes returned to their baseline levels after the regain period. CONCLUSIONS The study highlights the dynamic nature of gut microbiota and its response to fat loss and regain in non-obese fitness/physique competitors and provides novel insights into how competitive sports and sports nutrition can influence the gut ecosystem.
Collapse
Affiliation(s)
- Anastasiia Driuchina
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Ville Isola
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Juha J Hulmi
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Vera M Salmi
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Jukka Hintikka
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Juha P Ahtiainen
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
| | - Satu Pekkala
- University of Jyväskylä, Faculty of Sport and Health Sciences, Jyväskylä, Finland
- Turku University Hospital, Department of Clinical Microbiology, Turku, Finland
| |
Collapse
|
9
|
Qing L, Qian X, Zhu H, Wang J, Sun J, Jin Z, Tang X, Zhao Y, Wang G, Zhao J, Chen W, Tian P. Maternal-infant probiotic transmission mitigates early-life stress-induced autism in mice. Gut Microbes 2025; 17:2456584. [PMID: 39931863 PMCID: PMC11817528 DOI: 10.1080/19490976.2025.2456584] [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: 11/09/2024] [Revised: 12/14/2024] [Accepted: 01/13/2025] [Indexed: 02/14/2025] Open
Abstract
Autism, a disorder influenced by both genetic and environmental factors, presents significant challenges for prevention and treatment. While maternal-infant gut microbiota has been a focus in autism research, preventive strategies targeting maternal gut microbiota remain underexplored. This study demonstrates that prenatal probiotic intake can effectively prevent maternal separation-induced autistic-like behaviors in offspring without altering the embryonic neurodevelopment in mice. Using specific PCR primers and cross-fostering experiments, we traced the vertical transmission of probiotics, primarily via fecal/vaginal contamination. Early probiotic colonization conferred resilience against stress-induced gut pathogenic microbes and Th17-mediated peripheral inflammation while significantly inhibiting hypermyelination and neuroinflammation linked to systemic inflammation. Microbial metabolites like tyrosol and xanthurenic acid alleviated neuroinflammation and hypermyelination in vitro, though the causal relationship among neuroinflammation, hypermyelination, and autism in vivo requires further validation. These findings underscore the importance of the maternal-infant microbiota transmission window in autism prevention and highlight the clinical potential of prenatal probiotic interventions.
Collapse
Affiliation(s)
- Li Qing
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xin Qian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Huiyue Zhu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jingyu Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jingge Sun
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Zhiying Jin
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Xinyu Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Yingqi Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Gang Wang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R. China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou, P. R. China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, P. R. China
| | - Peijun Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, P. R. China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, P. R. China
| |
Collapse
|
10
|
Cantu-Jungles TM, Agamennone V, Van den Broek TJ, Schuren FHJ, Hamaker B. Systematically-designed mixtures outperform single fibers for gut microbiota support. Gut Microbes 2025; 17:2442521. [PMID: 39704614 DOI: 10.1080/19490976.2024.2442521] [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: 05/15/2024] [Revised: 10/03/2024] [Accepted: 12/09/2024] [Indexed: 12/21/2024] Open
Abstract
Dietary fiber interventions to modulate the gut microbiota have largely relied on isolated fibers or specific fiber sources. We hypothesized that fibers systematically blended could promote more health-related bacterial groups. Initially, pooled in vitro fecal fermentations were used to design dietary fiber mixtures to support complementary microbial groups related to health. Then, microbial responses were compared for the designed mixtures versus their single fiber components in vitro using fecal samples from a separate cohort of 10 healthy adults. The designed fiber mixtures outperformed individual fibers in supporting bacterial taxa across donors resulting in superior alpha diversity and unexpected higher SCFA production. Moreover, unique shifts in community structure and specific taxa were observed for fiber mixtures that were not observed for single fibers, suggesting a synergistic effect when certain fibers are put together. Fiber mixture responses were remarkably more consistent than individual fibers across donors in promoting several taxa, especially butyrate producers from the Clostridium cluster XIVa. This is the first demonstration of synergistic fiber interactions for superior support of a diverse group of important beneficial microbes consistent across people, and unexpectedly high SCFA production. Overall, harnessing the synergistic potential of designed fiber mixtures represents a promising and more efficacious avenue for future prebiotic development.
Collapse
Affiliation(s)
- T M Cantu-Jungles
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, USA
| | - V Agamennone
- Microbiology and Systems Biology Group, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, the Netherlands
| | - T J Van den Broek
- Microbiology and Systems Biology Group, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, the Netherlands
| | - F H J Schuren
- Microbiology and Systems Biology Group, The Netherlands Organization for Applied Scientific Research (TNO), Zeist, the Netherlands
| | - B Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
11
|
Akagbosu CO, McCauley KE, Namasivayam S, Romero-Soto HN, O’Brien W, Bacorn M, Bohrnsen E, Schwarz B, Mistry S, Burns AS, Perez-Chaparro PJ, Chen Q, LaPoint P, Patel A, Krausfeldt LE, Subramanian P, Sellers BA, Cheung F, Apps R, Douagi I, Levy S, Nadler EP, Hourigan SK. Gut microbiome shifts in adolescents after sleeve gastrectomy with increased oral-associated taxa and pro-inflammatory potential. Gut Microbes 2025; 17:2467833. [PMID: 39971742 PMCID: PMC11845021 DOI: 10.1080/19490976.2025.2467833] [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: 09/26/2024] [Revised: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025] Open
Abstract
Bariatric surgery is highly effective in achieving weight loss in children and adolescents with severe obesity, however the underlying mechanisms are incompletely understood, and gut microbiome changes are unknown. Here, we show that adolescents exhibit significant gut microbiome and metabolome shifts several months after laparoscopic vertical sleeve gastrectomy (VSG), with increased alpha diversity and notably with enrichment of oral-associated taxa. To assess causality of the microbiome/metabolome changes in phenotype, pre-VSG and post-VSG stool was transplanted into germ-free mice. Post-VSG stool was not associated with any beneficial outcomes such as adiposity reduction compared pre-VSG stool. However, post-VSG stool exhibited a potentially inflammatory phenotype with increased intestinal Th17 and decreased regulatory T cells. Concomitantly, we found elevated fecal calprotectin and an enrichment of proinflammatory pathways in a subset of adolescents post-VSG. We show that in some adolescents, microbiome changes post-VSG may have inflammatory potential, which may be of importance considering the increased incidence of inflammatory bowel disease post-VSG.
Collapse
Affiliation(s)
- Cynthia O. Akagbosu
- Department of Gastroenterology, Weill Cornell Medicine, New York, New York, USA
| | - Kathryn E. McCauley
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sivaranjani Namasivayam
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Hector N. Romero-Soto
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Wade O’Brien
- Dartmouth Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire, USA
| | - Mickayla Bacorn
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Eric Bohrnsen
- Research Technologies Branch, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Benjamin Schwarz
- Research Technologies Branch, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, Division of Intramural Research, Rocky Mountain Laboratories, National Institutes of Health, Hamilton, Montana, USA
| | - Shreni Mistry
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew S. Burns
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - P. Juliana Perez-Chaparro
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Qing Chen
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Phoebe LaPoint
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Anal Patel
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Lauren E. Krausfeldt
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Poorani Subramanian
- Bioinformatics and Computational Biosciences Branch National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Brian A. Sellers
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Foo Cheung
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Richard Apps
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Iyadh Douagi
- NIH Center for Human Immunology, Autoimmunity, and Inflammation (CHI), Bethesda, Maryland, USA
| | - Shira Levy
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Suchitra K. Hourigan
- Clinical Microbiome Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
12
|
Hwang HG, Park JW, Lee HJ, Ko MY, Ka M, Lee YK, Choi J, In SA, Lee YE, Lee S, Kim MS, Kim JY. Akkermansia muciniphila reverses neuronal atrophy in Negr1 knockout mice with depression-like phenotypes. Gut Microbes 2025; 17:2508424. [PMID: 40388597 PMCID: PMC12091914 DOI: 10.1080/19490976.2025.2508424] [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: 01/16/2025] [Revised: 04/07/2025] [Accepted: 05/14/2025] [Indexed: 05/21/2025] Open
Abstract
Genetic predispositions can shape the gut microbiome, which in turn modulates host gene expression and impacts host physiology. The complex interplay between host genetics and the gut microbiome likely contributes to the development of neuropsychiatric disorders, yet the mechanisms behind these interactions remain largely unexplored. In this study, we investigated the gut microbiota in Negr1 knockout (KO) mice, which exhibit anxiety- and depression-like behaviors, as NEGR1 (neuronal growth regulator 1) is a cell adhesion molecule linked to neuronal development and neuropsychiatric disorders. Our findings show significant early-life alterations in the gut microbiota composition of Negr1 KO mice, most notably a marked reduction in Akkermansia spp. along with reduced dendritic arborization and spine density in the nucleus accumbens (NAc) and the dentate gyrus (DG) of the hippocampus. Remarkably, daily administration of an Akkermansia strain isolated from wild-type mice reversed the neuronal structural abnormalities and ameliorated anxiety- and depression-like behaviors in Negr1 KO mice. Transcriptomic profiling revealed upregulation of mitochondrial genome-encoded genes in the NAc and hippocampus of Negr1 KO mice, along with a predisposition toward a pro-inflammatory state in the colon of Negr1 KO mice. The Akkermansia supplementation downregulated these mitochondrial genes in the NAc and hippocampus and upregulated genes involved in T cell activation and immune homeostasis in the colon. These findings demonstrate a novel gene-microbiome interaction in the pathophysiology of Negr1 KO mice, positioning Akkermansia spp. as a key mediator that improves neuronal atrophy and modulates anxiety- and depression-like behaviors. Our study provides compelling evidence for bidirectional interactions between host genetics and the gut microbiome in modulating neuropsychiatric phenotypes, offering new insights for addressing genetically influenced mental disorders.
Collapse
Affiliation(s)
- Hee-Gon Hwang
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Ji-Woo Park
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Hyo-Jin Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Moon Yi Ko
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Minhan Ka
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, Republic of Korea
| | - Yun Kyung Lee
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, Republic of Korea
| | - Jaeyoon Choi
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science, Soonchunhyang University, Cheonan, Republic of Korea
| | - Su-A In
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Ye-Eun Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Soojin Lee
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Min-Soo Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| | - Jeong-Yoon Kim
- Department of Microbiology and Molecular Biology, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
13
|
Singh S, Abu Y, Antoine D, Gomez D, Tao J, Truitt B, Roy S. Probiotic supplementation mitigates sex-dependent nociceptive changes and gut dysbiosis induced by prenatal opioid exposure. Gut Microbes 2025; 17:2464942. [PMID: 39950489 PMCID: PMC11834462 DOI: 10.1080/19490976.2025.2464942] [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: 06/20/2024] [Revised: 11/21/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
The gut microbiome has emerged as a promising target for modulating adverse effects of opioid exposure due to its significant role in health and disease. Opioid use disorder (OUD) has become increasingly prevalent, specifically in women of reproductive age, contributing to an increased incidence of offspring exposed to opioids in utero. Recent studies have shown that prenatal opioid exposure (POE) is associated with notable changes to the maternal gut microbiome, with subsequent implications for the offspring's microbiome and other adverse outcomes. However, the role of the gut microbiome in mediating sex-based differences in pain sensitivity has not yet been investigated. In this study, both male and female C57BL/6 offspring were used to determine sex-based differences in nociception and gut microbial composition as a result of POE. Our data reveals significant sex-based differences in offspring prenatally exposed to opioids. The gut microbiome of opioid-exposed females showed an enrichment of commensal bacteria including Lactobacillus compared to opioid-exposed males. Additionally, POE females demonstrated decreased nociceptive sensitivity, while males demonstrated increased nociceptive sensitivity. RNA sequencing of the prefrontal cortex showed sex-based differences in several canonical pathways, including an increase in the opioid signaling pathway of opioid-exposed females, which was not observed in males. Microbiome modification via maternal probiotic supplementation attenuated sex-based differences throughout the early stages of life. Together, our study provides further insight on sex-based differences arising from POE and highlights the pivotal role of the gut microbiome as a modifiable target for mitigating its negative effects.
Collapse
Affiliation(s)
- Salma Singh
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
| | - Yaa Abu
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
| | - Danielle Antoine
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
- Department of Neuroscience, School of Medicine, University of Miami Miller, Miami, USA
| | - Daniel Gomez
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
| | - Junyi Tao
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
| | - Bridget Truitt
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
- Department of Neuroscience, School of Medicine, University of Miami Miller, Miami, USA
| | - Sabita Roy
- Department of Surgery, School of Medicine, University of Miami Miller, Miami, USA
| |
Collapse
|
14
|
Valentin C, Brito Rodrigues P, Verce M, Delbauve S, La Palombara L, Demaret F, Allard J, Salmon I, Cani PD, Köhler A, Everard A, Flamand V. Maternal probiotic exposure enhances CD8 T cell protective neonatal immunity and modulates offspring metabolome to control influenza virus infection. Gut Microbes 2025; 17:2442526. [PMID: 39710590 DOI: 10.1080/19490976.2024.2442526] [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: 09/10/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024] Open
Abstract
Maternal gut microbiota composition contributes to the status of the neonatal immune system and could influence the early life higher susceptibility to viral respiratory infections. Using a novel protocol of murine maternal probiotic supplementation, we report that perinatal exposure to Lacticaseibacillus rhamnosus (L.rh) or Bifidobacterium animalis subsp. lactis (B.lac) increases the influenza A/PR8 virus (IAV) clearance in neonates. Following either supplementation, type 1 conventional dendritic cells (cDC1) were amplified in the lymph nodes leading to an enhanced IAV antigen-experienced IFN-γ producing effector CD8 T cells in neonates and IAV-specific resident memory CD8 T cells in adulthood. This was compatible with a higher protection of the offspring upon a secondary infection. Interestingly, only mice born to L.rh supplemented mothers further displayed an increased activation of IFN-γ producing virtual memory CD8 T cells and a production of IL-10 by CD4 and CD8 T cells that could explain a better control of the lung damages upon infection. In the offspring and the mothers, no disturbance of the gut microbiota was observed but, as analyzed through an untargeted metabolomic approach, both exposures modified neonatal plasma metabolites. Among them, we further demonstrated that genistein and 3-(3-hydroxyphenyl)propionic acid recapitulate viral clearance or cDC1 activation in neonates exposed to IAV. We conclude that maternal L.rh or B.lac supplementation confers the neonates specific metabolomic modulations with a better CD8 T cell-mediated immune protection against IAV infection.
