1
|
Xiang M, Qiao L, Han Q, Zha Y, Sui X, Wang Q. Effects of Supplementation With Different Specificities of Dietary Fiber on Health-Related Indicators in Adults With Overweight or Obesity: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Nutr Rev 2025; 83:e1544-e1563. [PMID: 39821284 DOI: 10.1093/nutrit/nuae193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025] Open
Abstract
CONTEXT Dietary fiber (DF) exhibits variations in its chemical and physical complexity, as well as in its utilization by the gut microbiota. However, the impact of these differences on the health status of adults with overweight or obesity remains unclear. OBJECTIVE This meta-analysis aimed to explore the varying effects of supplementing with different specificities of DF on the health of adults with overweight or obesity, providing guidance on selecting DF supplementation to improve health status. DATA SOURCES The literature search encompassed 4 electronic databases-PubMed, Cochrane Library, Web of Science, and EMBASE-and was conducted between January 1, 2012, and November 10, 2023. Randomized controlled trials comparing DF with placebo treatment, without energy restriction, were included. DATA EXTRACTION Two independent reviewers extracted data using a standardized form, resolving discrepancies through discussion. The data included study characteristics, participant demographics, DF specifications, and outcome measures. DATA ANALYSIS Random-effects models and the generic inverse variance method were used to analyze data, assuming varying outcomes based on DF specificity. Meta-regression assessed the impact of population, duration, and dosage. Publication bias was evaluated using funnel plots and Egger's and Begg's tests. The analysis included 34 trials (n = 1804) examining DF supplementation at 1.5 to 40 g/day for 3 to 16 weeks. DF supplementation significantly reduced glycated hemoglobin (HbA1c) by 0.13%, fasting insulin by 0.82 μIU/mL, and homeostatic model assessment of insulin resistance (HOMA-IR) by 0.33 in adults with overweight or obesity. Subgroup analyses based on DF specificity revealed differences in effects on HbA1c, fasting insulin, and systolic blood pressure. The low-specificity subgroup showed significant heterogeneity in body weight, body mass index, HbA1c, fasting insulin, and HOMA-IR, with a decrease in fasting insulin by 1.09 μIU/mL. The low-to-intermediate-specificity subgroup had reductions in HbA1c by 0.8%, fasting insulin by 2.08 μIU/mL, and HOMA-IR by 0.61. The intermediate-specificity subgroup experienced a 2.85-kg decrease in body weight and a 9.03-mg/dL increase in LDL cholesterol. The mixed subgroup showed an increase in systolic blood pressure by 3.85 mmHg. CONCLUSION Supplementing with different specificities of DF may have distinct effects on health-related indicators in adults with overweight or obesity. Considering individuals' gut microbiota composition and specific health goals is recommended when selecting DF supplementation for adults with overweight or obesity. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42023432920.
Collapse
Affiliation(s)
- Mai Xiang
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
- Key Lab of Sports Nutrition, State General Administration of Sport of China, Beijing 100029, China
- College of Exercise Science, Beijing Sport University, Beijing 100084, China
| | - Li Qiao
- Beijing Competitor Sports Nutrition Research Institute, Beijing 100029, China
| | - Qi Han
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
- Key Lab of Sports Nutrition, State General Administration of Sport of China, Beijing 100029, China
| | - Yu Zha
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
- Key Lab of Sports Nutrition, State General Administration of Sport of China, Beijing 100029, China
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xuemei Sui
- Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC 29208, United States
| | - Qirong Wang
- Sports Nutrition Center, National Institute of Sports Medicine, Beijing 100029, China
- Key Lab of Sports Nutrition, State General Administration of Sport of China, Beijing 100029, China
| |
Collapse
|
2
|
Wang C, Dong T, Rong X, Yang Y, Mou J, Li J, Ge J, Mu X, Jiang J. Microbiome in prostate cancer: pathogenic mechanisms, multi-omics diagnostics, and synergistic therapies. J Cancer Res Clin Oncol 2025; 151:178. [PMID: 40450182 DOI: 10.1007/s00432-025-06187-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/31/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND Prostate cancer (PCa) is a leading cause of cancer-related deaths in men, with the microbiome emerging as a significant factor in its development and progression. Understanding the microbiome's role could provide new insights into PCa pathogenesis and treatment. OBJECTIVE This review aims to explore the interactions between the microbiome and PCa, focusing on microbial imbalances and their effects on immune responses, inflammation, and hormone levels. It also discusses advanced research techniques and the potential for microbiome modulation in PCa management. METHODS The review synthesizes current literature on the microbiome's role in PCa, highlighting differences in microbial composition between cancerous and healthy prostate tissues. It examines techniques such as high-throughput sequencing and metagenomics and explores the mechanisms through which the microbiome influences PCa. CONCLUSIONS The review reveals substantial microbial differences in prostate tissues of PCa patients compared to healthy individuals, indicating a potential link between microbiome alterations and disease progression. It highlights the promise of microbiome-based strategies for diagnosis and treatment and underscores the need for further research into personalized, microbiome-centric approaches for PCa management.
Collapse
Affiliation(s)
- Chengran Wang
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China
| | - Tianqi Dong
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China
| | - Xin'ao Rong
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China
| | - Yuce Yang
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China
| | - Jianhui Mou
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China
| | - Jiaqi Li
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China
| | - Jianli Ge
- Orthopaedic Medical Center, The Second Hospital of Jilin University, Changchun, 130041, Jilin Province, China
| | - Xupeng Mu
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China.
| | - Jinlan Jiang
- Department of' Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, , 130033, Jilin Province, China.
| |
Collapse
|
3
|
Bauch A, Baur J, Honold I, Willmann M, Weber GL, Müller S, Sodenkamp S, Peter S, Schoppmeier U, Laske C. Prognostic Value of a Multivariate Gut Microbiome Model for Progression from Normal Cognition to Mild Cognitive Impairment Within 4 Years. Int J Mol Sci 2025; 26:4735. [PMID: 40429881 PMCID: PMC12112180 DOI: 10.3390/ijms26104735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 05/07/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Little is known about the dysbiosis of the gut microbiome in patients with mild cognitive impairment (MCI) potentially at risk for the development of Alzheimer's disease (AD). So far, only cross-sectional differences and not longitudinal changes and their prognostic significance have been in the scope of research in MCI. Therefore, we investigated the ability of longitudinal taxonomic and functional gut microbiome data from 100 healthy controls (HC) to predict the progression from normal cognition to MCI over a 4-year follow-up period (4yFU). Logistic regression models were built with baseline features that best discriminated between the two groups using an ANOVA-type statistical analysis. The best model for the discrimination of MCI converters was based on functional data using Gene Ontology (GO), which included 14 features. This model achieved an area under the receiver operating characteristic curve (AUROC) of 0.84 at baseline, 0.78 at the 1-year follow-up (1yFU), and 0.75 at 4yFU. This functional model outperformed the taxonomic model, which included 38 genera features, in terms of descriptive performance and showed comparable efficacy to combined analyses integrating functional, taxonomic, and clinical characteristics. Thus, gut microbiome algorithms have the potential to predict MCI conversion in HCs over a 4-year period, offering a promising innovative supplement for early AD identification.