Collapse
Affiliation(s)
- Clara Valentin
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| | - Patricia Brito Rodrigues
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium
| | - Marko Verce
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium
| | - Sandrine Delbauve
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| | - Léa La Palombara
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| | - Florine Demaret
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| | - Justine Allard
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles, Gosselies, Belgium
| | - Isabelle Salmon
- DIAPath, Center for Microscopy and Molecular Imaging, Université Libre de Bruxelles, Gosselies, Belgium
| | - Patrice D Cani
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Arnaud Köhler
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) Department, WEL Research Institute, Wavre, Belgium
| | - Véronique Flamand
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| |
Collapse
|
15
|
Castonguay-Paradis S, Demers-Potvin É, Rochefort G, Lacroix S, Perron J, Martin C, Flamand N, Raymond F, Di Marzo V, Veilleux A. Seasonal variations in circulating endocannabinoidome mediators and gut microbiota composition in humans. Gut Microbes 2025; 17:2476563. [PMID: 40111342 PMCID: PMC11926903 DOI: 10.1080/19490976.2025.2476563] [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: 05/27/2024] [Revised: 01/20/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND The human gut microbiome-endocannabinoidome axis is crucial for several homeostatic processes, including inflammation and energy metabolism, and is influenced by many endogenous and exogenous factors, such as dietary habits. Changes in the gut microbiome in response to seasonal variations were previously reported and tentatively attributed to shifts in dietary patterns. However, there is a need for longitudinal studies in industrialized populations to comprehensively explore seasonal variations independently of lifestyle confounding factors. OBJECTIVE To investigate the longitudinal effects of seasonal variations on the composition of the gut microbiome and the circulating levels of endocannabinoidome mediators in humans, while elucidating the contributing factors underlying these changes. METHODS Plasma and fecal samples were collected at the end of both the winter and summer in a longitudinal cohort of 48 individuals living in Québec City (Canada). Dietary habits, medical history, fecal microbiota taxonomic composition and plasma levels of circulating N‑acyl‑ethanolamines (NAEs) and 2‑monoacyl-glycerols (2‑MAGs) were obtained at each time point. RESULTS Lower circulating levels of most NAEs were observed at the end of summer. These changes were accompanied by a reduction in the relative abundance of the Bifidobacteriaceae and Lachnospiraceae families, along with an increase in the abundance of the Bacteroidaceae and Ruminococcaceae families. These seasonal variations were not associated with concurrent changes in adiposity parameters, dietary intakes, physical activity habits, or vitamin D status. Importantly, the magnitude of the shift in gut microbiota composition from winter to summer was found to be associated with the seasonal variations in circulating endocannabinoidome (eCBome) mediators. CONCLUSION This study identified specific seasonal changes in gut microbiota composition and circulating levels of several NAEs, which were not associated with vitamin D status and lifestyle habits. It underscores the importance of the gut microbiota-endocannabinoidome axis in the pathophysiology of seasonal changes, and of considering seasons in clinical trials on these systems.
Collapse
Affiliation(s)
- Sophie Castonguay-Paradis
- Centre Nutrition, santé et société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Québec, QC, Canada
- École de nutrition, Faculté des sciences de l’agriculture et de l’alimentation (FSAA), Université Laval, Québec, QC, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval ,Québec, Qc, Canada
| | - Élisabeth Demers-Potvin
- Centre Nutrition, santé et société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Québec, QC, Canada
- École de nutrition, Faculté des sciences de l’agriculture et de l’alimentation (FSAA), Université Laval, Québec, QC, Canada
| | - Gabrielle Rochefort
- Centre Nutrition, santé et société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Québec, QC, Canada
- École de nutrition, Faculté des sciences de l’agriculture et de l’alimentation (FSAA), Université Laval, Québec, QC, Canada
| | - Sébastien Lacroix
- Centre Nutrition, santé et société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Québec, QC, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval ,Québec, Qc, Canada
| | - Julie Perron
- Centre Nutrition, santé et société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Québec, QC, Canada
- École de nutrition, Faculté des sciences de l’agriculture et de l’alimentation (FSAA), Université Laval, Québec, QC, Canada
| | - Cyril Martin
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval ,Québec, Qc, Canada
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), Québec, QC, Canada
| | - Nicolas Flamand
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval ,Québec, Qc, Canada
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), Québec, QC, Canada
- Département de médecine, Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Frédéric Raymond
- Centre Nutrition, santé et société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Québec, QC, Canada
- École de nutrition, Faculté des sciences de l’agriculture et de l’alimentation (FSAA), Université Laval, Québec, QC, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval ,Québec, Qc, Canada
| | - Vincenzo Di Marzo
- Centre Nutrition, santé et société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Québec, QC, Canada
- École de nutrition, Faculté des sciences de l’agriculture et de l’alimentation (FSAA), Université Laval, Québec, QC, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval ,Québec, Qc, Canada
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), Québec, QC, Canada
- Département de médecine, Faculté de médecine, Université Laval, Québec, QC, Canada
- Joint International Unit on Chemical and Biomolecular Research on the Microbiome and its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu) between Université Laval and Consiglio Nazionale delle Ricerche, Institute of Biomolecular Chemistry, Italy
| | - Alain Veilleux
- Centre Nutrition, santé et société (NUTRISS), Institut sur la nutrition et les aliments fonctionnels (INAF), Québec, QC, Canada
- École de nutrition, Faculté des sciences de l’agriculture et de l’alimentation (FSAA), Université Laval, Québec, QC, Canada
- Canada Research Excellence Chair in the Microbiome-Endocannabinoidome mediators Axis in Metabolic Health (CERC-MEND), Université Laval ,Québec, Qc, Canada
| |
Collapse
|
16
|
Moore M, Whittington HD, Knickmeyer R, Azcarate-Peril MA, Bruno-Bárcena JM. Non-stochastic reassembly of a metabolically cohesive gut consortium shaped by N-acetyl-lactosamine-enriched fibers. Gut Microbes 2025; 17:2440120. [PMID: 39695352 DOI: 10.1080/19490976.2024.2440120] [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: 04/30/2024] [Revised: 10/15/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Diet is one of the main factors shaping the human microbiome, yet our understanding of how specific dietary components influence microbial consortia assembly and subsequent stability in response to press disturbances - such as increasing resource availability (feeding rate) - is still incomplete. This study explores the reproducible re-assembly, metabolic interplay, and compositional stability within microbial consortia derived from pooled stool samples of three healthy infants. Using a single-step packed-bed reactor (PBR) system, we assessed the reassembly and metabolic output of consortia exposed to lactose, glucose, galacto-oligosaccharides (GOS), and humanized GOS (hGOS). Our findings reveal that complex carbohydrates, especially those containing low inclusion (~1.25 gL-1) components present in human milk, such as N-acetyl-lactosamine (LacNAc), promote taxonomic, and metabolic stability under varying feeding rates, as shown by diversity metrics and network analysis. Targeted metabolomics highlighted distinct metabolic responses to different carbohydrates: GOS was linked to increased lactate, lactose to propionate, sucrose to butyrate, and CO2, and the introduction of bile salts with GOS or hGOS resulted in butyrate reduction and increased hydrogen production. This study validates the use of single-step PBRs for reliably studying microbial consortium stability and functionality in response to nutritional press disturbances, offering insights into the dietary modulation of microbial consortia and their ecological dynamics.
Collapse
Affiliation(s)
- Madison Moore
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Hunter D Whittington
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| | - Rebecca Knickmeyer
- Department of Psychiatry, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - M Andrea Azcarate-Peril
- Department of Medicine, Division of Gastroenterology and Hepatology, and UNC Microbiome Core, Center for Gastrointestinal Biology and Disease (CGIBD), School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jose M Bruno-Bárcena
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
17
|
Charlesson B, Jones J, Abbiss C, Peeling P, Watts S, Christophersen C. Training load influences gut microbiome of highly trained rowing athletes. J Int Soc Sports Nutr 2025; 22:2507952. [PMID: 40400144 PMCID: PMC12100958 DOI: 10.1080/15502783.2025.2507952] [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: 02/07/2025] [Accepted: 05/08/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Despite the importance of the gut microbiome on physical performance and health, little is known on the impact of training on an athlete's gut health. OBJECTIVE This study investigates the effect of training load on markers of gut health. METHODS Whole stool (24 h) samples were collected from 23 highly trained rowers (mean ± SD; age 19.2 ± 1.1 y; weight 80.1 ± 11.4 kg; height 1.83 ± 0.09 m) following periods of high (HT) and low training load (LT). The microbiome and short-chain fatty acid concentrations were characterized from the whole stool samples. Three-day weighted food records were used to determine diet quality (ADIcore), macronutrient, and fiber intakes during HT and LT. RESULTS By design, training duration (147%) and intensity (130%) were greater during (HT), compared with (LT) (p < 0.001). Carbohydrate, fat, protein, and fiber intake remained stable, but ADIcore was higher in HT (55 ± 10) compared with LT (49 ± 9; t(15) = 2.78, p = 0.014; CI: 1.34 to 10.155). Stool frequency (1.11 ± 0.47 vs 0.67 ± 0.76; p = 0.007) was lower in HT compared with LT, and a greater number of participants were unable to produce a stool sample during LT (8% vs 47%). Short chain fatty acid (SCFA), propionic (120.64 ± 30.06 mm vs 91.35 ± 34.91 mm; p = 0.007), and butyric acid (104.76 ± 50.02 vs 64.23 ± 22.05 mm, p = 0.003) concentrations were lower in HT compared with LT. Alpha diversity, Shannon-Wiener diversity index (3.43 ± 0.37 vs 3.67 ± 0.34, p = 0.09) was lower in HT than LT. The abundance of the dominant Bacteroidia was greater at HT compared to LT and ratio of firmicutes to Bacteroidota (n = 16, 1.31 ± 1.19 vs 4.29 ± 3.88, t(15) = -3.44, p = 0.04, CI = -4.82 to -1.13) was lower in HT compared to LT. CONCLUSION Results of this study indicate that gut microbiome, SCFA concentrations, stool frequency, and diet quality vary between periods of high and low training load in athletes. The relationship between these factors and impact of such changes in gut health is currently unclear and warrants further investigation.
Collapse
Affiliation(s)
- B. Charlesson
- Edith Cowan University, School of Medical and Health Science, Perth, Australia
- Western Australian Institute of Sport, Perth, Australia
| | - J. Jones
- Edith Cowan University, School of Medical and Health Science, Perth, Australia
| | - C. Abbiss
- Edith Cowan University, School of Medical and Health Science, Perth, Australia
| | - P. Peeling
- Western Australian Institute of Sport, Perth, Australia
- University of Western Australia, School of Human Sciences, Perth, Australia
| | - S. Watts
- Western Australian Institute of Sport, Perth, Australia
- University of Western Australia, School of Human Sciences, Perth, Australia
| | - C.T. Christophersen
- Edith Cowan University, School of Medical and Health Science, Perth, Australia
| |
Collapse
|
18
|
Mitra D, Armijo GK, Ober EH, Baker SM, Turner HC, Broustas CG. MIIST305 mitigates gastrointestinal acute radiation syndrome injury and ameliorates radiation-induced gut microbiome dysbiosis. Gut Microbes 2025; 17:2458189. [PMID: 39930324 PMCID: PMC11817531 DOI: 10.1080/19490976.2025.2458189] [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: 11/11/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 02/14/2025] Open
Abstract
High-dose radiation exposure results in gastrointestinal (GI) acute radiation syndrome identified by the destruction of mucosal layer, intestinal growth barrier dysfunction, and aberrant inflammatory responses. Further, radiation causes gut microbiome dysbiosis characterized by diminished microbial diversity, mostly commensal bacteria, and the spread of bacterial pathogens that trigger the recruitment of immune cells and the production of pro-inflammatory factors that lead to further GI tissue damage. Currently, there are no U.S. Food and Drug Administration (FDA) approved countermeasures that can treat radiation-induced GI injuries. To meet this critical need, Synedgen Inc. has developed a glycopolymer radiomitigator (MIIST305) that is specifically targeted to the GI tract, which acts by intercalating into the mucus layer and the glycocalyx of intestinal epithelial cells that could potentially ameliorate the deleterious effects of radiation. Male C57BL/6J adult mice were exposed to 13 Gy partial body X-irradiation with 5% bone marrow shielding and MIIST305 was administered on days 1, 3, and 5 post-irradiation. Approximately 85% of the animals survived the irradiation exposure and were apparently healthy until the end of the 30-day study period. In contrast, no control, Vehicle-treated animals survived past day 10 at this radiation dose. We show that MIIST305 improved intestinal epithelial barrier function and suppressed systemic inflammatory responses mediated by radiation-induced pro-inflammatory cytokines. Taxonomic profiling and community structure of the fecal and colonic mucosa microbiota demonstrated that MIIST305 treatment increased microbial diversity and restored abundance of beneficial commensal bacteria, including Lactobacillus and Bifidobacterium genera while suppressing potentially pathogenic bacteria Enterococcus and Staphylococcus compared with Vehicle-treated animals. In summary, MIIST305 is a novel GI-targeted therapeutic that greatly enhances survival in mice exposed to lethal radiation and protects the GI tract from injury by restoring a balanced gut microbiota and reducing pro-inflammatory responses. Further development of this drug as an FDA-approved medical countermeasure is of critical importance.
Collapse
Affiliation(s)
- Debmalya Mitra
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Gabriel K. Armijo
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Elizabeth H. Ober
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | | | - Helen C. Turner
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Constantinos G. Broustas
- Center for Radiological Research, Columbia University Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
19
|
Uriot O, Deschamps C, Scanzi J, Brun M, Kerckhove N, Dualé C, Fournier E, Durif C, Denis S, Dapoigny M, Langella P, Alric M, Etienne-Mesmin L, Stéphanie BD. Gut microbial dysbiosis associated to diarrheic irritable bowel syndrome can be efficiently simulated in the Mucosal ARtificial COLon (M-ARCOL). Bioengineered 2025; 16:2458362. [PMID: 39902883 PMCID: PMC11796540 DOI: 10.1080/21655979.2025.2458362] [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: 07/08/2024] [Revised: 01/08/2025] [Accepted: 01/16/2025] [Indexed: 02/06/2025] Open
Abstract
Irritable bowel syndrome (IBS) is a common chronic gastrointestinal disorder, with diarrhea-predominant IBS (IBS-D) as the most frequent subtype. The implication of gut microbiota in the disease's etiology is not fully understood. In vitro gut systems can offer a great alternative to in vivo assays in preclinical studies, but no model reproducing IBS-related dysbiotic microbiota has been developed. Thanks to a large literature review, a new Mucosal ARtifical COLon (M-ARCOL) adapted to IBS-D physicochemical and nutritional conditions was set-up. To validate the model and further exploit its potential in a mechanistic study, in vitro fermentations were performed using bioreactors inoculated with stools from healthy individuals (n = 4) or IBS-D patients (n = 4), when the M-ARCOL was set-up under healthy or IBS-D conditions. Setting IBS-D parameters in M-ARCOL inoculated with IBS-D stools maintained the key microbial features associated to the disease in vivo, validating the new system. In particular, compared to the healthy control, the IBS-D model was characterized by a decreased bacterial diversity, together with a lower abundance of Rikenellaceae and Prevotellaceae, but a higher level of Proteobacteria and Akkermansiaceae. Of interest, applying IBS-D parameters to healthy stools was not sufficient to trigger IBS-D dysbiosis and applying healthy parameters to IBS-D stools was not enough to restore microbial balance. This validated IBS-D colonic model can be used as a robust in vitro platform for studies focusing on gut microbes in the absence of the host, as well as for testing food and microbiota-related interventions aimed at personalized restoration of gut microbiota eubiosis.
Collapse
Affiliation(s)
- Ophélie Uriot
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne – INRAE, Clermont-Ferrand, Puy-de-Dôme,France
| | - Charlotte Deschamps
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne – INRAE, Clermont-Ferrand, Puy-de-Dôme,France
| | - Julien Scanzi
- UMR INSERM 1107 NEURO-DOL, Université Clermont Auvergne, Clermont-Ferrand, Puy-de-Dôme,France
- Service de Gastroentérologie, Centre Hospitalo-Universitaire, Clermont-Ferrand, Puy-de-Dôme,France
- Service de Gastroentérologie, Centre Hospitalier de Thiers, Thiers, Puy-de-Dôme, France
| | - Morgane Brun
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne – INRAE, Clermont-Ferrand, Puy-de-Dôme,France
| | - Nicolas Kerckhove
- UMR INSERM 1107 NEURO-DOL, Université Clermont Auvergne, Clermont-Ferrand, Puy-de-Dôme,France
- Service de Pharmacologie médicale, Centre Hospitalo-Universitaire, Clermont-Ferrand, Puy-de-Dôme,France
| | - Christian Dualé
- CIC INSERM 1405, Centre Hospitalo-Universitaire, Clermont-Ferrand, Puy-de-Dôme,France
| | - Elora Fournier
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne – INRAE, Clermont-Ferrand, Puy-de-Dôme,France
| | - Claude Durif
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne – INRAE, Clermont-Ferrand, Puy-de-Dôme,France
| | - Sylvain Denis
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne – INRAE, Clermont-Ferrand, Puy-de-Dôme,France
| | - Michel Dapoigny
- UMR INSERM 1107 NEURO-DOL, Université Clermont Auvergne, Clermont-Ferrand, Puy-de-Dôme,France
- Service de Gastroentérologie, Centre Hospitalo-Universitaire, Clermont-Ferrand, Puy-de-Dôme,France
| | - Philippe Langella
- Micalis, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, Yvelines,France
| | - Monique Alric
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne – INRAE, Clermont-Ferrand, Puy-de-Dôme,France
| | - Lucie Etienne-Mesmin
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne – INRAE, Clermont-Ferrand, Puy-de-Dôme,France
| | - Blanquet-Diot Stéphanie
- UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, Université Clermont Auvergne – INRAE, Clermont-Ferrand, Puy-de-Dôme,France
| |
Collapse
|
20
|
Prince N, Peralta Marzal LN, Roussin L, Monnoye M, Philippe C, Maximin E, Ahmed S, Salenius K, Lin J, Autio R, Adolfs Y, Pasterkamp RJ, Garssen J, Naudon L, Rabot S, Kraneveld AD, Perez-Pardo P. Mouse strain-specific responses along the gut-brain axis upon fecal microbiota transplantation from children with autism. Gut Microbes 2025; 17:2447822. [PMID: 39773319 PMCID: PMC11730631 DOI: 10.1080/19490976.2024.2447822] [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: 07/18/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025] Open
Abstract
Several factors are linked to the pathophysiology of autism spectrum disorders (ASD); however, the molecular mechanisms of the condition remain unknown. As intestinal problems and gut microbiota dysbiosis are associated with ASD development and severity, recent studies have focused on elucidating the microbiota-gut-brain axis' involvement. This study aims to explore mechanisms through which gut microbiota might influence ASD. Briefly, we depleted the microbiota of conventional male BALB/cAnNCrl (Balb/c) and C57BL/6J (BL/6) mice prior to human fecal microbiota transplantation (hFMT) with samples from children with ASD or their neurotypical siblings. We found mouse strain-specific responses to ASD hFMT. Notably, Balb/c mice exhibit decreased exploratory and social behavior, and show evidence of intestinal, systemic, and central inflammation accompanied with metabolic shifts. BL/6 mice show less changes after hFMT. Our results reveal that gut microbiota alone induce changes in ASD-like behavior, and highlight the importance of mouse strain selection when investigating multifactorial conditions like ASD.
Collapse
Affiliation(s)
- Naika Prince
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Lucia N. Peralta Marzal
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Léa Roussin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Magali Monnoye
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Catherine Philippe
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Elise Maximin
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Sabbir Ahmed
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Karoliina Salenius
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Centre, Tampere, Finland
| | - Jake Lin
- Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Reija Autio
- Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Youri Adolfs
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - R. Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Danone Nutricia Research, Utrecht, Netherlands
| | - Laurent Naudon
- Université Paris-Saclay, INRAE, AgroParisTech, CNRS, Micalis Institute, Jouy-en-Josas, France
| | - Sylvie Rabot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Aletta D. Kraneveld
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Department of Neuroscience, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Paula Perez-Pardo
- Division of Pharmacology, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
21
|
Budzinski L, Kang GU, Riedel R, Sempert T, Lietz L, Maier R, Büttner J, Bochow B, Tordai MT, Shah A, Abbas A, Momtaz T, Krause JL, Kempkens R, Lehman K, Heinz GA, Benken AE, Bartsch S, Necke K, Hoffmann U, Mashreghi MF, Biesen R, Kallinich T, Alexander T, Jessen B, Weidinger C, Siegmund B, Radbruch A, Schirbel A, Moser B, Chang HD. Single-cell microbiota phenotyping reveals distinct disease and therapy-associated signatures in Crohn's disease. Gut Microbes 2025; 17:2452250. [PMID: 39815413 PMCID: PMC11740678 DOI: 10.1080/19490976.2025.2452250] [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: 08/29/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025] Open
Abstract
IgA-coated fractions of the intestinal microbiota of Crohn's disease (CD) patients have been shown to contain taxa that hallmark the compositional dysbiosis in CD microbiomes. However, the correlation between other cellular properties of intestinal bacteria and disease has not been explored further, especially for features that are not directly driven by the host immune-system, e.g. the expression of surface sugars by bacteria. By sorting and sequencing IgA-coated and lectin-stained fractions from CD patients microbiota and healthy controls, we found that lectin-stained bacteria were distinct from IgA-coated bacteria, but still displayed specific differences between CD and healthy controls. To exploit the discriminatory potential of both, immunoglobulin coated bacteria and the altered surface sugar expression of bacteria in CD, we developed a multiplexed single cell-based analysis approach for intestinal microbiota. By multi-parameter microbiota flow cytometry (mMFC) we characterized the intestinal microbiota of 55 CD patients and 44 healthy controls for 11-parameters in total, comprising host-immunoglobulin coating and the presence of distinct surface sugar moieties. The data were analyzed by machine-learning to assess disease-specific marker patterns in the microbiota phenotype. mMFC captured detailed characteristics of CD microbiota and identified patterns to classify CD patients. In addition, we identified phenotypic signatures in the CD microbiota which not only reflected remission after 6 weeks of anti-TNF treatment, but were also able to predict remission before the start of an adalimumab treatment course in a pilot study. We here present the proof-of-concept demonstrating that multi-parameter single-cell bacterial phenotyping by mMFC could be a novel tool with high translational potential to expand current microbiome investigations by phenotyping of bacteria to identify disease- and therapy-associated cellular alterations and to reveal novel target properties of bacteria for functional assays and therapeutic approaches.
Collapse
Affiliation(s)
- Lisa Budzinski
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department for Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Gi-Ung Kang
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - René Riedel
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Bioinformatics and Computational Biology, Department of Cardiology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Toni Sempert
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Leonie Lietz
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department for Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - René Maier
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Janine Büttner
- Department of Hepatology and Gastroenterology, Campus Charité Mitte, Charité, Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bettina Bochow
- Department of Hepatology and Gastroenterology, Campus Charité Mitte, Charité, Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcell T. Tordai
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Aayushi Shah
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department for Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Amro Abbas
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Tanisha Momtaz
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- School of Pharmacy, BRAC University, Dhaka, Bangladesh
| | - Jannike L. Krause
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Robin Kempkens
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Katrin Lehman
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Gitta A. Heinz
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Anne E. Benken
- Department of Rheumatology, Campus Charité Mitte, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Stefanie Bartsch
- Department of Paediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Campus Virchow, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Kathleen Necke
- Department of Paediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Campus Virchow, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Ute Hoffmann
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Mir-Farzin Mashreghi
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Robert Biesen
- Department of Rheumatology, Campus Charité Mitte, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tilmann Kallinich
- Department of Paediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité Campus Virchow, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Tobias Alexander
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department of Rheumatology, Campus Charité Mitte, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bosse Jessen
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Carl Weidinger
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Campus Benjamin Franklin, Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- BIH Charité Clinician Scientist Program
| | - Andreas Radbruch
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
| | - Anja Schirbel
- Department of Hepatology and Gastroenterology, Campus Charité Mitte, Charité, Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Benjamin Moser
- Department of Hepatology and Gastroenterology, Campus Charité Mitte, Charité, Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- DRK Kliniken Berlin, Clinic for internal medicine – Gastroenterology, Haematology and Oncology, Nephrology, Centre for chronic gastrointestinal inflammations, Berlin, Germany
| | - Hyun-Dong Chang
- German Rheumatology Research Centre Berlin – A Leibniz Institute, Berlin, Germany
- Department for Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| |
Collapse
|
22
|
Kordahi MC, Daniel N, Gewirtz AT, Chassaing B. Mucus-penetrating microbiota drive chronic low-grade intestinal inflammation and metabolic dysregulation. Gut Microbes 2025; 17:2455790. [PMID: 39865067 PMCID: PMC11776472 DOI: 10.1080/19490976.2025.2455790] [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: 01/19/2024] [Revised: 12/20/2024] [Accepted: 01/14/2025] [Indexed: 01/28/2025] Open
Abstract
Metabolic syndrome is, in humans, associated with alterations in the composition and localization of the intestinal microbiota, including encroachment of bacteria within the colon's inner mucus layer. Possible promoters of these events include dietary emulsifiers, such as carboxymethylcellulose (CMC) and polysorbate-80 (P80), which, in mice, result in altered microbiota composition, encroachment, low-grade inflammation and metabolic syndrome. While assessments of gut microbiota composition have largely focused on fecal/luminal samples, we hypothesize an outsized role for changes in mucus microbiota in driving low-grade inflammation and its consequences. In support of this notion, we herein report that both CMC and P80 led to stark changes in the mucus microbiome, markedly distinct from those observed in feces. Moreover, transfer of mucus microbiota from CMC- and P80-fed mice to germfree mice resulted in microbiota encroachment, low-grade inflammation, and various features of metabolic syndrome. Thus, we conclude that mucus-associated bacteria are pivotal determinants of intestinal inflammatory tone and host metabolism.
Collapse
Affiliation(s)
- Melissa C. Kordahi
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, CNRS UMR6047, Paris, France
- Mucosal microbiota in chronic inflammatory diseases, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris, France
| | - Noëmie Daniel
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, CNRS UMR6047, Paris, France
- Mucosal microbiota in chronic inflammatory diseases, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris, France
| | - Andrew T. Gewirtz
- Institute for Biomedical Sciences, Centre for Inflammation, Immunity and Infection, Digestive Disease Research Group, Georgia State University, Atlanta, GA, USA
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, CNRS UMR6047, Paris, France
- Mucosal microbiota in chronic inflammatory diseases, INSERM U1016, CNRS UMR8104, Université Paris Cité, Paris, France
- CHRU Nancy, IHU Infiny, Nancy, France
| |
Collapse
|
23
|
Martinez-Medina JN, Ghazisaeedi F, Kramer C, Ziegler JF, McParland V, Mönch PW, Siegmund B, Jarquín-Díaz VH, Fulde M, Forslund-Startceva SK. Mucosal washes are useful for sampling intestinal mucus-associated microbiota despite low biomass. Gut Microbes 2025; 17:2464296. [PMID: 39980334 PMCID: PMC11849919 DOI: 10.1080/19490976.2025.2464296] [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: 12/26/2023] [Revised: 10/24/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
Understanding the dynamic relationship between mucus-associated microbiota and host health is critical. However, studies predominantly using stool samples may not accurately represent these bacterial communities. Here, we investigated the mucus-associated microbiota in the gastrointestinal tract of mice and the terminal ileum of humans using different sample types: mucosal washes, brushes, scrapings, and intestinal contents in mice and biopsies, brushes and mucosal washes in humans. We used DNA quantification and 16S rRNA amplicon sequencing to evaluate the comparability of the information yielded from the different sample types under a controlled benchmark. In mice, mucosal washes and brushes had comparative bacterial DNA and host DNA contamination than scraping samples. Similarly, in humans, washes outperformed biopsies in bacterial DNA content. Read counts and microbiota alpha diversity remained remarkably similar in mice and between brushes and washes in humans. The composition of the microbiota varied based on the subsegment and sample type in mice and sample type in humans. We conclude that washes and brushes reduce host contamination without inducing substantial compositional bias when sampling mucosal microbiota. Our findings suggest that mucosal washes and brushes are a viable alternative to biopsies in humans and scrapings in mice, thereby improving the transferability of results across hosts. Our study highlights the importance of focusing on mucus-associated microbiota to better capture host-microbiome interactions at their closer interface.
Collapse
Affiliation(s)
- Jennifer N. Martinez-Medina
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
| | - Fereshteh Ghazisaeedi
- Institute of Microbiology and Epizootics, School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
| | - Catharina Kramer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Jörn F Ziegler
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
| | - Victoria McParland
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
| | - Paul W. Mönch
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Britta Siegmund
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Víctor Hugo Jarquín-Díaz
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
| | - Marcus Fulde
- Institute of Microbiology and Epizootics, School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
| | - Sofia K. Forslund-Startceva
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Structural and Computational Biology, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
24
|
Mörkl S, Narrath M, Schlotmann D, Sallmutter MT, Putz J, Lang J, Brandstätter A, Pilz R, Karl Lackner H, Goswami N, Steuber B, Tatzer J, Lackner S, Holasek S, Painold A, Jauk E, Wenninger J, Horvath A, Spicher N, Barth A, Butler MI, Wagner-Skacel J. Multi-species probiotic supplement enhances vagal nerve function - results of a randomized controlled trial in patients with depression and healthy controls. Gut Microbes 2025; 17:2492377. [PMID: 40298641 PMCID: PMC12045568 DOI: 10.1080/19490976.2025.2492377] [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: 10/03/2024] [Revised: 03/04/2025] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Major depression (MD) significantly impacts individual well-being and society. The vagus nerve plays a pivotal role in the gut-brain axis, facilitating bidirectional communication between these systems. Recent meta-analyses suggest potential antidepressant effects of probiotics, although their mechanisms remain unclear. This study aimed to assess the impact of a multi-species probiotic (OMNi-BiOTiC® STRESS Repair) on vagus nerve function in 43 MD patients and 43 healthy controls (HC). Participants received either probiotics or placebo twice daily. Serum and stool samples were collected at baseline, 7 days, 28 days, and 3 months. Vagus nerve (VN) function was evaluated using 24-hour electrocardiography (ECG) for heart rate variability (HRV), alongside stool microbiome analysis via 16S rRNA sequencing. After 3 months, MD patients receiving probiotics demonstrated significantly improved morning VN function compared to HC. MD participants who were in the probiotic group showed a significant increase in Christensellales, particularly Akkermansia muciniphila along with improved sleep parameters (use of sleep medication, sleep latency) as measured by the Pittsburgh Sleep Quality Inventory (PSI). This study highlights potential physiological benefits of probiotics in MD, potentially mediated through VN stimulation. Understanding these mechanisms could lead to novel therapeutic approaches for MD management.