Collapse
Affiliation(s)
- Anne Bauch
- Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany; (I.H.); (G.L.W.); (S.M.); (S.S.); (C.L.)
| | - Julia Baur
- Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany; (I.H.); (G.L.W.); (S.M.); (S.S.); (C.L.)
| | - Iris Honold
- Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany; (I.H.); (G.L.W.); (S.M.); (S.S.); (C.L.)
| | - Matthias Willmann
- SYNLAB MVZ Leinfelden-Echterdingen GmbH, Labor Dr. Bayer, 70771 Leinfelden-Echterdingen, Germany;
| | - Greta Louise Weber
- Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany; (I.H.); (G.L.W.); (S.M.); (S.S.); (C.L.)
| | - Stephan Müller
- Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany; (I.H.); (G.L.W.); (S.M.); (S.S.); (C.L.)
| | - Sebastian Sodenkamp
- Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany; (I.H.); (G.L.W.); (S.M.); (S.S.); (C.L.)
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Silke Peter
- Institute of Medical Microbiology and Hygiene, University of Tübingen, 72076 Tübingen, Germany; (S.P.); (U.S.)
| | - Ulrich Schoppmeier
- Institute of Medical Microbiology and Hygiene, University of Tübingen, 72076 Tübingen, Germany; (S.P.); (U.S.)
| | - Christoph Laske
- Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany; (I.H.); (G.L.W.); (S.M.); (S.S.); (C.L.)
- German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Section for Dementia Research, Hertie Institute for Clinical Brain Research, Department of Psychiatry and Psychotherapy, University of Tübingen, 72076 Tübingen, Germany
| |
Collapse
|
4
|
Pallen MJ, Ponsero AJ, Telatin A, Moss CJ, Baker D, Heavens D, Davidson GL. Faecal metagenomes of great tits and blue tits provide insights into host, diet, pathogens and microbial biodiversity. Access Microbiol 2025; 7:000910.v3. [PMID: 40302838 PMCID: PMC12038002 DOI: 10.1099/acmi.0.000910.v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
Background. The vertebrate gut microbiome plays crucial roles in host health and disease. However, there is limited information on the microbiomes of wild birds, most of which is restricted to barcode sequences. We therefore explored the use of shotgun metagenomics on the faecal microbiomes of two wild bird species widely used as model organisms in ecological studies: the great tit (Parus major) and the Eurasian blue tit (Cyanistes caeruleus). Results. Short-read sequencing of five faecal samples generated a metagenomic dataset, revealing substantial variation in composition between samples. Reference-based profiling with Kraken2 identified key differences in the ratios of reads assigned to host, diet and microbes. Some samples showed high abundance of potential pathogens, including siadenoviruses, coccidian parasites and the antimicrobial-resistant bacterial species Serratia fonticola. From metagenome assemblies, we obtained complete mitochondrial genomes from the host species and from Isospora spp., while metagenome-assembled genomes documented new prokaryotic species. Conclusions. Here, we have shown the utility of shotgun metagenomics in uncovering microbial diversity beyond what is possible with 16S rRNA gene sequencing. These findings provide a foundation for future hypothesis testing and microbiome manipulation to improve fitness in wild bird populations. The study also highlights the potential role of wild birds in the dissemination of antimicrobial resistance.
Collapse
Affiliation(s)
- Mark J. Pallen
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- University of East Anglia, Norwich Research Park, Norwich, UK
| | | | - Andrea Telatin
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Cara-Jane Moss
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - David Baker
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Darren Heavens
- Earlham Institute, Norwich Research Park, Norwich, Norfolk, NR4 7UZ UK
| | - Gabrielle L. Davidson
- University of East Anglia, Norwich Research Park, Norwich, UK
- University of Cambridge, Downing Street, Cambridge, CB2 3EB, UK
| |
Collapse
|
5
|
Wang H, Su Q, Sun H, Meng Y, Xing X, Zheng H, Li Y. Unexpected Microbial and Genetic Diversity in the Gut of Chinese Giant Salamander. Integr Zool 2025. [PMID: 40165002 DOI: 10.1111/1749-4877.12976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/23/2025] [Accepted: 03/06/2025] [Indexed: 04/02/2025]
Abstract
The gut microbiome is crucial for animal health, yet the diversity of the critically endangered Chinese giant salamander's gut microbiota remains largely uncharacterized. In this study, we first conducted a comprehensive landscape survey of the gut microbiome of the Chinese giant salamander using 16S rRNA sequencing across a wide geographic range, identifying a distinct microbial cluster within its habitat. Subsequently, using shotgun metagenomes, we recovered 1518 metagenome-assembled genomes. Notably, 85% of the newly identified genomes could not be assigned to any known bacterial species, indicating a significant presence of novel taxa in Chinese giant salamander intestines. We observed substantial species-level variations in the gut microbiome across different age groups, with some novel species uniquely enriched in specific age populations. From the gut symbionts, we established a gene catalog comprising 3 278 107 non-redundant protein-coding genes, of which 7733 were annotated into recognized KEGG orthology groups. Additionally, we found that the gut microbiota of the Chinese giant salamander exhibits enhanced functional capacities explicitly in lipid metabolism and assimilatory sulfate reduction. Significant variations in the abundance of related enzyme-encoding genes across age groups suggest the unique roles of microbial metabolism in salamander health. By identifying microbial genomes and constructing an integrated gene catalog from metagenomic data, we significantly expand the resources available for research on the gut microbiome of the Chinese giant salamander, paving the way for further investigations into its ecological and health-related implications.
Collapse
Affiliation(s)
- Hongjian Wang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qinzhi Su
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Huihui Sun
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yujie Meng
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xinhui Xing
- Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Hao Zheng
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yiyuan Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
6
|
Príncipe LA, Marchi PH, Cesar CGL, Amaral AR, Duarte KKS, Finardi GLF, Souza JM, Balieiro JCC, Vendramini THA. Evaluation of enzymatically hydrolyzed poultry byproduct meal effects on fecal microbiota and pressure variables in elderly obese cats. Front Vet Sci 2025; 12:1530260. [PMID: 40191086 PMCID: PMC11969457 DOI: 10.3389/fvets.2025.1530260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/17/2025] [Indexed: 04/09/2025] Open
Abstract
Arterial hypertension is influenced by the intestinal microbiota and its metabolites, which play a crucial role in host health. Dietary peptides are multifunctional molecules with therapeutic potential for managing hypertension. This study aimed to evaluate the impact of incorporating enzymatically hydrolyzed poultry byproduct meal (EHPM-c) into extruded dry diets on the fecal microbiota and blood pressure parameters of elderly obese cats. Eighteen owners of neutered, clinically healthy male and female cats of various breeds were randomly assigned to two groups: control (30.8%, conventional poultry byproduct meal-CPM-c) and test (17.07%, CPM-c + 12.0% EHPM-c). Clinical values of systolic blood pressure, serum aldosterone concentrations, angiotensin-converting enzyme I activity, and fecal microbiota using 16S rRNA were measured. Data were processed using SAS software (PROC MIXED, PROC GLIMMIX, and PROC CORR; p < 0.05). Both groups exhibited high microbial alpha diversity, with no significant differences in beta diversity. Although the inclusion of 12.0% EHPM-c had no measurable effect on blood pressure, both diets promoted beneficial modulation of the fecal microbiota, improving intestinal health. These findings underscore the importance of diet in maintaining gut homeostasis in obese senior cats. While the inclusion of 12.0% EHPM-c did not significantly alter blood pressure parameters, the modulation of the fecal microbiota suggests a potential role in maintaining intestinal health. These results highlight the need for further studies to explore different inclusion levels and longer intervention periods.