Collapse
Affiliation(s)
- Sabrina Mörkl
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Martin Narrath
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Daria Schlotmann
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Marie-Therese Sallmutter
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Julia Putz
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Julia Lang
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Brandstätter
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Rene Pilz
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Helmut Karl Lackner
- Division of Physiology und Pathophysiology, Medical University of Graz, Graz, Austria
| | - Nandu Goswami
- Division of Physiology und Pathophysiology, Medical University of Graz, Graz, Austria
- Gravitational Physiology and Medicine Research Unit, Division of Physiology und Pathophysiology, Medical University of Graz, Graz, Austria
- Center for Space and Aviation Health, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Bianca Steuber
- Division of Physiology und Pathophysiology, Medical University of Graz, Graz, Austria
- Gravitational Physiology and Medicine Research Unit, Division of Physiology und Pathophysiology, Medical University of Graz, Graz, Austria
| | - Jasmin Tatzer
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Sonja Lackner
- Division of Immunology, Medical University of Graz, Graz, Austria
| | - Sandra Holasek
- Division of Immunology, Medical University of Graz, Graz, Austria
| | - Annamaria Painold
- Division of Psychiatry and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Emanuel Jauk
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Julian Wenninger
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| | - Angela Horvath
- Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria
| | - Nicolai Spicher
- Department of Medical Informatics, University Medical Center Göttingen, Göttingen, Germany
| | - Asmus Barth
- Department of Medical Informatics, University Medical Center Göttingen, Göttingen, Germany
| | - Mary I Butler
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Jolana Wagner-Skacel
- Division of Medical Psychology, Psychosomatics and Psychotherapeutic Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
25
|
Katona BW, Shukla A, Hu W, Nyul T, Dudzik C, Arvanitis A, Clay D, Dungan M, Weber M, Tu V, Hao F, Gan S, Chau L, Buchner AM, Falk GW, Jaffe DL, Ginsberg G, Palmer SN, Zhan X, Patterson AD, Bittinger K, Ni J. Microbiota and metabolite-based prediction tool for colonic polyposis with and without a known genetic driver. Gut Microbes 2025; 17:2474141. [PMID: 40069167 PMCID: PMC11913376 DOI: 10.1080/19490976.2025.2474141] [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: 10/16/2024] [Revised: 01/22/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
Despite extensive investigations into the microbiome and metabolome changes associated with colon polyps and colorectal cancer (CRC), the microbiome and metabolome profiles of individuals with colonic polyposis, including those with (Gene-pos) and without (Gene-neg) a known genetic driver, remain comparatively unexplored. Using colon biopsies, polyps, and stool from patients with Gene-pos adenomatous polyposis (N = 9), Gene-neg adenomatous polyposis (N = 18), and serrated polyposis syndrome (SPS, N = 11), we demonstrated through 16S rRNA sequencing that the mucosa-associated microbiota in individuals with colonic polyposis is representative of the microbiota associated with small polyps, and that both Gene-pos and SPS cohorts exhibit differential microbiota populations relative to Gene-neg polyposis cohorts. Furthermore, we used these differential microbiota taxa to perform linear discriminant analysis to differentiate Gene-neg subjects from Gene-pos and from SPS subjects with an accuracy of 89% and 93% respectively. Stool metabolites were quantified via 1H NMR, revealing an increase in alanine in SPS subjects relative to non-polyposis subjects, and Partial Least Squares Discriminant Analysis (PLS-DA) analysis indicated that the proportion of leucine to tyrosine in fecal samples may be predictive of SPS. Use of these microbial and metabolomic signatures may allow for better diagnostric and risk-stratification tools for colonic polyposis patients and their families as well as promote development of microbiome-targeted approaches for polyp prevention.
Collapse
Affiliation(s)
- Bryson W. Katona
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ashutosh Shukla
- Division of Digestive & Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Weiming Hu
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Thomas Nyul
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Christina Dudzik
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Alex Arvanitis
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel Clay
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michaela Dungan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Marina Weber
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Vincent Tu
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fuhua Hao
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA, USA
| | - Shuheng Gan
- Peter O’Donnell Jr. School of Public Health, Quantitative Biomedical Research Center, Center for the Genetics and Host Defense, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lillian Chau
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Anna M. Buchner
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gary W. Falk
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David L. Jaffe
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gregory Ginsberg
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Suzette N. Palmer
- Peter O’Donnell Jr. School of Public Health, Quantitative Biomedical Research Center, Center for the Genetics and Host Defense, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaowei Zhan
- Peter O’Donnell Jr. School of Public Health, Quantitative Biomedical Research Center, Center for the Genetics and Host Defense, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Josephine Ni
- Division of Digestive & Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
26
|
Zünd JN, Mujezinovic D, Reichlin M, Plüss S, Caflisch M, Robinson S, Lacroix C, Pugin B. Novel cross-feeding human gut microbes metabolizing tryptophan to indole-3-propionate. Gut Microbes 2025; 17:2501195. [PMID: 40336187 PMCID: PMC12064059 DOI: 10.1080/19490976.2025.2501195] [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: 09/02/2024] [Revised: 04/26/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Tryptophan-derived indoles produced by the gut microbiota, particularly indole-3-propionate (IPA), are key compounds associated with gastrointestinal balance and overall health. Reduced levels of IPA have been associated with inflammatory bowel disease, type 2 diabetes, and colorectal cancer. Since fiber-rich diets have been shown to promote IPA, we aimed to decipher fiber-specific effects and identify associated IPA-producing taxa in a range of healthy individuals. We cultured fecal microbiota from 16 adults with tryptophan and eight different dietary fibers and monitored community shifts by 16S rRNA gene amplicon sequencing and tryptophan-derived indoles using targeted liquid chromatography with diode array detection. The concentrations and types of indoles produced were donor-specific, with pectin strongly promoting IPA production in certain donors. IPA production was not associated with any known IPA producer but with the pectin-utilizing species Lachnospira eligens, which produced indole-3-lactate (ILA) in vitro, the IPA precursor. Supplementation of ILA in additional fecal microbiota cultures (n = 6) revealed its effective use as a substrate for IPA production. We identified a novel IPA producer, Enterocloster aldenensis, which produced IPA exclusively from ILA but not from tryptophan. Co-culture of L. eligens and E. aldenensis resulted in IPA production, providing new evidence for an ILA cross-feeding mechanism that may contribute to the IPA-promoting effects observed with pectin. Overall, we highlight the potential for targeted dietary interventions to promote beneficial gut taxa and metabolites.
Collapse
Affiliation(s)
- Janina N. Zünd
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Denisa Mujezinovic
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Markus Reichlin
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Serafina Plüss
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Marina Caflisch
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Serina Robinson
- Department of Environmental Microbiology, Eawag, Dübendorf, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| | - Benoit Pugin
- Laboratory of Food Biotechnology, Department of Health Sciences and Technology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
27
|
Jardon KM, Umanets A, Gijbels A, Trouwborst I, Hul GB, Siebelink E, Vliex LM, Bastings JJ, Argamasilla R, Chenal E, Venema K, Afman LA, Goossens GH, Blaak EE. Distinct gut microbiota and metabolome features of tissue-specific insulin resistance in overweight and obesity. Gut Microbes 2025; 17:2501185. [PMID: 40336254 PMCID: PMC12064058 DOI: 10.1080/19490976.2025.2501185] [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: 09/20/2024] [Revised: 02/24/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Insulin resistance (IR) is an early marker of cardiometabolic deterioration which may develop heterogeneously in key metabolic organs, including the liver (LIR) and skeletal muscle (MIR). This tissue-specific IR is characterized by distinct metabolic signatures, but the role of the gut microbiota in its etiology remains unclear. Here, we profiled the gut microbiota, its metabolites and the plasma metabolome in individuals with either a LIR or MIR phenotype (n = 233). We observed distinct microbial community structures LIR and MIR, and higher short-chain fatty acid (SCFA) producing bacteria, fecal SCFAs and branched-chain fatty acids and a higher postprandial plasma glucagon-like-peptide-1 response in LIR. In addition, we found variations in metabolome profiles and phenotype-specific associations between microbial taxa and functional metabolite groups. Overall, our study highlights association between gut microbiota and its metabolites composition with IR heterogeneity that can be targeted in precision-based strategies to improve cardiometabolic health. Clinicaltrials.gov registration: NCT03708419.
Collapse
Affiliation(s)
- Kelly M. Jardon
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Alexander Umanets
- Centre for Healthy Eating & Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
- Chair Group Youth Food and Health, Faculty of Science and Engineering, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Anouk Gijbels
- TiFN, Wageningen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Inez Trouwborst
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Gabby B. Hul
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Els Siebelink
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Lars M.M. Vliex
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Jacco J.A.J. Bastings
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | | | | | - Koen Venema
- Centre for Healthy Eating & Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Lydia A. Afman
- TiFN, Wageningen, The Netherlands
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Gijs H. Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Ellen E. Blaak
- TiFN, Wageningen, The Netherlands
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, The Netherlands
| |
Collapse
|
28
|
Khan S, McWhorter AR, Willson NL, Andrews DM, Underwood GJ, Moore RJ, Hao Van TT, Chousalkar KK. Vaccine protection of broilers against various doses of wild-type Salmonella Typhimurium and changes in gut microbiota. Vet Q 2025; 45:1-14. [PMID: 39721950 DOI: 10.1080/01652176.2024.2440428] [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: 08/30/2024] [Revised: 10/24/2024] [Accepted: 12/01/2024] [Indexed: 12/28/2024] Open
Abstract
This study evaluated the impact of vaccine diluents (peptone or water) on the protective effects of Salmonella Typhimurium (S. Typhimurium) vaccine. Vaccinated broilers were challenged with different doses of wild-type S. Typhimurium through dust. At the time of cull, vaccine load was highest in caeca and lowest in spleen. Wild-type S. Typhimurium was detectable after 24 hrs only in the vaccinated birds challenged with 108 CFU and positive control. S. Typhimurium load was lower in the organs of the groups challenged with 104 and 106 compared to the 108 CFU group. The caecal microbiota alpha diversity of the vaccinated or vaccinated and challenged chickens differed from the positive and negative control groups. Beta diversity of the positive control clustered separately from all other treatment groups, showing that vaccine caused minimal changes in gut microbiota structure. The vaccinated and/or wild-type challenged chickens showed significantly higher abundance of Anaerostignum, Lachnoclostridium, Intestinimonas, Colidextribacter, Monoglobus, Acetanaerobacterium and Subdoligranulum. Outcomes from this study demonstrate that the vaccine effectively protected broiler chickens from S. Typhimurium infection and helped maintain a more stable gut microbiota structure, reducing the impact of S. Typhimurium on gut health. Vaccine diluent did not affect gut microbiota composition.
Collapse
Affiliation(s)
- Samiullah Khan
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, Australia
| | - Andrea R McWhorter
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, Australia
| | - Nicky-Lee Willson
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, Australia
| | | | | | | | | | - Kapil K Chousalkar
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, Australia
| |
Collapse
|
29
|
Gustafson KL, Rodriguez TR, McAdams ZL, Coghill LM, Ericsson AC, Franklin CL. Failure of colonization following gut microbiota transfer exacerbates DSS-induced colitis. Gut Microbes 2025; 17:2447815. [PMID: 39812347 PMCID: PMC11740679 DOI: 10.1080/19490976.2024.2447815] [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: 09/25/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025] Open
Abstract
To study the impact of differing specific pathogen-free gut microbiomes (GMs) on a murine model of inflammatory bowel disease, selected GMs were transferred using embryo transfer (ET), cross-fostering (CF), and co-housing (CH). Prior work showed that the GM transfer method and the microbial composition of donor and recipient GMs can influence microbial colonization and disease phenotypes in dextran sodium sulfate-induced colitis. When a low richness GM was transferred to a recipient with a high richness GM via CH, the donor GM failed to successfully colonize, and a more severe disease phenotype resulted when compared to ET or CF, where colonization was successful. By comparing CH and gastric gavage for fecal material transfer, we isolated the microbial component of this effect and determined that differences in disease severity and survival were associated with microbial factors rather than the transfer method itself. Mice receiving a low richness GM via CH and gastric gavage exhibited greater disease severity and higher expression of pro-inflammatory immune mediators compared to those receiving a high richness GM. This study provides valuable insights into the role of GM composition and colonization in disease modulation.
Collapse
Affiliation(s)
- Kevin L. Gustafson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Trevor R. Rodriguez
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
| | - Zachary L. McAdams
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- Molecular Pathogenesis and Therapeutics Program, University of Missouri, Columbia, MO, USA
| | - Lyndon M. Coghill
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- University of Missouri Bioinformatics and Analytics Core, University of Missouri, Columbia, MO, USA
| | - Aaron C. Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- University of Missouri College of Veterinary Medicine, Columbia, MO, USA
- University of Missouri Metagenomics Center, Columbia, MO, USA
| | - Craig L. Franklin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| |
Collapse
|
30
|
Organski AC, Rajwa B, Reddivari A, Jorgensen JS, Cross TWL. Gut microbiome-driven regulation of sex hormone homeostasis: a potential neuroendocrine connection. Gut Microbes 2025; 17:2476562. [PMID: 40071861 PMCID: PMC11913384 DOI: 10.1080/19490976.2025.2476562] [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: 10/02/2024] [Revised: 12/17/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025] Open
Abstract
The gut microbiome is known to have a bidirectional relationship with sex hormone homeostasis; however, its role in mediating interactions between the primary regulatory axes of sex hormones and their productions is yet to be fully understood. We utilized both conventionally raised and gnotobiotic mouse models to investigate the regulatory role of the gut microbiome on the hypothalamic-pituitary-gonadal (HPG) axis. Male and female conventionally raised mice underwent surgical modifications as follows: (1) hormonally intact controls; (2) gonadectomized males and females; (3) gonadectomized males and females supplemented with testosterone and estrogen, respectively. Fecal samples from these mice were used to colonize sex-matched, intact, germ-free recipient mice through fecal microbiota transplant (FMT). Serum gonadotropins, gonadal sex hormones, cecal microbiota, and the serum global metabolome were assessed. FMT recipients of gonadectomized-associated microbiota showed lower circulating gonadotropin levels than recipients of intact-associated microbiota, opposite to that of FMT donors. FMT recipients of gonadectomized-associated microbiota also had greater testicular weights compared to recipients of intact-associated microbiota. The gut microbiota composition of recipient mice differed significantly based on the FMT received, with the male microbiota having a more concerted impact in response to changes in the HPG axis. Network analyses showed that multiple metabolically unrelated pathways may be involved in driving differences in serum metabolites due to sex and microbiome received in the recipient mice. In sum, our findings indicate that the gut microbiome responds to the HPG axis and subsequently modulates its feedback mechanisms. A deeper understanding of interactions between the gut microbiota and the neuroendocrine-gonadal system may contribute to the development of therapies for sexually dimorphic diseases.
Collapse
Affiliation(s)
| | - Bartek Rajwa
- Bindley Bioscience, Purdue University, West Lafayette, IN, USA
| | - Anjali Reddivari
- Department of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, USA
| | - Joan S. Jorgensen
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Tzu-Wen L. Cross
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
| |
Collapse
|
31
|
Pryor JC, Nieva C, Talley NJ, Eslick GD, Duncanson K, Burns GL, Hoedt EC, Keely S. Microbial-derived peptidases are altered in celiac disease, non-celiac gluten sensitivity, and functional dyspepsia: a systematic review and re-analysis of the duodenal microbiome. Gut Microbes 2025; 17:2500063. [PMID: 40346812 PMCID: PMC12068744 DOI: 10.1080/19490976.2025.2500063] [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: 01/15/2025] [Revised: 03/20/2025] [Accepted: 04/25/2025] [Indexed: 05/12/2025] Open
Abstract
Dietary gluten triggers symptoms in patients with gluten-related disorders (GRDs) including celiac disease (CeD), non-celiac gluten sensitivity (NCGS), and subsets of patients with functional dyspepsia (FD). The gastrointestinal microbiota is altered in these patients when compared to healthy individuals. As the microbiota is crucial for the hydrolysis of gluten, we hypothesized that the capacity of the microbiota to digest gluten is reduced in these conditions. We systematically reviewed and re-analyzed published datasets to compare gastrointestinal microbiomes of GRD patients and identify signals explaining gluten responses. A systematic search of five databases was conducted to identify studies where the microbiota of CeD, NCGS, or FD patients was analyzed by 16S rRNA amplicon or shotgun metagenomic sequencing and compared to control populations. Where available, raw duodenal microbiota sequence data were re-analyzed with a consistent bioinformatic pipeline. Thirty articles met the inclusion criteria for this systematic review. Microbiota diversity metrics were not impacted by the diseases; however, genera including Streptococcus, Neisseria, and Lactobacillus were commonly altered in GRD patients. Re-analysis of duodenal 16S rRNA data was possible for five included articles but did not identify any consistent differentially abundant taxa. Predicted functional analysis of the microbiome revealed that peptidases including aminopeptidase, proline iminopeptidase, and Xaa-Pro dipeptidase are altered in CeD, NCGS, and FD, respectively. These microbial-derived peptidases hydrolyze bonds in proline-rich gluten peptides. While the gastrointestinal microbiota in patients with GRDs differ from controls, no distinct phenotype links them. However, alterations to the predicted functional capacity of the microbiome to produce gluten-hydrolyzing enzymes suggest that inappropriate digestion of gluten by the microbiome impacts host responses to dietary gluten in these conditions. These findings have implications for therapeutic management of GRDs, as treatment with gluten-degrading enzymes or tailored probiotics could improve disease outcomes by enhancing gluten digestion into non-reactive peptides.
Collapse
Affiliation(s)
- Jennifer C. Pryor
- College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Digestive Health, The University of Newcastle, Newcastle, NSW, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Cheenie Nieva
- College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Digestive Health, The University of Newcastle, Newcastle, NSW, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Nicholas J. Talley
- College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Digestive Health, The University of Newcastle, Newcastle, NSW, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Guy D. Eslick
- College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Digestive Health, The University of Newcastle, Newcastle, NSW, Australia
| | - Kerith Duncanson
- College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Digestive Health, The University of Newcastle, Newcastle, NSW, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Grace L. Burns
- College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Digestive Health, The University of Newcastle, Newcastle, NSW, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Emily C. Hoedt
- College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Digestive Health, The University of Newcastle, Newcastle, NSW, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Simon Keely
- College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, Australia
- National Health and Medical Research Council (NHMRC), Centre of Research Excellence in Digestive Health, The University of Newcastle, Newcastle, NSW, Australia
- Immune Health Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
32
|
Rompo T, Hayashi N, Teo E, Singhla T, Kunkaew C, Sripakdee D, Nambooppha B, Wanganurakkul S, Limwibulpong K, Sangarun K, Suwongsaksri N, Suphakarn S, Chotiphutthikul C, Matsui Y, Irie T, Yoshida A, Chintapitaksakul L, Misawa N, Nonaka N, Nakao R, Tiwananthagorn S. Strongyle nematode fauna in three ruminants in upper northern Thailand. Parasitol Int 2025; 108:103057. [PMID: 40043788 DOI: 10.1016/j.parint.2025.103057] [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: 12/26/2024] [Revised: 02/18/2025] [Accepted: 03/02/2025] [Indexed: 05/17/2025]
Abstract
Gastrointestinal parasites, particularly strongyle nematodes, pose a significant threat to the health of ruminants. Due to the technical limitations of microscopic and conventional PCR-based methods, the strongyle parasite fauna has not been well studied even in common livestock animals. The present study aimed to investigate the prevalence and diversity of strongyle nematodes in three ruminant species in northern Thailand using a nemabiome approach. Fecal samples were collected from buffaloes, cattle, and goats that were raised for meat and for dairy in four provinces in northern Thailand. Strongyle infections were determined using egg flotation and McMaster techniques followed by DNA metabarcoding for species identification. The results showed high prevalence of strongyles especially in goats raised for meat (88 %), and in goats raised for dairy (72 %). Significantly more goats and cattle raised for meat were strongyle egg-positive compared to their dairy counterparts. Notably, deworming frequency was not significantly associated with strongyle egg-positivity in all ruminant groups apart from dairy goats. Nemabiome analysis identified 11 strongyle species across seven genera. Among the ruminant hosts, beef cattle exhibited the highest strongyle richness. Additionally, the dominance of specific strongyle species influenced the differences observed in diversity indices. This research is the first to apply the nemabiome approach to assess strongyle nematode diversity in northern Thailand, providing valuable insights into nematode community compositions. These findings emphasize the importance of molecular techniques for parasite monitoring and the development of targeted control strategies.
Collapse
Affiliation(s)
- Thanakorn Rompo
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Naoki Hayashi
- Laboratory of Parasitology, Graduate School of Infectious Diseases, Faculty of Veterinary Medicine, Hokkaido University, Japan; Division of Parasitology, Veterinary Research Unit, International Institute for Zoonosis Control, Hokkaido University, Japan
| | - Ernest Teo
- Laboratory of Parasitology, Graduate School of Infectious Diseases, Faculty of Veterinary Medicine, Hokkaido University, Japan
| | - Tawatchai Singhla
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chakorn Kunkaew
- Department of Animal Science and Fisheries, Faculty of Science and Agricultural Technology, Rajamangala University of Technology Lanna, Lampang, Thailand
| | | | | | - Saruda Wanganurakkul
- Veterinary Research and Development Center - Eastern Region, Department of Livestock Development, Chon Buri, Thailand
| | - Kanthanis Limwibulpong
- Veterinary Research and Development Center - Eastern Region, Department of Livestock Development, Chon Buri, Thailand
| | - Kanyatip Sangarun
- Veterinary Research and Development Center - Eastern Region, Department of Livestock Development, Chon Buri, Thailand
| | - Napatsorn Suwongsaksri
- Veterinary Research and Development Center - Eastern Region, Department of Livestock Development, Chon Buri, Thailand
| | - Saravalee Suphakarn
- Veterinary Research and Development Center - Eastern Region, Department of Livestock Development, Chon Buri, Thailand
| | | | - Yuto Matsui
- Center for Animal Disease Control (CADIC), University of Miyazaki, Miyazaki, Japan
| | - Takao Irie
- Center for Animal Disease Control (CADIC), University of Miyazaki, Miyazaki, Japan; Laboratory of Veterinary Parasitic Diseases, Department of Veterinary Sciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Ayako Yoshida
- Center for Animal Disease Control (CADIC), University of Miyazaki, Miyazaki, Japan; Laboratory of Veterinary Parasitic Diseases, Department of Veterinary Sciences, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | | | - Naoaki Misawa
- Center for Animal Disease Control (CADIC), University of Miyazaki, Miyazaki, Japan
| | - Nariaki Nonaka
- Laboratory of Parasitology, Graduate School of Infectious Diseases, Faculty of Veterinary Medicine, Hokkaido University, Japan; Division of Parasitology, Veterinary Research Unit, International Institute for Zoonosis Control, Hokkaido University, Japan
| | - Ryo Nakao
- Laboratory of Parasitology, Graduate School of Infectious Diseases, Faculty of Veterinary Medicine, Hokkaido University, Japan; Division of Parasitology, Veterinary Research Unit, International Institute for Zoonosis Control, Hokkaido University, Japan.
| | - Saruda Tiwananthagorn
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand; Research Center of Producing and Development of Products and Innovations for Animal Health and Production, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
33
|
Chang VC, Purandare V, Li S, Andreotti G, Hua X, Wan Y, Dagnall CL, Jones K, Hicks BD, Hutchinson A, Yano Y, Dalton KR, Lee M, Parks CG, London SJ, Sandler DP, Gail MH, Shi J, Hofmann JN, Sinha R, Abnet CC, Vogtmann E, Beane Freeman LE. Animal farming and the oral microbiome in the Agricultural Health Study. ENVIRONMENTAL RESEARCH 2025; 281:121964. [PMID: 40436194 DOI: 10.1016/j.envres.2025.121964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 05/08/2025] [Accepted: 05/24/2025] [Indexed: 06/11/2025]
Abstract
BACKGROUND Raising farm animals imparts various exposures that may shape the human microbiome. The oral microbiome has been increasingly implicated in disease development. Animal farming has also been associated with certain chronic diseases such as cancer; however, underlying biological mechanisms are unclear. We investigated associations between raising farm animals and the oral microbiome in the Agricultural Health Study. METHODS This analysis included 1,245 participants (865 farmers and 380 spouses) who provided oral wash specimens and information on types and numbers of specific animals raised on their farms within 2 years before sample collection. The oral microbiome was measured by sequencing the V4 region of the 16S ribosomal RNA gene. We evaluated associations of farm animal exposures with alpha and beta diversity metrics (within- and between-sample diversity, respectively), as well as presence and relative abundance of specific bacterial genera. All analyses adjusted for potential confounders (e.g., age, sex, smoking, alcohol consumption). RESULTS Overall, 63 % of participants raised farm animals, most commonly cattle (46 %) and hogs (20 %). Those who raised a large number of hogs (≥2,000 vs. no hogs) had higher alpha diversity. Conversely, raising sheep/goats and raising larger numbers of poultry were associated with lower alpha diversity. Beta diversity was not significantly different between participants with and without any farm animals. Participants raising any farm animals had higher relative abundance of Porphyromonas and lower relative abundances of Prevotella and Ruminococcaceae UCG-014. Several genera were more likely to be absent with specific animal exposures (e.g., Capnocytophaga for cattle and sheep/goats; Corynebacterium, Dialister, Stomatobaculum, and Solobacterium for sheep/goats and poultry). CONCLUSIONS This was the largest study of farm animal exposures and the human microbiome to date. Findings suggest that raising specific farm animals may influence the oral microbiome, supporting the need to further investigate the potential role of animal farming in disease etiology.
Collapse
Affiliation(s)
- Vicky C Chang
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA.
| | - Vaishnavi Purandare
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Shilan Li
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Gabriella Andreotti
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Xing Hua
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA; Frederick National Laboratory for Cancer Research/Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Yunhu Wan
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA; Frederick National Laboratory for Cancer Research/Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Casey L Dagnall
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA; Frederick National Laboratory for Cancer Research/Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Kristine Jones
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA; Frederick National Laboratory for Cancer Research/Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Belynda D Hicks
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA; Frederick National Laboratory for Cancer Research/Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Amy Hutchinson
- Cancer Genomics Research Laboratory, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA; Frederick National Laboratory for Cancer Research/Leidos Biomedical Research, Inc., Frederick, MD, USA
| | - Yukiko Yano
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kathryn R Dalton
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Mikyeong Lee
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Christine G Parks
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Stephanie J London
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Mitchell H Gail
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jianxin Shi
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jonathan N Hofmann
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Rashmi Sinha
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Christian C Abnet
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Emily Vogtmann
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Laura E Beane Freeman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
34
|
Wang L, Shen Y, Chen B, Nie H, Gan L, Luo A, He Q, Zhong K, Gao H. Spatial environments of stack and layer shape the quality of Chinese sauce aroma Daqu: Characteristics, functions, and relationships. Int J Food Microbiol 2025; 440:111287. [PMID: 40460797 DOI: 10.1016/j.ijfoodmicro.2025.111287] [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: 12/16/2024] [Revised: 05/14/2025] [Accepted: 05/25/2025] [Indexed: 06/11/2025]
Abstract
The Chinese sauce aroma liquor Daqu (CSDQ) is an important raw material and starter in Baijiu processing, which is formed through solid-state fermentation in an open environment. The spatial environment influences the qualities of CSDQ. In this study, the subtypes of CSDQ were defined by types and layers. Based on multiomic methods, the nutrient substances, fermentation-related parameters, flavor compounds, microbial diversity, and bacterial function of different subtypes were comprehensively evaluated. The obtained results revealed that YQ had the most types of characteristic bacteria, WQ provided the most yeast and the highest fermentation power, and BQ had the most pyrazines and Bacillales. The relationships between subtypes or biochemical factors were presented. Trace bacteria exhibited temperate amplification in YQ and BQ, which may be the essential outcome of CSDQs as natural culture mediums. Our findings provided a better understanding of solid-state fermentation in the Baijiu industry.