Collapse
Affiliation(s)
- Leonardo A. Príncipe
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Pedro H. Marchi
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Cinthia G. L. Cesar
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Andressa R. Amaral
- Veterinary Nutrology Service, Veterinary Teaching Hospital, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | - Kelly K. S. Duarte
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Gabriela L. F. Finardi
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Jennifer M. Souza
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Júlio C. C. Balieiro
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
| | - Thiago H. A. Vendramini
- Pet Nutrology Research Center, Department of Animal Nutrition and Production, School of Veterinary Medicine and Animal Science, University of São Paulo, Pirassununga, Brazil
- Veterinary Nutrology Service, Veterinary Teaching Hospital, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
7
|
Zhra M, Elahi MA, Tariq A, Abu-Zaid A, Yaqinuddin A. Sirtuins and Gut Microbiota: Dynamics in Health and a Journey from Metabolic Dysfunction to Hepatocellular Carcinoma. Cells 2025; 14:466. [PMID: 40136715 PMCID: PMC11941559 DOI: 10.3390/cells14060466] [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: 01/27/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction leading to non-alcoholic fatty liver disease (NAFLD) exhibits distinct molecular and immune signatures that are influenced by factors like gut microbiota. The gut microbiome interacts with the liver via a bidirectional relationship with the gut-liver axis. Microbial metabolites, sirtuins, and immune responses are pivotal in different metabolic diseases. This extensive review explores the complex and multifaceted interrelationship between sirtuins and gut microbiota, highlighting their importance in health and disease, particularly metabolic dysfunction and hepatocellular carcinoma (HCC). Sirtuins (SIRTs), classified as a group of NAD+-dependent deacetylases, serve as crucial modulators of a wide spectrum of cellular functions, including metabolic pathways, the inflammatory response, and the process of senescence. Their subcellular localization and diverse functions link them to various health conditions, including NAFLD and cancer. Concurrently, the gut microbiota, comprising diverse microorganisms, significantly influences host metabolism and immune responses. Recent findings indicate that sirtuins modulate gut microbiota composition and function, while the microbiota can affect sirtuin activity. This bidirectional relationship is particularly relevant in metabolic disorders, where dysbiosis contributes to disease progression. The review highlights recent findings on the roles of specific sirtuins in maintaining gut health and their implications in metabolic dysfunction and HCC development. Understanding these interactions offers potential therapeutic avenues for managing diseases linked to metabolic dysregulation and liver pathology.
Collapse
Affiliation(s)
- Mahmoud Zhra
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Muhammad Affan Elahi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.A.E.); (A.A.-Z.)
| | - Aamira Tariq
- Department of Biosciences, COMSATS University Islamabad, Islamabad Campus, Islamabad 45550, Pakistan
| | - Ahmed Abu-Zaid
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.A.E.); (A.A.-Z.)
| | - Ahmed Yaqinuddin
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| |
Collapse
|
8
|
Plesz SB, Adlan LG, Büki A, Makra N, Ligeti B, Ágg B, Szabó D, Zádori ZS, Ferdinandy P, Horvath G, Kekesi G. Dysbiosis is associated with the behavioral phenotype observed in the triple-hit Wisket rat model of schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111276. [PMID: 39880276 DOI: 10.1016/j.pnpbp.2025.111276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 01/16/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
Comorbidities between gastrointestinal diseases and psychiatric disorders have been widely reported, with the gut-brain axis implicated as a potential biological basis. Thus, dysbiosis may play an important role in the etiology of schizophrenia, which is barely detected. Triple-hit Wisket model rats exhibit various schizophrenia-like behavioral phenotypes. The present study aimed to compare the diversity and abundance of gut microbiota in Wisket model and control rats; furthermore, to correlate the microbial taxonomic profiles to indices of behavioral change. Tail-flick and Ambitus tests were used to assess acute heat pain sensitivity, and record exploration and locomotor activity along with motivation in young adult, control and Wisket model rats. Fecal microbiota composition was profiled by deep sequencing of bacterial 16S rRNA, and it was correlated to behavioral phenotype. Wisket rats exhibited significantly decreased pain sensitivity, lower locomotor activity and exploration, and impaired motivation compared with controls. No significant differences were observed in bacterial alpha diversity between the groups; however, clear differences in community structure were observed. Wisket rats showed decreases in several genera of Firmicutes and Saccharimonas, and increases in Bacteriodetes and Helicobacter phyla compared with controls. Correlation analysis revealed significant associations between the microbiota profile and the behavioral phenotype. This is the first demonstration that fecal microbiota composition is markedly altered in a triple-hit schizophrenia rat model, suggesting the contribution of the microbiota-gut-brain axis in the development of the schizophrenia-like behavioral phenotype. Thus targeting the gut microbiota may be a novel approach to treat such impairments.
Collapse
Affiliation(s)
- Szonja B Plesz
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.
| | - Leatitia G Adlan
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.
| | - Alexandra Büki
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.
| | - Nóra Makra
- HUN-REN Human Microbiota Study Group, Institute of Medical Microbiology, Semmelweis University, Budapest, Hungary.
| | - Balázs Ligeti
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary.
| | - Bence Ágg
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary.
| | - Dóra Szabó
- HUN-REN Human Microbiota Study Group, Institute of Medical Microbiology, Semmelweis University, Budapest, Hungary.
| | - Zoltán S Zádori
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
| | - Péter Ferdinandy
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary.
| | - Gyongyi Horvath
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.
| | - Gabriella Kekesi
- Department of Physiology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.
| |
Collapse
|
9
|
Liu N, Zhang Z, Zhang J, Ma W, Wang C. Effects of N-Carbamylglutamate supplementation on cecal morphology, microbiota composition, and short-chain fatty acids contents of broiler breeder roosters. Sci Rep 2025; 15:7489. [PMID: 40032942 PMCID: PMC11876567 DOI: 10.1038/s41598-025-91577-w] [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: 12/11/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
The objective of this study was to assess the effects of N-Carbamylglutamate (NCG) supplementation on cecal morphology, microbiota composition, and short-chain fatty acids (SCFAs) contents in broiler breeder roosters. A total of 72 11-week-old Zhuanghe Dagu broiler breeder roosters with a similar initial body weight (1.53 ± 0.06 kg) were randomly allocated into two groups. Each group had 3 replicates with 12 birds per replicate. The experimental period lasted 42 days. All birds underwent the same production practices, except for the dietary conditions. It was found that an increase in cecal muscularis thickness and villi epithelium thickness. The NCG supplementation was found to have regulatory effects on the composition of cecal microbiota. Additionally, the study observed an increase in the content of butyric acid in the cecum of broiler breeder roosters fed with the NCG-containing control diet compared to those fed with the basal diet. Spearman correlation analysis showed that the variation of cecal microbiota was closely related to the production of butyric acid as well as the improvement of muscularis and villi epithelium thickness in cecum. The increase of butyric acid content in cecum was positively correlated with the improvement of cecal muscularis and villi epithelium thickness. In conclusion, the findings of this study indicate that dietary supplementation of NCG in broiler breeder roosters can positively influence cecal morphology, microbiota composition, and butyric production.