Collapse
Affiliation(s)
- Lingchang Wang
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Yi Shen
- Sichuan Langjiu Group Co., Ltd, Luzhou 646523, China
| | - Bo Chen
- Sichuan Langjiu Group Co., Ltd, Luzhou 646523, China
| | - Hongfang Nie
- Sichuan Langjiu Group Co., Ltd, Luzhou 646523, China
| | - Langfei Gan
- Sichuan Langjiu Group Co., Ltd, Luzhou 646523, China
| | - Aimin Luo
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China.
| | - Qiang He
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| | - Kai Zhong
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China.
| | - Hong Gao
- College of Biomass Science and Engineering and Healthy Food Evaluation Research Center, Sichuan University, Chengdu 610065, China
| |
Collapse
|
35
|
Vermote L, Chun BH, Khan SA, De Vuyst L, Jeon CO, Weckx S. Metagenomic and meta-metabolomic analysis of traditional Korean rice vinegar productions shows a large variability between producers. Int J Food Microbiol 2025; 440:111283. [PMID: 40460798 DOI: 10.1016/j.ijfoodmicro.2025.111283] [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: 02/05/2025] [Revised: 04/30/2025] [Accepted: 05/24/2025] [Indexed: 06/11/2025]
Abstract
Cereal vinegars have been used for thousands of years, especially in Asian countries. These vinegars are still produced in a traditional way by a spontaneous, consecutive, alcoholic and acetic acid fermentation process in open vats under non-sterile conditions, which can lead to an unstable and inconsistent flavor and quality. The present study characterized the microbial diversity of complete, traditional Korean rice vinegar productions at two producers (A and B), from steamed rice to rice vinegar, applying high-throughput amplicon-based and shotgun metagenomic sequencing, in combination with meta-metabolomic analysis. Functional analysis based on metagenome-assembled genomes provided insights into the genetic potential of the different microorganisms involved. Producer A used nuruk, a traditional starter, and seed vinegar to start the alcoholic and acetic acid fermentation phases, respectively, which resulted in highly controlled productions even when different fermentation vessels were used. Producer B used only nuruk to start the vinegar productions, and the spontaneous inoculation of acetic acid bacteria did fail in one of the productions. The addition of nuruk resulted in a simultaneous rice starch saccharification and alcoholic fermentation phase characterized by producer-specific moulds, yeasts, and lactic acid bacteria (LAB). During the acetic acid fermentation phase at both producers (a) novel Acetobacter species, related to A. pasteurianus was found. The simultaneous presence of several LAB species made it hard to link them with the production of specific metabolites. Also, the species contributing to ester formation, important for the flavor, was not clear and requires further research.
Collapse
Affiliation(s)
- Louise Vermote
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Department of Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Byung Hee Chun
- Department of Life Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea
| | - Shehzad Abid Khan
- Department of Life Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea
| | - Luc De Vuyst
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Department of Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium
| | - Che Ok Jeon
- Department of Life Science, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, Republic of Korea
| | - Stefan Weckx
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Department of Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, 1050 Brussels, Belgium.
| |
Collapse
|
36
|
Kang D, Lu H, Kang T, Zhang Y, Ge Z, Zhang L, Peng Y. Heterogeneous microstructure induces floatation in high-rate anammox granules. WATER RESEARCH X 2025; 28:100319. [PMID: 40028193 PMCID: PMC11871469 DOI: 10.1016/j.wroa.2025.100319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/11/2024] [Accepted: 02/09/2025] [Indexed: 03/05/2025]
Abstract
The floatation of anammox granules can be a serious challenge in practical wastewater treatment, as it can deteriorate reactor performance and cause bacterial loss. To deepen the understanding of floatation mechanism, in this study, both the floating (F-AnGS) and settling anammox granules (S-AnGS) from a high-rate anammox reactor were comparatively investigated. F-AnGS demonstrated 1.6 times higher specific anammox activity compared to S-AnGS, but only 65 % of produced gas could be successfully released, as quantified by anaerobic respirometry. In addition to the overall EPS accumulation, F-AnGS exhibited a heterogeneous microstructure distinct from that of S-AnGS, as revealed by 3D X-ray microscopic imaging at the single granule level. The heterogeneous distribution of EPS, which can form a dense surface layer, was the main cause for granule floatation. The heterogeneous microstructure of F-AnGS can reduce the distance between microorganisms and enhance the metabolic interaction between anammox bacteria and heterotrophs. The abundance of community members did not have a significant variation, but the functional genes related to anammox and partial denitrification pathway were significantly increased, indicating the enhanced nitrite loop in F-AnGS. This study proposed new structural insights into mechanism of anammox granule floatation, suggesting the appropriate activity control of granule-based anammox process.
Collapse
Affiliation(s)
- Da Kang
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, PR China
| | - Huifeng Lu
- Zhejiang Water Healer Environmental Technology Co., Ltd, Hangzhou, PR China
| | - Tingting Kang
- Zhejiang Water Healer Environmental Technology Co., Ltd, Hangzhou, PR China
| | - Yihan Zhang
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, PR China
| | - Zheng Ge
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, PR China
| | - Liang Zhang
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, PR China
| | - Yongzhen Peng
- National Engineering Laboratory for Advanced Municipal Wastewater Treatment and Reuse Technology, Department of Environmental Engineering, Beijing University of Technology, PR China
| |
Collapse
|
37
|
Zhang L, Wang H, Liu X, Zhan H, Schneidewind U, Krause S, Jin M, Liang X, Liu Y, Li P. Denitrification dominates nitrate attenuation and nitrous oxide effluxes under water table fluctuations. JOURNAL OF HAZARDOUS MATERIALS 2025; 493:138325. [PMID: 40273856 DOI: 10.1016/j.jhazmat.2025.138325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 03/02/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Agricultural soils in riparian zones near rivers often experience frequent water table fluctuations, which can lead to increased nitrogen losses and greenhouse gas emissions via the nitrogen biogeochemical processes. However, the influence of water table fluctuations on the multiple nitrogen transformation processes that dominate nitrate attenuation and nitrous oxide (N2O) effluxes remains poorly understood. In this study, the dynamic changes in depth-dependent nitrate attenuation and soil N2O effluxes, and the responses of microbial communities influenced by water table fluctuations were studied using a series of large column experiments. Our results revealed that dissolved oxygen (DO) concentrations at a depth of -10 cm in sand columns with three different grain sizes (fine→medium→coarse) oscillated, producing oxidizing conditions during drainage and reducing conditions during imbibition periods. DO micro-sensors installed in a layered (sand and sandy loam) column as well as in two sandy loam columns with different regimes in induced water table changes all revealed steady hypoxic conditions. The diversity of the microbial community was significantly correlated with total nitrogen, total organic carbon, and nitrate concentrations, as well as potential denitrification rates. The dominant microbial populations related to the nrfA gene were Methanothrix and Sedimentibacte, whereas those related to denitrification (nirK, nirS, and nosZ) were Pseudomonas and Sulfuricaulis. These findings improve our understanding of the effects of water table fluctuations on groundwater nitrate loss in riparian corridors.
Collapse
Affiliation(s)
- Lin Zhang
- Key Laboratory of Marine Environment and Ecology, Ministry of Education, College of Environmental Science and Engineering, Ocean University of China, Qingdao 266100, China; School of Environmental Studies, China University of Geosciences, Wuhan 430078, China; School of Geography, Earth and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Helin Wang
- State Key Laboratory of Geomicrobiology and Environmental Changes, China University of Geosciences, Wuhan 430078, China
| | - Xiaohan Liu
- State Key Laboratory of Geomicrobiology and Environmental Changes, China University of Geosciences, Wuhan 430078, China
| | - Hongbin Zhan
- Department of Geology and Geophysics, Texas A&M University, College Station, TX 77843-3115, USA
| | - Uwe Schneidewind
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom
| | - Stefan Krause
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, United Kingdom; Univ Lyon, Université Lyon 1 Claude Bernard, ENTPE, CNRS, UMR 5023 LEHNA, 3 Rue M. Audin, Vaulx-en-Velin Cedex 69518, France
| | - Menggui Jin
- School of Environmental Studies, China University of Geosciences, Wuhan 430078, China; State Key Laboratory of Geomicrobiology and Environmental Changes, China University of Geosciences, Wuhan 430078, China.
| | - Xing Liang
- School of Environmental Studies, China University of Geosciences, Wuhan 430078, China
| | - Yanfeng Liu
- School of Environmental Studies, China University of Geosciences, Wuhan 430078, China
| | - Ping Li
- State Key Laboratory of Geomicrobiology and Environmental Changes, China University of Geosciences, Wuhan 430078, China.
| |
Collapse
|
38
|
Yan J, Li X, Pan Z, Lin X, Zuo Q, Zhou J, Zhou S, Sui F, Zhang L, Fischel MHH. Red mud causes dynamic changes in the soil microbial community and cadmium fractions in a slightly cadmium-contaminated paddy soil. JOURNAL OF HAZARDOUS MATERIALS 2025; 493:138349. [PMID: 40267706 DOI: 10.1016/j.jhazmat.2025.138349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
Red mud is a highly alkaline industrial by-product rich in iron oxides with great potential for soil cadmium remediation. Although the stabilization of Cd by red mud is well reported in rice potted and field experiments, the influence of red mud on microbial communities in paddy soil and the contribution of soil microbial communities subjected to red mud in Cd stabilization remain unknown. This study used high-throughput sequencing and bioinformatics, combined with a sequential extraction procedure, to determine the microbiological mechanisms of rice Cd reduction by red mud and information on the corresponding soil Cd fraction. The results showed that red mud significantly increased the soil pH and iron and manganese oxide-bound Cd fractions. Red mud application influenced the microbial beta diversity rather than the alpha diversity, especially for bacteria. Unique taxa associated with iron reduction (e.g., phylum Firmicutes and genus Anaeromyxobacter) were enriched at the rice-filling stage, which may contribute to the stabilization of Cd. Red mud application caused little difference in the fungal communities. A 2 % red mud amendment successfully decreased the grain Cd content in a high Cd-accumulating rice cultivar by 72 %. Red mud effectively reduces Cd accumulation in the short-term and demonstrates potential for remedial applications. This study provides microbiological evidence for stabilizing Cd in red mud, but its long-term environmental impact and field applicability require further research.
Collapse
Affiliation(s)
- Jiali Yan
- School of Civil Engineering and Architecture, Chuzhou University, Chuzhou 239000, China
| | - Xuwei Li
- Nanjing Institute of Environmental Science, Ministry of Ecology and Environment of the People's Republic of China, Nanjing 210042, China
| | - Zhengguo Pan
- School of Civil Engineering and Architecture, Chuzhou University, Chuzhou 239000, China
| | - Xiaochen Lin
- Nanjing Institute of Environmental Science, Ministry of Ecology and Environment of the People's Republic of China, Nanjing 210042, China
| | - Qinglin Zuo
- School of Resources and Environmental Engineering, Anhui University, Hefei 230601, China
| | - Jiankang Zhou
- Key Laboratory of Soil Pollution Control and Remediation of Henan Province, College of Resources and Environment, Henan Agricultural University, Zhengzhou, China
| | - Shiqi Zhou
- School of Civil Engineering and Architecture, Chuzhou University, Chuzhou 239000, China
| | - Fuqing Sui
- Key Laboratory of Soil Pollution Control and Remediation of Henan Province, College of Resources and Environment, Henan Agricultural University, Zhengzhou, China.
| | - Lei Zhang
- School of Civil Engineering and Architecture, Chuzhou University, Chuzhou 239000, China.
| | - Matthew H H Fischel
- Sustainable Agricultural Systems Laboratory, USDA-Agricultural Research Service, Beltsville, MD 20705, USA
| |
Collapse
|
39
|
Castagnoli R, Pala F, Subramanian P, Oguz C, Schwarz B, Lim AI, Burns AS, Fontana E, Bosticardo M, Corsino C, Angelova A, Delmonte OM, Kenney H, Riley D, Smith G, Ott de Bruin L, Oikonomou V, Dos Santos Dias L, Fink D, Bohrnsen E, Kimzey CD, Marseglia GL, Alva-Lozada G, Bergerson JR, Brett A, Brigatti KW, Dimitrova D, Dutmer CM, Freeman AF, Ale H, Holland SM, Licciardi F, Pasic S, Poskitt LE, Potts DE, Dasso JF, Sharapova SO, Strauss KA, Ward BR, Yilmaz M, Kuhns DB, Lionakis MS, Daley SR, Kong HH, Segre JA, Villa A, Pittaluga S, Walter JE, Vujkovic-Cvijin I, Belkaid Y, Notarangelo LD. Immunopathological and microbial signatures of inflammatory bowel disease in partial RAG deficiency. J Exp Med 2025; 222:e20241993. [PMID: 40314722 PMCID: PMC12047384 DOI: 10.1084/jem.20241993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 03/16/2025] [Accepted: 04/10/2025] [Indexed: 05/03/2025] Open
Abstract
Partial RAG deficiency (pRD) can manifest with systemic and tissue-specific immune dysregulation, with inflammatory bowel disease (IBD) in 15% of the patients. We aimed at identifying the immunopathological and microbial signatures associated with IBD in patients with pRD and in a mouse model of pRD (Rag1w/w) with spontaneous development of colitis. pRD patients with IBD and Rag1w/w mice showed a systemic and colonic Th1/Th17 inflammatory signature. Restriction of fecal microbial diversity, abundance of pathogenic bacteria, and depletion of microbial species producing short-chain fatty acid were observed, which were associated with impaired induction of lamina propria peripheral Treg cells in Rag1w/w mice. The use of vedolizumab in Rag1w/w mice and of ustekinumab in a pRD patient were ineffective. Antibiotics ameliorated gut inflammation in Rag1w/w mice, but only bone marrow transplantation (BMT) rescued the immunopathological and microbial signatures. Our findings shed new light in the pathophysiology of gut inflammation in pRD and establish a curative role for BMT to resolve the disease phenotype.