Collapse
Affiliation(s)
- Na Liu
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, 121001, China
| | - Zhe Zhang
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, 121001, China
| | - Jiehui Zhang
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, 121001, China
| | - Wei Ma
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, 121001, China
| | - Chunqiang Wang
- College of Animal Science and Veterinary Medicine, Jinzhou Medical University, Jinzhou, 121001, China.
| |
Collapse
|
10
|
Blanquet L, Serra D, Marrinhas C, Almeida A. Exploring Gut Microbiota-Targeted Therapies for Canine Idiopathic Epilepsy. Int J Mol Sci 2025; 26:1742. [PMID: 40004205 PMCID: PMC11855791 DOI: 10.3390/ijms26041742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Epilepsy stands out as one of the most prevalent chronic neurological conditions affecting companion animals. Recent research has increasingly focused on exploring the role of gut microbiota in influencing neurological conditions, like epilepsy. This influence stems from the bidirectional communication pathways between gut bacteria and the brain, which involve metabolic, neural, immunological, and endocrine mechanisms. In fact, a balanced and stable gut microbiota is essential to maintaining normal gut physiology and ensuring appropriate signaling along the gut-brain axis. Conversely, dysbiosis can have detrimental effects on gut physiology and may contribute to the development or exacerbation of neurological conditions, including epilepsy. Considering these findings, this review article aims to deepen the understanding of the mechanisms underlying the microbiota-gut-brain connection in the context of canine idiopathic epilepsy. Moreover, this review presents recent data on innovative gut-related therapeutic strategies for canine idiopathic epilepsy treatment.
Collapse
Affiliation(s)
- Luann Blanquet
- EUVG—Escola Universitária Vasco de Gama, Campus Universitário, Av. José R. Sousa Fernandes 197, 3020-210 Coimbra, Portugal
| | - Diana Serra
- EUVG—Escola Universitária Vasco de Gama, Campus Universitário, Av. José R. Sousa Fernandes 197, 3020-210 Coimbra, Portugal
- CIVG—Centro de Investigação Vasco da Gama, EUVG—Escola Universitária Vasco da Gama, Campus Universitário-Bloco B, Av. José R. Sousa Fernandes 197, 3020-210 Coimbra, Portugal
- CNC—Centro de Neurociências e Biologia Celular, Universidade de Coimbra, 3004-531 Coimbra, Portugal
| | - Carla Marrinhas
- Onevet Hospital Veterinário do Baixo Vouga, Estrada Nacional 1, 355, 3750-742 Águeda, Portugal
| | - Anabela Almeida
- EUVG—Escola Universitária Vasco de Gama, Campus Universitário, Av. José R. Sousa Fernandes 197, 3020-210 Coimbra, Portugal
- CIVG—Centro de Investigação Vasco da Gama, EUVG—Escola Universitária Vasco da Gama, Campus Universitário-Bloco B, Av. José R. Sousa Fernandes 197, 3020-210 Coimbra, Portugal
- CIBIT/ICNAS—Instituto de Imagem Biomédica e Investigação Translacional de Coimbra, Universidade de Coimbra, Polo 3, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| |
Collapse
|
11
|
Yan Q, Chen Y, Gao EB, Lu Y, Wu J, Qiu H. The characteristics of intestinal microflora in infants with rotavirus enteritis, changes in microflora before and after treatment and their clinical values. Sci Rep 2025; 15:4312. [PMID: 39910252 PMCID: PMC11799311 DOI: 10.1038/s41598-025-88312-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/28/2025] [Indexed: 02/07/2025] Open
Abstract
Rotavirus (RV) is a leading pathogen causing diarrhea in children. In this study, a total of 51 fecal samples from children with RV enteritis, 29 post-treatment fecal samples, and 38 fecal samples from age-matched healthy controls were collected. Microbial DNA was isolated from the samples followed by high throughput Illumina sequencing targeting 16 S rRNA gene. Compared to the healthy group, the RV-infected group exhibited reduced microbial diversity. Both groups shared Firmicutes as the dominant phylum. Additionally, the abundance of Proteobacteria increased significantly in the RV-infected group. At the genus level, among the top 50 most abundant genera, 34 showed significant differences, with these differential genera correlating with certain clinical indicators such as dehydration levels and C-reactive protein (CRP). Notably, there were no significant differences in the microbiota before and after treatment in RV-infected children. Only 8.82% (3/34) of the differential genera in the post-treatment group showed a recovery trend towards the healthy state. This study enhances the understanding of how RV infection alters the gut microbiota structure in children and provides a scientific basis for improving clinical diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Qiyan Yan
- Department of Pediatrics, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Yuanyuan Chen
- Department of Pediatrics, Jinhua Women's and Children's Hospital, Jinhua, 321000, Zhejiang, China
| | - E-Bin Gao
- School of Life Sciences, Jiangsu University, Zhenjiang, 212000, Jiangsu, China
| | - Yanbo Lu
- Department of Pediatrics, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Junhua Wu
- Department of Pediatrics, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China.
| | - Haiyan Qiu
- Department of Pediatrics, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China.
| |
Collapse
|
12
|
Ayyanar MP, Vijayan M. A review on gut microbiota and miRNA crosstalk: implications for Alzheimer's disease. GeroScience 2025; 47:339-385. [PMID: 39562408 PMCID: PMC11872870 DOI: 10.1007/s11357-024-01432-5] [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: 09/23/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and progressive neuronal damage. Recent research has highlighted the significant roles of the gut microbiota and microRNAs (miRNAs) in the pathogenesis of AD. This review explores the intricate interaction between gut microbiota and miRNAs, emphasizing their combined impact on Alzheimer's progression. First, we discuss the bidirectional communication within the gut-brain axis and how gut dysbiosis contributes to neuroinflammation and neurodegeneration in AD. Changes in gut microbiota composition in Alzheimer's patients have been linked to inflammation, which exacerbates disease progression. Next, we delve into the biology of miRNAs, focusing on their roles in gene regulation, neurodevelopment, and neurodegeneration. Dysregulated miRNAs are implicated in AD pathogenesis, influencing key processes like inflammation, tau pathology, and amyloid deposition. We then examine how the gut microbiota modulates miRNA expression, particularly in the brain, potentially altering neuroinflammatory responses and synaptic plasticity. The interplay between gut microbiota and miRNAs also affects blood-brain barrier integrity, further contributing to Alzheimer's pathology. Lastly, we explore therapeutic strategies targeting this gut microbiota-miRNA axis, including probiotics, prebiotics, and dietary interventions, aiming to modulate miRNA expression and improve AD outcomes. While promising, challenges remain in fully elucidating these interactions and translating them into effective therapies. This review highlights the importance of understanding the gut microbiota-miRNA relationship in AD, offering potential pathways for novel therapeutic approaches aimed at mitigating the disease's progression.
Collapse
Affiliation(s)
- Maruthu Pandian Ayyanar
- Department of Biology, The Gandhigram Rural Institute (Deemed to be University), Gandhigram, 624302, Tamil Nadu, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| |
Collapse
|
13
|
Veerapandian R, Paudyal A, Schneider SM, Lee STM, Vediyappan G. A mouse model of immunosuppression facilitates oral Candida albicans biofilms, bacterial dysbiosis and dissemination of infection. Front Cell Infect Microbiol 2025; 14:1467896. [PMID: 39902181 PMCID: PMC11788080 DOI: 10.3389/fcimb.2024.1467896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 12/26/2024] [Indexed: 02/05/2025] Open
Abstract
Opportunistic pathogens are a major threat to people, especially those with impaired immune systems. Two of the most important microbes in this category are the fungus Candida albicans and Gram-positive bacteria of the genus Enterococcus, which share overlapping niches in the oral cavity, gastrointestinal and urogenital tracts. The clinical importance of oral C. albicans biofilm and its interaction with the host under immunosuppressive conditions remains largely understudied. Here, we used a mouse model of oropharyngeal candidiasis (OPC) with cortisone acetate injection on alternate days and a continuous supply of C. albicans in drinking water for three days, resulting in immunosuppression. Results showed abundant growth of resident oral bacteria and a strong C. albicans biofilm on the tongue consisting of hyphae which damaged papillae, the epidermal layer, and invaded tongue tissue with the accumulation of inflammatory cells as demonstrated by Grocott's methenamine silver and hematoxylin and eosin staining, respectively. The dispersed microbes from the oral biofilm colonized the gastrointestinal (GI) tract and damaged its integrity, disseminating microbes to other organs. Although no visible damage was observed in the kidney and liver, except increased lipid vacuoles in the liver cells, C. albicans was found in the liver homogenate. Intriguingly, we found co-occurrence of Enterococcus faecalis in the tongue, liver, and stool of immunosuppressed control and C. albicans infected organs. Targeted 16S rRNA and ITS2 amplicon sequencing of microbes from the fecal samples of mice confirmed the above results in the stool samples and revealed an inverse correlation of beneficial microbes in the dysbiosis condition. Our study shows that mucosal-oral infection of C. albicans under immunosuppressed conditions causes tissue damage and invasion in local and distant organs; the invasion may be aided by the overgrowth of the resident endogenous Enterobacteriaceae and other members, including the opportunistic pathogen Enterococcus faecalis.
Collapse
Affiliation(s)
- Raja Veerapandian
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Anuja Paudyal
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Sarah M. Schneider
- Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, United States
| | - Sonny T. M. Lee
- Division of Biology, Kansas State University, Manhattan, KS, United States
| | - Govindsamy Vediyappan
- Division of Biology, Kansas State University, Manhattan, KS, United States
- Diagnostic Medicine and Pathobiology, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
14
|
Ying J, Zhang MW, Wei KC, Wong SH, Subramaniam M. Influential articles in autism and gut microbiota: bibliometric profile and research trends. Front Microbiol 2025; 15:1401597. [PMID: 39850141 PMCID: PMC11755156 DOI: 10.3389/fmicb.2024.1401597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025] Open
Abstract
Objective Autism spectrum disorder (ASD) is a common neurodevelopmental disorder. Increasing evidence suggests that it is potentially related to gut microbiota, but no prior bibliometric analysis has been performed to explore the most influential works in the relationships between ASD and gut microbiota. In this study, we conducted an in-depth analysis of the most-cited articles in this field, aiming to provide insights to the existing body of research and guide future directions. Methods A search strategy was constructed and conducted in the Web of Science database to identify the 100 most-cited papers in ASD and gut microbiota. The Biblioshiny package in R was used to analyze and visualize the relevant information, including citation counts, country distributions, authors, journals, and thematic analysis. Correlation and comparison analyses were performed using SPSS software. Results The top 100 influential manuscripts were published between 2000 and 2021, with a total citation of 40,662. The average number of citations annually increased over the years and was significantly correlated to the year of publication (r = 0.481, p < 0.01, Spearman's rho test). The United States was involved in the highest number of publications (n = 42). The number of publications in the journal was not significantly related to the journal's latest impact factor (r = 0.016, p > 0.05, Spearman's rho test). Co-occurrence network and thematic analysis identified several important areas, such as microbial metabolites of short-chain fatty acids and overlaps with irritable bowel syndrome. Conclusion This bibliometric analysis provides the key information of the most influential studies in the area of ASD and gut microbiota, and suggests the hot topics and future directions. The findings of this study can serve as a valuable reference for researchers and policymakers, guiding the development and implementation of the scientific research strategies in this area.
Collapse
Affiliation(s)
- Jiangbo Ying
- Department of Developmental Psychiatry, Institute of Mental Health, Singapore, Singapore
| | | | - Ker-Chiah Wei
- Department of Developmental Psychiatry, Institute of Mental Health, Singapore, Singapore
| | - Sunny H. Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Gastroenterology and Hepatology, Tan Tock Seng Hospital, Singapore, Singapore
| | | |
Collapse
|
15
|
Doyle C, Wall K, Fanning S, McMahon BJ. Making sense of sentinels: wildlife as the One Health bridge for environmental antimicrobial resistance surveillance. J Appl Microbiol 2025; 136:lxaf017. [PMID: 39805713 DOI: 10.1093/jambio/lxaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/25/2024] [Accepted: 01/12/2025] [Indexed: 01/16/2025]
Abstract
Antimicrobial resistance (AMR), arising from decades of imprudent anthropogenic use of antimicrobials in healthcare and agriculture, is considered one of the greatest One Health crises facing healthcare globally. Antimicrobial pollutants released from human-associated sources are intensifying resistance evolution in the environment. Due to various ecological factors, wildlife interact with these polluted ecosystems, acquiring resistant bacteria and genes. Although wildlife are recognized reservoirs and disseminators of AMR in the environment, current AMR surveillance systems still primarily focus on clinical and agricultural settings, neglecting this environmental dimension. Wildlife can serve as valuable sentinels of AMR in the environment, reflecting ecosystem health, and the effectiveness of mitigation strategies. This review explores knowledge gaps surrounding the ecological factors influencing AMR acquisition and dissemination in wildlife, and highlights limitations in current surveillance systems and policy instruments that do not sufficiently address the environmental component of AMR. We discuss the underutilized opportunity of using wildlife as sentinel species in a holistic, One Health-centred AMR surveillance system. By better integrating wildlife into systematic AMR surveillance and policy, and leveraging advances in high-throughput technologies, we can track and predict resistance evolution, assess the ecological impacts, and better understand the complex dynamics of environmental transmission of AMR across ecosystems.