Collapse
Affiliation(s)
- Riccardo Castagnoli
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Poorani Subramanian
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cihan Oguz
- Integrated Data Sciences Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Schwarz
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Ai Ing Lim
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrew S. Burns
- NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | | | - Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cristina Corsino
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Angelina Angelova
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ottavia M. Delmonte
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Heather Kenney
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Deanna Riley
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Grace Smith
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lisa Ott de Bruin
- Willem-Alexander Children’s Hospital, Department of Pediatrics, Pediatric Stem Cell Transplantation Program, Leiden University Medical Center, Leiden, Netherlands
| | - Vasileios Oikonomou
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Lucas Dos Santos Dias
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Danielle Fink
- Neutrophil Monitoring Lab, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Eric Bohrnsen
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Cole D. Kimzey
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Gian Luigi Marseglia
- Pediatric Unit, Department of Clinical, Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, Pavia, Italy
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Guisela Alva-Lozada
- Allergy and Immunology Division Hospital Nacional Edgardo Rebagliati Martins, Lima, Peru
| | - Jenna R.E. Bergerson
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ana Brett
- Hospital Pediátrico, Unidade Local de Saúde de Coimbra, Coimbra, Portugal
- Clínica Universitária de Pediatria, Faculdade de Medicina, Universidade de Coimbra, Coimbra, Portugal
| | | | - Dimana Dimitrova
- Experimental Transplantation and Immunotherapy Branch, National Cancer Institute of the National Institutes of Health, Bethesda, MD, USA
| | - Cullen M. Dutmer
- Allergy and Immunology, Children’s Healthcare of Atlanta, Emory University School of Medicine, Atlanta, GA, USA
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hanadys Ale
- Division of Immunology, Allergy and Rheumatology, Joe DiMaggio Children’s Hospital, Memorial Healthcare System, Hollywood, FL, USA
| | - Steven M. Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Francesco Licciardi
- Immuno-reumatologia, Pediatria Specialistica Universitaria, Ospedale Infantile Regina Margherita, Torino, Italy
| | - Srdjan Pasic
- Department of Pediatric Immunology, Mother and Child Health Institute, Medical Faculty, University of Belgrade, Belgrade, Serbia
| | | | - David E. Potts
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Joseph F. Dasso
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Svetlana O. Sharapova
- Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | | | - Brant R. Ward
- Division of Allergy and Immunology, Children’s National Hospital, Washington, DC, USA
| | - Melis Yilmaz
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Douglas B. Kuhns
- Neutrophil Monitoring Lab, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Michail S. Lionakis
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Stephen R. Daley
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Australia
| | - Heidi H. Kong
- Cutaneous Microbiome and Inflammation Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Julia A. Segre
- Microbial Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anna Villa
- San Raffaele-Telethon Institute for Gene Therapy (SR-Tiget), IRCSS San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Stefania Pittaluga
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jolan E. Walter
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children’s Hospital, St. Petersburg, FL, USA
| | - Ivan Vujkovic-Cvijin
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Immunology, Institut Pasteur, Paris, France
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
40
|
Thangadurai T, Dobretsov S, Aeby G. Exploring bacterial diversity in Acropora pharaonis: Implications for coral health and growth anomalies. Microb Pathog 2025; 205:107616. [PMID: 40294758 DOI: 10.1016/j.micpath.2025.107616] [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: 10/21/2024] [Revised: 03/07/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Coral growth anomalies (GA) affect many coral genera across the world, yet the etiology of GAs remains unknown, with limited knowledge of associated bacteria. In this study, we investigated bacterial associations between the growth anomalies (GAs) and healthy (H) portions of coral colonies in Acropora faraonis for two seasons to understand microbial dynamics. Additionally, we examined bacteria in water (W), which could be affecting coral bacterial communities. We found that alpha diversity remained consistent between healthy and GA coral tissues, but their relative abundances differed significantly. Notably, differential analysis revealed the abundance of Endozoicomonas spp., differed significantly between GA and H tissue, although it remains the dominant genus in both GA and H tissue. The high relative abundance of Endozoicomonas spp. in both GA and healthy tissue underscores its potential role in maintaining coral health. Structural modifications in GAs, such as changes in polyp sizes or densities, could be responsible for these differences in bacterial abundance. Similarly, microbial community composition remained consistent between seasons but differed in abundance again. We found differences between microbial communities of GAs and water, but no significant differences were observed between GAs and H, and no previously established bacterial pathogens were detected in GA tissue. These findings describe bacterial community patterns in GAs, but their potential role in its pathogenesis remains unknown. Further metagenomic and meta-transcriptomic analyses are needed to understand potential bacterial involvement in GAs. Additionally, investigating viruses and fungi in GA tissue is recommended to gain deeper insights into GA pathogenesis.
Collapse
Affiliation(s)
- Thinesh Thangadurai
- Department of Marine Science and Fisheries, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al Khoud 123 PO Box 34, Muscat, Oman
| | - Sergey Dobretsov
- Department of Marine Science and Fisheries, College of Agricultural and Marine Sciences, Sultan Qaboos University, Al Khoud 123 PO Box 34, Muscat, Oman; UNESCO Chair in Marine Biotechnology, Center of Excellence in Marine Biotechnology, Sultan Qaboos University, Al Khoud 123 PO Box 50, Muscat, Oman.
| | - Greta Aeby
- Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar
| |
Collapse
|
41
|
Wang W, Chen Y, Rasic M, Ascoli C, Hatch JE, Nemsick NK, Deschamp AR, Davis SD, Sanders DB, Ranganathan S, Stick S, Perkins DL, Thomas Ferkol, Finn PW. An observational study of the lung microbiome and lung function in young children with cystic fibrosis across two countries with differing antibiotic practices. Microb Pathog 2025; 205:107628. [PMID: 40288428 DOI: 10.1016/j.micpath.2025.107628] [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: 10/04/2024] [Revised: 04/16/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Cystic fibrosis (CF) lung disease begins early, and prophylactic antibiotics have been used to prevent Staphylococcus aureus infection. This study examined the lung microbiome in two countries with differing antibiotic practices and its relationship to lung function in young children with CF. METHODS A binational, longitudinal, observational study was performed to define the lower airway microbiome in infants with CF. 16S rRNA sequencing was performed using lavage fluid to characterize the lung microbiota in 45 infants with and without prophylactic antibiotic therapy at an average age of approximately 3 months and 14 months. The association between pulmonary function, bacterial community diversities, and taxa was assessed. RESULTS Expected CF bacterial genera and non-traditional bacteria, such as Streptococcus, were identified as core taxa. Microbial community shifts were observed in infants who received antibiotic prophylaxis, with lower alpha diversity (ANOVA, P < 0.05) and a higher proportion of Streptococcus at the first visit. Beta diversity (FEV0.5z; MiRKAT, P < 0.05) and Streptococcus were associated with FEV0.5z (LASSO and linear regression, β < 0). Functional annotation suggested that alteration of lung microbiota may be linked to antimicrobial resistance. CONCLUSIONS Lung microbial diversity in infants with CF varied between the two countries, particularly during early infancy. A shift in the lung microbiome toward a higher relative abundance of Streptococcus was associated with reduced pulmonary function.
Collapse
Affiliation(s)
- Wangfei Wang
- Richard and Loan Hill Department of Biomedical Engineering, College of Engineering and Medicine, University of Illinois at Chicago, Chicago, IL, USA; Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Yang Chen
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA; Department of Biological Sciences, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Mladen Rasic
- Richard and Loan Hill Department of Biomedical Engineering, College of Engineering and Medicine, University of Illinois at Chicago, Chicago, IL, USA; Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Christian Ascoli
- Division of Pulmonary, Critical Care, Sleep and Allergy, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Joseph E Hatch
- Department of Pediatrics, UNC Children's, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Nicole K Nemsick
- Department of Biological Sciences, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Ashley R Deschamp
- Department of Pediatrics, University of Nebraska Medical Center, Children's Hospital and Medical Center, Omaha, NE, USA
| | - Stephanie D Davis
- Department of Pediatrics, UNC Children's, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Don B Sanders
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sarath Ranganathan
- Department of Pediatrics, University of Melbourne, Infection and Immunity, Murdoch Children's Research Institute, Parkville, Australia
| | - Stephen Stick
- Department of Pediatrics, University of Western Australia, Centre for Child Health Research, Perth, Australia
| | - David L Perkins
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA; Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| | - Thomas Ferkol
- Department of Pediatrics, UNC Children's, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| | - Patricia W Finn
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM, USA; Department of Biological Sciences, College of Liberal Arts and Sciences, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
42
|
Peng Q, Zhou H, Zheng H, Xie G. Investigating the role of primary fungi in Huangjiu fermentation: Insights from flavor orientation and synthetic microbiomes. Food Microbiol 2025; 129:104765. [PMID: 40086991 DOI: 10.1016/j.fm.2025.104765] [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: 10/25/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
Huangjiu, a traditional alcoholic beverage, presents a complex fermentation ecosystem primarily influenced by specific fungal species. This study utilized a culture-dependent approach and amplicon sequencing to explore fungal community succession during Huangjiu fermentation. Key fungi identified include Saccharomyces cerevisiae, Aspergillus species (flavus, oryzae, niger), Saccharomycopsis fibuligera, Thermomyces lanuginosus, Rhizopus arrhizus, Issatchenkia orientalis, Wickerhamomyces anomalus, and Diutina rugosa. Employing a synthetic microbiome, we developed a dual-strain fermentation system to evaluate the impact of these fungi on Huangjiu's organoleptic properties. Introduction of these fungi significantly altered the flavor profile, enhancing 23 volatile organic compounds (VOCs), with S. fibuligera notably increasing nine distinct VOCs. While molds contributed to bitterness by increasing bitter amino acids, S. fibuligera effectively mitigated these components, enhancing the beverage's alcohol body, smoothness, and balance. These findings provide crucial insights for optimizing Huangjiu fermentation to improve its quality and appeal.
Collapse
Affiliation(s)
- Qi Peng
- National Engineering Research Center for Chinese CRW (branch center), School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Huihui Zhou
- National Engineering Research Center for Chinese CRW (branch center), School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Huajun Zheng
- National Engineering Research Center for Chinese CRW (branch center), School of Life and Environmental Sciences, Shaoxing University, Shaoxing, 312000, Zhejiang, China
| | - Guangfa Xie
- Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, College of Biology and Environmental Engineering, Zhejiang Shuren University, Hangzhou, 310015, Zhejiang, China.
| |
Collapse
|
43
|
Montiel-Corona V, Buitrón G. Light/dark cycles and iron supplementation to enhance the simultaneous production of polyhydroxyalkanoates, 5-aminolevulinic acid, coenzyme Q 10, and pigments through photofermentation. BIORESOURCE TECHNOLOGY 2025; 429:132513. [PMID: 40222488 DOI: 10.1016/j.biortech.2025.132513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
This study aimed to obtain 5-aminolevulinic acid (5-ALA), Coenzyme Q10 (CoQ10), polyhydroxyalkanoates (PHA), carotenoids, and bacteriochlorophylls (Bchl) through the photofermentation of residual wine lees. Light/dark cycles and iron supplementation were evaluated. Under 12-hour light/dark cycles, the production of 5-ALA, CoQ10, carotenoids, and Bchl increased by 1.7, 2.8, 1.7, and 2.4 times, respectively, compared to the continuous illumination control, while PHA production decreased from 511 to 445 mg/L. Combining light/dark cycles with iron supplementation enhanced the biomass production rate. The CoQ10 content increased by 4.9 times (reaching 8.8 mg/g-dw), carotenoids by 3.7 times (6.4 mg/g-dw), and Bchl by 6.4 times (17.9 mg/g-dw) compared to the control treatment, while maintaining 5-ALA at 5.3 µmol/L and PHA at 377 mg/L. The combination of light/dark cycles and iron provides a triple benefit: increased production of value-added substances, enhanced biomass production rate, and improved organic matter removal, making it an attractive option for winery effluent treatment and valorization.
Collapse
Affiliation(s)
- Virginia Montiel-Corona
- Laboratory for Research on Advanced Processes for Water Treatment, Unidad Académica Juriquilla, Instituto de Ingeniería, Universidad Nacional Autónoma de México, Blvd. Juriquilla 3001, Querétaro 76230, México
| | - Germán Buitrón
- Laboratory for Research on Advanced Processes for Water Treatment, Unidad Académica Juriquilla, Instituto de Ingeniería, Universidad Nacional Autónoma de México, Blvd. Juriquilla 3001, Querétaro 76230, México.
| |
Collapse
|
44
|
Li B, Liang C, Xu B, Song P, Liu D, Zhang J, Gu H, Jiang F, Gao H, Cai Z, Zhang T. Extreme winter environment dominates gut microbiota and metabolome of white-lipped deer. Microbiol Res 2025; 297:128182. [PMID: 40252261 DOI: 10.1016/j.micres.2025.128182] [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: 07/25/2024] [Revised: 03/23/2025] [Accepted: 04/10/2025] [Indexed: 04/21/2025]
Abstract
Qinghai-Tibet Plateau (QTP) is marked by harsh environments that drive the evolution of unique nutrient metabolism mechanism in indigenous animal gut microbiotas. Yet, responses of these microbiotas to different extreme environments remain poorly understood. White-lipped deer (Przewalskium albirostris), a native endangered species in the QTP, serves as an ideal model to study how gut microbiotas adapt to season and human disturbances. Here, a multi-omics integrated analysis of 16S rRNA, metagenomics, and untargeted metabolomics was performed to investigate the composition, function, and metabolic characteristics of gut microbiota in White-lipped deer across different seasons and living environments. Our results revealed that extreme winter environment dominated the composition, function, and metabolism of gut microbiota in white-lipped deer. The white-lipped deer exhibited an enriched gut microbiota associated with producing short-chain fatty acids in winter, with core feature genera including norank_o_Rhodospirillales, Rikenellaceae_RC9_gut_group, and unclassified_c_Clostridia. However, potential pathogenic bacteria and few short-chain fatty acid producers, with core feature genera including norank_f_p-2534-18B5_gut_group, Cellulosilyticum, and Paeniclostridium, showed enrichment in captivity. Pathways associated with carbohydrate metabolism, amino acid metabolism, and immune regulation showed enrichment in winter group as an adaptation to the cold and food scarcity. Among these, Rikenellaceae_RC9_gut_group and unclassified_c_Clostridia contributed significantly to these metabolic pathways. The gut microbiota of white-lipped deer exhibited enrichment in pathways related to intestinal inflammation and enhanced immune regulation to alleviate the stress of captivity. Among these, norank_f_p-2534-18B5_gut_group contributed the most to these pathways. Butyric, valeric, and valproic acids were significantly more abundant in the winter group, while 3-hydroxybutyric and (S)-beta-aminoisobutyric acids were higher in the captive group. Furthermore, enriched metabolites and associated pathways in both groups further supported the inferences on metagenomic functions. This study confirms the key role of specific gut microbiota in adapting to high-altitude winters and anthropogenic disturbances, emphasizing its importance for environmental resilience in wild, high-altitude mammals.
Collapse
Affiliation(s)
- Bin Li
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Chengbo Liang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Bo Xu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Pengfei Song
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | | | | | - Haifeng Gu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Feng Jiang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Hongmei Gao
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China
| | - Zhenyuan Cai
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China.
| | - Tongzuo Zhang
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, China; Qinghai Provincial Key Laboratory of Animal Ecological Genomics, Xining, China.
| |
Collapse
|
45
|
Ahn JS, Lee YB, Han EJ, Choi YJ, Kim DH, Kwok SK, Choi HK, Chung HJ. Identification of specific gut microbes and their therapeutic potential in ameliorating systemic lupus erythematosus in a mouse model. Life Sci 2025; 374:123684. [PMID: 40320135 DOI: 10.1016/j.lfs.2025.123684] [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: 02/13/2025] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/11/2025]
Abstract
AIMS The gut microbiome significantly influences autoimmune diseases, including systemic lupus erythematosus (SLE). This study aimed to characterize the gut microbiome and metabolome in SLE and evaluate the therapeutic potential of specific microbial supplementation in MRL/lpr mice. MATERIALS AND METHODS MRL/lpr mice, a well-established model for SLE, were used to analyze gut microbiome changes before and after SLE symptom onset. 16S rRNA sequencing and GC-MS-based metabolic profiling were performed to identify key microbial species and associated metabolites. Selected microbes were supplemented in MRL/lpr mice for 10 weeks, and their effects on SLE symptoms and Th17/Treg balance were evaluated. KEY FINDINGS Eisenbergiella massiliensis, Lacrimispora saccharolytica, and Hungatella xylanolytica were significantly decreased in MRL/lpr mice following the onset of SLE symptoms. These microbes were strongly correlated with specific metabolites, including 5-cholestanol, cholesterol, p-cresol, and indole. Supplementation with these microbes alleviated SLE symptoms and modulated the Th17/Treg balance. SIGNIFICANCE This study highlights the critical role of gut microbiota in immune regulation and SLE symptom relief. Targeted microbial supplementation may serve as a novel therapeutic strategy for managing SLE.