Collapse
Affiliation(s)
- Caoimhe Doyle
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
- UCD-Centre for Food Safety, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Katie Wall
- UCD-Centre for Food Safety, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Séamus Fanning
- UCD-Centre for Food Safety, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Belfield, Dublin 4, Ireland
| | - Barry J McMahon
- UCD School of Agriculture and Food Science, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
16
|
Swanson KS, Allenspach K, Amos G, Auchtung TA, Bassett SA, Bjørnvad CR, Everaert N, Martín-Orúe SM, Ricke SC, Ryan EP, Fahey GC. Use of biotics in animals: impact on nutrition, health, and food production. J Anim Sci 2025; 103:skaf061. [PMID: 40036559 PMCID: PMC12010704 DOI: 10.1093/jas/skaf061] [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: 09/30/2024] [Accepted: 02/26/2025] [Indexed: 03/06/2025] Open
Abstract
Probiotics, prebiotics, and other biotic substances are not only effective ways to promote a healthy gastrointestinal tract, an effective immune system, and the overall health of humans, but also in agricultural and companion animals. Because key differences exist in regard to gastrointestinal tract anatomy and physiology, dietary management and feeding strategy, and disease susceptibility, however, biotic types and amounts often differ according to host species and life stage. Despite these differences, the literature demonstrates the value of biotics in agricultural and companion animal species. While high variability in responsiveness and efficacy has been reported, biotic substances may be effectively used to improve digestion, reduce morbidity, increase growth rate and/or efficiency in agricultural animals and promote gastrointestinal health and immune response in companion animals. As the oversight of antibiotic use intensifies, the population density of animals and humans increases, and production strategies of agricultural animals are more heavily scrutinized, the importance of biotics and other health promotors will continue to increase in the future. To date, the effects of animal biotic use have focused primarily on the farm, home, or veterinary clinic. In the future, their impact must be viewed on a larger scale. As global "One Health" approaches seek to reduce antimicrobial use and resistance and there are increasing demands for sustainable and safe food production, biotics will continue to be an important part of the solution. As knowledge of gastrointestinal microbiomes grows and the biotic field develops, more targeted and effective strategies for health promotion in these species are expected. At the 2023 International Scientific Association for Probiotics and Prebiotics meeting, experts were invited to participate in a discussion group focused on "The Use of Probiotics and Prebiotics in Agricultural and Companion Animals". This review reports the outcomes of that discussion, including the documented use of probiotics, prebiotics, and other biotic substances to promote health or treat disease in agricultural and companion animals, provide implications of animal biotic use on human health, and provide perspective on how scientific advances may impact the development and improvement of biotics in the future.
Collapse
Affiliation(s)
- Kelly S Swanson
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Karin Allenspach
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Gregory Amos
- Waltham Petcare Science Institute, Melton Mowbray LE13, UK
| | | | - Shalome A Bassett
- Fonterra Limited, Fonterra Research & Development Centre, Palmerston North 4442, New Zealand
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand
| | - Charlotte R Bjørnvad
- Department of Veterinary Clinical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Susana M Martín-Orúe
- Department of Animal and Food Science, Animal Nutrition and Welfare Service, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Steven C Ricke
- Department of Animal and Dairy Sciences, Meat Science and Animal Biologics Discovery Program, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Elizabeth P Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO 80524, USA
| | - George C Fahey
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
17
|
Castañeda S, Poveda C, Suarez-Reyes C, Wu Y, Haugen N, Patiño LH, Weatherhead JE, Ramírez JD. Microbiota dynamics during Ascaris suum larval migration: Implications for host microbial communities in a murine model. Microb Pathog 2025; 198:107122. [PMID: 39549928 DOI: 10.1016/j.micpath.2024.107122] [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: 07/29/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
The complex interactions between parasites, their hosts, and associated microbiota hold significant implications for host health and disease outcomes. Helminths like Ascaris lumbricoides and Ascaris suum can significantly alter the host's intestinal microbiota, affecting both parasite biology and host pathology. Despite extensive research on host-microbiota changes due to helminth infections, the study of helminth-associated microbiota remains limited. This study aims to characterize the microbiota associated with Ascaris larvae and surrounding host tissues at distinct developmental stages (day 4, day 8, day 14), during larval migration through the liver, lungs, and intestine, and its impact on the host's microbiota in a murine model. Twenty mice were infected with 2500 embryonated A. suum eggs via oral gavage. Five Ascaris-infected mice and age-matched naïve mice were euthanized at 4-, 8-, and 14-days post-infection (DPI). Stool, intestine, liver, and lung samples were collected. Larvae were isolated from embryonated eggs in vitro, from the liver at 4 DPI, and the lung at 8 DPI. Utilizing 16S rRNA sequencing, we analyzed bacterial diversity in samples from different Ascaris stages and host tissues. Our results revealed a total of 8040 amplicon sequence variants (ASVs) with Ascaris samples displaying the highest diversity. Notably, Ascaris-larvae associated microbiota differed significantly from that of the host, with higher diversity observed in the parasite. Differential abundance analysis identified distinct taxonomic patterns, highlighting specific genera such as Bradyrhizobium, Achromobacter, and Pseudomonas in Ascaris. Our findings suggest that Ascaris harbors a unique microbiota that potentially exchanges bacteria with the host during larval migration. These insights pave the way for further research into the ecological and functional dynamics of helminth-microbiota interactions, which may inform novel therapeutic strategies targeting these microbial relationships to mitigate helminth infections and improve host health outcomes.
Collapse
Affiliation(s)
- Sergio Castañeda
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Cristina Poveda
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA; Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA; National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Charlie Suarez-Reyes
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Yifan Wu
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Noah Haugen
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Luz H Patiño
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Jill E Weatherhead
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA.
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia; Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
18
|
Bruno P, Schüler T, Rosshart SP. Born to be wild: utilizing natural microbiota for reliable biomedical research. Trends Immunol 2025; 46:17-28. [PMID: 39690004 DOI: 10.1016/j.it.2024.11.013] [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: 10/11/2024] [Revised: 11/11/2024] [Accepted: 11/19/2024] [Indexed: 12/19/2024]
Abstract
Laboratory mice housed under specific pathogen-free (SPF) conditions are the standard model in biomedical research. However, experiments with a particular inbred mouse strain performed in different laboratories often yield inconsistent or conflicting data due to housing-specific variations in the composition and diversity of SPF microbiota. These variations affect immune and nonimmune cell functions, leading to systemic physiological changes. Consequently, microbiota-dependent inconsistencies have raised general doubts regarding the suitability of mice as model organisms. Since stability positively correlates with biological diversity, we postulate that increasing species diversity can improve microbiota stability and mouse physiology, enhancing robustness, reproducibility, and experimental validity. Similar to the generation of inbred mouse strains in the last century, we suggest a worldwide initiative to define a transplantable 'wild' microbiota that stably colonizes mice irrespective of housing conditions.
Collapse
Affiliation(s)
- Philipp Bruno
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany.
| | - Stephan P Rosshart
- Department of Microbiome Research, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany; Department of Medicine II, Faculty of Medicine, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
19
|
Veerapagu M, Jeya K, Sankara Narayanan A. Gastrointestinal microbiome engineering in pig. HUMAN AND ANIMAL MICROBIOME ENGINEERING 2025:265-290. [DOI: 10.1016/b978-0-443-22348-8.00016-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
20
|
Lai Y, Huang X, Sun H, Hui Q, Hu S. Research Progress in the Relationship between Intestinal Flora and Diabetes Mellitus. Endocr Metab Immune Disord Drug Targets 2025; 25:281-290. [PMID: 38956918 DOI: 10.2174/0118715303308965240624054156] [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: 02/06/2024] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024]
Abstract
Diabetes mellitus is a common chronic metabolic disease characterized by a high incidence and disability rate. Intestinal flora refers to the microbial community that lives in the intestines and plays a crucial role in maintaining intestinal health and the human immune system. In recent years, an increasing body of research has revealed a close relationship between intestinal flora and diabetes. The pathophysiological mechanisms between them have also been constantly uncovered, and the regulation of intestinal flora has shown promising efficacy in the adjuvant treatment of diabetes. This study mainly summarized the characteristics and mechanisms of intestinal flora in patients with diabetes in recent years, as well as the methods of regulating intestinal flora to prevent and treat diabetes, and prospected the future research direction. This will offer a theoretical basis for the clinical adjuvant treatment of diabetes with intestinal flora and the development of new drugs.
Collapse
Affiliation(s)
- Yingji Lai
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xianfeng Huang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hongwei Sun
- Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qi Hui
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shanshan Hu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
21
|
Wu Z, Bian M, Zhang H, Wang M, Wang P, Shao Y, Shen L, Zhu G. Compositional characteristics of the gut microbiome in patients with uremia. INDIAN J PATHOL MICR 2025; 68:42-50. [PMID: 39011618 DOI: 10.4103/ijpm.ijpm_554_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 04/05/2024] [Indexed: 07/17/2024] Open
Abstract
ABSTRACT During acute or chronic uremia, the cumulative harmful effects of uremic toxins result in numerous health problems and, ultimately, mortality. Previous research has identified that uremic retention solutes originate from the gut microbiome, indicating that uremia may be closely associated with gut microbiome dysbiosis. To deepen our understanding of the compositional characteristics of the gut microbiome in patients with uremia and thereby promote precision medicine in the treatment of uremia, we conducted a study of the compositional characteristics of the gut microbiome in 20 patients with uremia. The gut microbiome diversity of uremic patients and the control group showed certain differences. Nonmetric multidimensional scaling analysis showed that the beta diversity of the gut microbiome of uremic patients was significantly different from that of the healthy control individuals, with a distinct clustering effect in the uremic patient group, and it also showed a similarly distinct clustering effect in the healthy control group. The Chao1 index and Sobs index were significantly lower in the uremic patient group than in the healthy control group ( P < 0.05). By analyzing the composition and abundance distribution of the gut microbiome in the uremic patient group and healthy control group, we found that the relative abundance of the gut microbiome constituents Fusobacteriota , Enterobacteriaceae, Oscillospirales, Ruminococcaceae, and Lachnospiraceae was significantly increased in the intestines of uremic patients. We also detected the rare taxa Erysipelotrichaceae, which was present only in the uremic patient group. Predictive functional analysis suggested that an increased abundance of Ruminococcaceae and Lachnospirales, which are associated with indoxyl sulfate and phenylacetyl glutamine, and an increased abundance of Oscillospirales, which is associated with pyruvate metabolism, in uremic patients may strongly influence the gut environment according to renal function, resulting in dysbiosis associated with uremic toxin production. Rare taxa such as Erysipelotrichaceae have been suggested to be detrimental to intestinal disease. Further research into these gut microbiomes may provide new ideas for the prevention and treatment of uremia with the gut microbiome.
Collapse
Affiliation(s)
- Zirui Wu
- College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Mingjie Bian
- College of Life Sciences, Anhui Normal University, Wuhu, China
- Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, and Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, Anhui Normal University, Wuhu, Anhui, China
| | - Hong Zhang
- College of Life Sciences, Anhui Normal University, Wuhu, China
- Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, and Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, Anhui Normal University, Wuhu, Anhui, China
| | - Mengli Wang
- College of Life Sciences, Anhui Normal University, Wuhu, China
- Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, and Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, Anhui Normal University, Wuhu, Anhui, China
| | - Peng Wang
- College of Life Sciences, Anhui Normal University, Wuhu, China
- Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, and Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, Anhui Normal University, Wuhu, Anhui, China
| | - Yunxia Shao
- College of Life Sciences, Anhui Normal University, Wuhu, China
- Second People's Hospital of Wuhu, Anhui, China
- Department of Neohrology, Wuhu Hospital, East China Normal University, Wuhu, China
| | - Liang Shen
- College of Life Sciences, Anhui Normal University, Wuhu, China
- Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, and Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, Anhui Normal University, Wuhu, Anhui, China
| | - Guoping Zhu
- College of Life Sciences, Anhui Normal University, Wuhu, China
- Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, and Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, Anhui Normal University, Wuhu, Anhui, China
| |
Collapse
|
22
|
Cui L, Xie Y, Luo K, Wang M, Liu L, Li C, Tian X. Physiological and intestinal microbiota responses of sea cucumber Apostichopus japonicus to various stress and signatures of intestinal microbiota dysbiosis. Front Microbiol 2024; 15:1528275. [PMID: 39780943 PMCID: PMC11708840 DOI: 10.3389/fmicb.2024.1528275] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Identifying the signatures of intestinal dysbiosis caused by common stresses is fundamental to establishing efficient health monitoring strategies for sea cucumber. This study investigated the impact of six common stress experienced frequently in aquaculture on the growth performance, intestinal homeostasis and microbiota of sea cucumber, including thermal (23°C), hypoosmotic (22‰ salinity), ammonium (0.5 mg/L NH4 +-N), and nitrite (0.25 mg/L NO2 --N) stress exposure for 30 days, as well as starvation and crowding (6 kg/m3 density) stress exposure for 60 days. Results demonstrated that all stress led to reduced growth performance and digestive capacity of sea cucumber, along with varying degrees of oxidative stress and immune responses. Various stresses significantly altered the diversity, community structure (except for crowding stress), and composition of intestinal microbiota. The ratios of Bacteroidota: Proteobacteria (B: P) and Firmicutes: Proteobacteria (F: P) declined markedly compared to the control. Potentially pathogenic bacteria of Shewanellaceae, Vibrionaceae, and Moraxellaceae significantly increased under crowding, ammonium, and nitrite stress, respectively, whereas beneficial microbes of Achromobacter and Rhodobacteraceae were, respectively, enriched under hypoosmotic and starvation stresses. The complexity and stability of microbial ecological networks were further altered by these stresses. KEGG predictions revealed the reduced functional pathways of intestinal microbiota involved in host immunity under different stresses. Correlation analysis further confirmed a strong link between microbiota response and host immunity under different stresses. The increased abundance of Verrucomicrobia species could also be identified as the sensitive indicator for diagnosing whether the host was under stressful pressure by random forest analysis.