Collapse
Affiliation(s)
- Ji-Seon Ahn
- Honam Regional Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea
| | - Ye-Been Lee
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Eui-Jeong Han
- Honam Regional Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea
| | - Yu-Jin Choi
- Honam Regional Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea; Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Da-Hye Kim
- Honam Regional Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea; Department of Biomedical Sciences and Institute for Medical Science, Jeonbuk National University Medical School, Jeonju, Jeonbuk 54907, Republic of Korea
| | - Seung Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyung-Kyoon Choi
- College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea.
| | - Hea-Jong Chung
- Honam Regional Center, Korea Basic Science Institute, Gwangju 61751, Republic of Korea; College of Pharmacy, Chung-Ang University, Seoul 06974, Republic of Korea; Department of Bio-Analysis Science, University of Science & Technology, Daejeon 34413, Republic of Korea.
| |
Collapse
|
46
|
Diniz BC, Wilfert P, Sorokin DY, van Loosdrecht MCM. Anaerobic digestion at high-pH and alkalinity for biomethane production: Insights into methane yield, biomethane purity, and process performance. BIORESOURCE TECHNOLOGY 2025; 429:132505. [PMID: 40220921 DOI: 10.1016/j.biortech.2025.132505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/21/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
The role of high-pH conditions in anaerobic digestion (AD) has traditionally been confined to it's use in pre-treatment processes. However, operating AD at elevated pH and alkalinity offers significant advantages, including in-situ upgrading of biogas to biomethane. This study examines the potential and scalability of AD under these conditions (pH ∼ 9.3; alkalinity ∼ 0.5 eq/L). The substrate used was the alkaline waste generated from the extraction of extracellular polymeric substances (EPS) from aerobic granular sludge (AGS), and the inoculum used was a haloalkaliphile microbial community from soda lake sediments. To evaluate the system's performance, the organic loading rate (OLR) was incrementally increased. The highest methane production obtained was 8.4 ± 0.1 mL/day/gVSadded at a hydraulic retention time (HRT) of 15 days and an OLR of 1 kgVS/day/m3. At this loading rate, methanogenesis became the rate limiting conversion. The maximum volatile solids conversion was 48.1 ± 1.1 %. Throughout the reactor operation, methane purity in the biogas consistently exceeded 90 % peaking at 96.0 ± 0.2 %, showcasing the potential for in-situ biogas purification under these conditions. In addition, no ammonia inhibition was observed, even with free-ammonia (NH3) concentrations reaching up to 14 mM. This study underscores the potential of high-pH anaerobic digestion as a sustainable method for both waste treatment and energy recovery.
Collapse
Affiliation(s)
- Beatriz C Diniz
- Department of Biotechnology, Faculty of Applied Sciences, Delft University of Technology, Van der Maasweg 9, Delft 2629 HZ, the Netherlands.
| | - Philipp Wilfert
- Department of Biotechnology, Faculty of Applied Sciences, Delft University of Technology, Van der Maasweg 9, Delft 2629 HZ, the Netherlands; Labor für Siedlungswasserwirtschaft und Abfalltechnik, Fachbereich Bauwesen, Technische Hochschule Lübeck 23562 Lübeck, Germany
| | - Dimitry Y Sorokin
- Department of Biotechnology, Faculty of Applied Sciences, Delft University of Technology, Van der Maasweg 9, Delft 2629 HZ, the Netherlands; Winogradsky Institute of Microbiology, Federal Research Centre of Biotechnology, Russian Academy of Sciences, Moscow, Russia
| | - Mark C M van Loosdrecht
- Department of Biotechnology, Faculty of Applied Sciences, Delft University of Technology, Van der Maasweg 9, Delft 2629 HZ, the Netherlands
| |
Collapse
|
47
|
Keeley N, Dunlop K, Laroche O, Hansen PK, Angell IL, Rudi K. An approach for quantifying the influence of fish farm waste on hard-bottom habitats. MARINE POLLUTION BULLETIN 2025; 217:118039. [PMID: 40315742 DOI: 10.1016/j.marpolbul.2025.118039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/20/2025] [Accepted: 04/21/2025] [Indexed: 05/04/2025]
Abstract
Sea-based fish farms release significant quantities of fish feaces, waste feed and associated contaminants into the surrounding environment, necessitating ecological effects-based monitoring. Traditionally, this has been achieved by measuring geochemical and biological properties of sediments obtained by a benthic grab. However, many modern high-capacity farms are situated over hard substrates, rendering conventional sampling methods ineffective and hindering environmental management. Fortunately, new possibilities have emerged with the advent of high-throughput sequencing, and in-particular, the use of microbial eDNA as an indicator of benthic enrichment, and new methods have been developed for obtaining the source material irrespective of substrate. Here, we first demonstrate how applying the default approach used by the world's largest salmon farming industry to hard substrates is essentially qualitative and can lead to inappropriate conclusions. We then show the results from a substrate independent benthic sampling (SIBS) method coupled with a structured quantitative visual assessment of seabed images. The approach utilizes three novel indicators that quantify the degree of visual organic loading (VOL) and ecological effects (VEE) and elucidate the magnitude and spatial extent of the waste influence field via an index (bMBI) derived from 16S eDNA. VOL and VEE were useful for quantifying and classifying effects beneath farms, but were of limited use further away, whereas the bMBI was both highly quantitative and sensitive in the immediate and more distant receiving environments. This dual approach provides a basis for the development of a quantitative hard-bottom monitoring system and should therefore permit effective environment management in the future.
Collapse
Affiliation(s)
- Nigel Keeley
- Institute of Marine Research, Framsenteret, PO Box 6606, Stakkevollan, 9296 Tromsø, Norway.
| | - Katherine Dunlop
- Institute of Marine Research, Framsenteret, PO Box 6606, Stakkevollan, 9296 Tromsø, Norway
| | | | - Pia Kupka Hansen
- Institute of Marine Research, Postbox 1870, Nordnes, N-5817 Bergen, Norway
| | | | - Knut Rudi
- Norwegian University of Life Sciences, Ås, Norway
| |
Collapse
|
48
|
Kakar FL, Aqeel H, Okoye F, Elbeshbishy E, Liss SN. Microbial shifts and VFA production in the optimization of anaerobic digestion by thermal hydrolysis coupled with vacuum fermentation. BIORESOURCE TECHNOLOGY 2025; 429:132481. [PMID: 40187500 DOI: 10.1016/j.biortech.2025.132481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
This study investigated a novel thermal-hydrolysis combined with a vacuum fermentation system for high-grade volatile fatty acids (VFA) recovery, and the corresponding changes in the microbial community. Four systems with and without hydrothermal pre-treatment (HTP) and vacuum were mobilized; results revealed that integration of HTP with vacuum has the highest potential in terms of VFA recovery, sludge disintegration, and solid reduction. HTP and vacuum fermentation systems were associated with the highest COD solubilization (45 %), and VFA yield (0.32 g COD/g VSS added). Vacuum fermenters with and without pre-treatment have the highest specific denitrification rates of 7.6 and 7.2 mg NO3-N/g VSS.h, respectively, compared to all other samples and control (acetate). Changes brought about by vacuum fermentation included a shift in the microbial community toward enriching fermenters, mainly Caprothermobacteria and Thermotagea, responsible for VFA production.
Collapse
Affiliation(s)
- Farokh Laqa Kakar
- Environmental Research Group for Resource Recovery, Department of Civil Engineering, Faculty of Engineering, Architecture and Science, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Hussain Aqeel
- Department of Chemistry and Biology, Faculty of Science, Toronto Metropolitan University 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Frances Okoye
- Environmental Research Group for Resource Recovery, Department of Civil Engineering, Faculty of Engineering, Architecture and Science, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Elsayed Elbeshbishy
- Environmental Research Group for Resource Recovery, Department of Civil Engineering, Faculty of Engineering, Architecture and Science, Toronto Metropolitan University, 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada
| | - Steven N Liss
- Department of Chemistry and Biology, Faculty of Science, Toronto Metropolitan University 350 Victoria Street, Toronto, Ontario M5B 2K3, Canada; School of Environmental Studies, Queen's University, Kingston, ON K7L 3N6, Canada; Department of Microbiology, Stellenbosch University, Private Bag, XI, Matieland, 7602 Stellenbosch, South Africa.
| |
Collapse
|
49
|
Liu X, Li Y, Yuan C, Zhao Y, Zhou L, Yan Y, Ren J, Liu Q. Sophocarpine suppresses MAPK-mediated inflammation by restoring gut microbiota in colorectal cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156833. [PMID: 40393246 DOI: 10.1016/j.phymed.2025.156833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/18/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND Colorectal cancer (CRC), as one of the most common cancers globally, poses a significant challenge to public health due to its high incidence and mortality rates. This underscores the need for continuous exploration of new therapeutic targets and effective drugs. Sophocarpine (SC), a natural compound derived from traditional Chinese medicine, holds considerable therapeutic potential in the treatment of CRC, however, the relevant mechanisms remains unclear. PURPOSE This study aims to explore the anti-tumor effects of SC against CRC by modulating gut microbiota, and uncover potential mechanisms linking SC's therapeutic effects to gut microbiota regulation by analyzing the impact of SC on microbiota composition and CRC progression. MATERIAL This study explores the impact of SC on the gut microbiota in CRC by constructing subcutaneous xenograft tumors of CRC and integrating 16S rRNA sequencing and RNA transcriptomic sequencing. The fecal microbiota transplantation (FMT) mouse model was used to validate the biological function of SC in correcting gut microbiota dysbiosis to treat CRC. Subsequently, we conducted in vitro studies on the molecular mechanisms by which SC regulates the gut microbiota as an effective hallmark of CRC treatment, using lipopolysaccharide (LPS) to simulate an inflammatory gut microbiota environment and P38 MAPK knockdown cell line. RESULTS SC significantly inhibited CRC cell proliferation with IC50 values of 2.547±0.256 μM for HCT116 and 2.851±0.332 μM for LoVo cells. In vivo experiments demonstrated that SC effectively suppressed tumor growth in xenograft models. 16S rRNA sequencing revealed that SC modulated gut microbiota composition, particularly affecting Bacteroides and Alistipes populations. SC significantly reduced the levels of inflammatory factors and inhibited the MAPK signaling pathway, as evidenced by decreased p-JNK, p-p38 MAPK, and p-NF-κB p65 expression. CONCLUSIONS Current clinical practice still lacks effective therapeutic agents targeting CRC through gut microbiota modulation. This study presents the first evidence that SC, a natural compound, exhibits dual-action therapeutic efficacy against CRC progression by simultaneously modulating gut microbial composition and suppressing MAPK pathway-mediated inflammatory responses. These findings highlight SC's novel therapeutic potential as a promising microbiota-regulating candidate for CRC intervention, offering an innovative approach that bridges microbial ecology with cancer signaling pathways.
Collapse
Affiliation(s)
- Xiangjun Liu
- Laboratory Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China
| | - Yu Li
- Laboratory Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China
| | - Chenyue Yuan
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China
| | - Yong Zhao
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China
| | - Lin Zhou
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China
| | - Yuting Yan
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China
| | - Jianlin Ren
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China.
| | - Qingzhong Liu
- Laboratory Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhijiang Road, Shanghai, 200071, China.
| |
Collapse
|
50
|
Smirnova AV, Verbeke TJ, Furgason CC, Albakistani EA, Nwosu FC, Kim JJ, Haupt ES, Sheremet A, Lee ES, Trang E, Richardson E, Dacks JB, Dunfield PF. Microbial community development in an oil sands pit lake. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 987:179764. [PMID: 40460542 DOI: 10.1016/j.scitotenv.2025.179764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 05/06/2025] [Accepted: 05/24/2025] [Indexed: 06/16/2025]
Abstract
Surface mining and extraction of oil sands in Canada produces fluid tailings that contain several compounds of concern for the environment. One option for mine reclamation is the construction of Pit Lakes (PLs) to contain and remediate these tailings. Ultimately, PLs should support food webs typical of boreal lakes. From 2015 to 2021, we applied 16S/18S rRNA gene amplicon sequencing and metagenomics to monitor prokaryotic and eukaryotic microbes in the only full-scale PL of the oil sands industry (Base Mine Lake or BML), and compared it to two control environments: a freshwater reservoir unaffected by tailings, and active tailings ponds receiving regular industrial input. Microbial communities in BML were always intermediate to the two control environments based on alpha and beta diversity analyses. BML communities were highly variable with year, season, and water depth, and contained fewer core species than the freshwater reservoir. Several hydrocarbon degraders and sulfur cycling bacteria were identified as indicator species of tailings ponds, while several phototrophs were indicative of freshwater. However, all of these species were abundant in BML, suggesting that the PL supports food webs characteristic of each control environment. Over the 6-year study, the relative abundances of some common freshwater phytoplankton (Cryptomonas, Mychonastes, Trebouxiophyceae, Cyanobium) and heterotrophic bacteria (Sporichthyaceae, Ca. Fonsibacter, Ilumatobacteraceae, Microbacteriaceae, Ca. Planktophila) increased in BML. The results suggest that microbial communities and processes in BML represent an intermediate state between a tailings pond and a natural freshwater system, and did not stabilize within 10 years of its creation.
Collapse
Affiliation(s)
- Angela V Smirnova
- Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada
| | - Tobin J Verbeke
- Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada
| | - Chantel C Furgason
- Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada
| | - Emad A Albakistani
- Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada
| | - Felix C Nwosu
- Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada
| | - Joong-Jae Kim
- Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada
| | - Evan S Haupt
- Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada
| | - Andriy Sheremet
- Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada
| | - Eun-Suk Lee
- Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada
| | - Esther Trang
- Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada
| | - Elisabeth Richardson
- Division of Infectious Diseases, Department of Medicine, Department of Biological Sciences, 1-001 CCIS, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Joel B Dacks
- Division of Infectious Diseases, Department of Medicine, Department of Biological Sciences, 1-001 CCIS, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | - Peter F Dunfield
- Department of Biological Sciences, University of Calgary, 2500 University Dr. NW, Calgary, Alberta T2N 1N4, Canada.
| |
Collapse
|