Collapse
Affiliation(s)
- Liang Cui
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Yumeng Xie
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Kai Luo
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Mingyang Wang
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Longzhen Liu
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
- The Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Changlin Li
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| | - Xiangli Tian
- The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, China
| |
Collapse
|
23
|
Rooney J, Rivera-de-Torre E, Li R, Mclean K, Price DR, Nisbet AJ, Laustsen AH, Jenkins TP, Hofmann A, Bakshi S, Zarkan A, Cantacessi C. Structural and functional analyses of nematode-derived antimicrobial peptides support the occurrence of direct mechanisms of worm-microbiota interactions. Comput Struct Biotechnol J 2024; 23:1522-1533. [PMID: 38633385 PMCID: PMC11021794 DOI: 10.1016/j.csbj.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
The complex relationships between gastrointestinal (GI) nematodes and the host gut microbiota have been implicated in key aspects of helminth disease and infection outcomes. Nevertheless, the direct and indirect mechanisms governing these interactions are, thus far, largely unknown. In this proof-of-concept study, we demonstrate that the excretory-secretory products (ESPs) and extracellular vesicles (EVs) of key GI nematodes contain peptides that, when recombinantly expressed, exert antimicrobial activity in vitro against Bacillus subtilis. In particular, using time-lapse microfluidics microscopy, we demonstrate that exposure of B. subtilis to a recombinant saposin-domain containing peptide from the 'brown stomach worm', Teladorsagia circumcincta, and a metridin-like ShK toxin from the 'barber's pole worm', Haemonchus contortus, results in cell lysis and significantly reduced growth rates. Data from this study support the hypothesis that GI nematodes may modulate the composition of the vertebrate gut microbiota directly via the secretion of antimicrobial peptides, and pave the way for future investigations aimed at deciphering the impact of such changes on the pathophysiology of GI helminth infection and disease.
Collapse
Affiliation(s)
- James Rooney
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | - Ruizhe Li
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Kevin Mclean
- Moredun Research Institute, Penicuik Midlothian, United Kingdom
| | | | | | - Andreas H. Laustsen
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Timothy P. Jenkins
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Andreas Hofmann
- Max Rubner-Institut, Federal Research Institute of Nutrition and Food, Kulmbach, Germany
- Melbourne Veterinary School, Faculty of Science, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Somenath Bakshi
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Ashraf Zarkan
- Department of Genetics, University of Cambridge, Cambridge, United Kingdom
| | - Cinzia Cantacessi
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
24
|
Fleischer R, Velling M, Peters W, Peterka T, Franke F, Vymyslická PJ, Rehbein S, Heurich M, Sommer S. Invasive Fascioloides magna infections impact gut microbiota in a definitive host in Europe. Int J Parasitol Parasites Wildl 2024; 25:101024. [PMID: 39687766 PMCID: PMC11648883 DOI: 10.1016/j.ijppaw.2024.101024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 11/23/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024]
Abstract
Invasive parasites that expand their natural range can be a threat to wildlife biodiversity and may pose a health risk to non-adapted, naive host species. The invasive giant liver fluke, Fascioloides magna, native to North America, has extended its range in Europe and uses mainly red deer (Cervus elaphus) as definitive hosts. The penetration of the intestinal barrier by the young flukes to reach the liver via the abdominal cavity as well as the release of fluke metabolism products and excreta with the bile and/or changes in the microbial community of the biliary system may enable the translocation of intestinal bacteria across the intestinal barrier and, in turn, could be associated with inflammation and changes in the intestinal bacterial community. The gut commensal community plays a key role in host nutrition and interacts with cells of the immune system to maintain host health. For this study, the gut bacterial community of red deer infected with F. magna and of non-infected red deer from one of the largest forest ecosystems in Central Europe, located on the border between the Czech Republic and Germany, was investigated. The individual fluke burden was associated with changes in the gut microbial composition of the gut of infected individuals, whereas the diversity and composition of the gut bacteria were only slightly different between fluke-infected and uninfected deer. Several bacterial taxa at the genus level were unique to individuals carrying either one or many liver flukes. Our results suggest that the microbiota of red deer is stable to perturbation by low numbers of F. magna. However, a larger parasite burden may cause changes in the gut microbial composition in definitive hosts implying that non-invasive fecal microbiome assessments could serve as indicator for wildlife health monitoring.
Collapse
Affiliation(s)
- Ramona Fleischer
- Institute of Evolutionary Ecology and Conservation Genomics, University of Ulm, Germany
| | - Marc Velling
- Faculty of Environment and Natural Resources, University of Freiburg, Germany
- Department of National Park Monitoring and Animal Management, Bavarian Forest National Park, Germany
| | - Wibke Peters
- Bavarian State Institute of Forestry, Research Unit Wildlife Biology and Management, Freising, Germany
| | - Tomáš Peterka
- Faculty of Forestry and Wood Sciences, Czech University of Life Sciences Prague, Praha - Suchdol, Czech Republic
- Šumava National Park, Vimperk, Czech Republic
| | - Frederik Franke
- Bavarian State Institute of Forestry, Research Unit Wildlife Biology and Management, Freising, Germany
| | - Pavla Jůnková Vymyslická
- Šumava National Park, Vimperk, Czech Republic
- Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Praha – Suchdol, Czech Republic
| | | | - Marco Heurich
- Faculty of Environment and Natural Resources, University of Freiburg, Germany
- Department of National Park Monitoring and Animal Management, Bavarian Forest National Park, Germany
- Institute for Forest and Wildlife Management, Inland Norway University of Applied Sciences, Koppang, Norway
| | - Simone Sommer
- Institute of Evolutionary Ecology and Conservation Genomics, University of Ulm, Germany
| |
Collapse
|
25
|
Lopez VA, Lim JJ, Seguin RP, Dempsey JL, Kunzman G, Cui JY, Xu L. Oral exposure to benzalkonium chlorides in male and female mice reveals alteration of the gut microbiome and bile acid profile. Toxicol Sci 2024; 202:265-277. [PMID: 39363503 PMCID: PMC11589104 DOI: 10.1093/toxsci/kfae116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024] Open
Abstract
Benzalkonium chlorides (BACs) are commonly used disinfectants in a variety of consumer and food-processing settings, and the COVID-19 pandemic has led to increased usage of BACs. The prevalence of BACs raises the concern that BAC exposure could disrupt the gastrointestinal microbiota, thus interfering with the beneficial functions of the microbes. We hypothesize that BAC exposure can alter the gut microbiome diversity and composition, which will disrupt bile acid (BA) homeostasis along the gut-liver axis. In this study, male and female mice were exposed orally to d7-C12- and d7-C16-BACs at 120 µg/g/d for 1 wk. UPLC-MS/MS analysis of liver, blood, and fecal samples of BAC-treated mice demonstrated the absorption and metabolism of BACs. Both parent BACs and their metabolites were detected in all exposed samples. Additionally, 16S rRNA sequencing was carried out on the bacterial DNA isolated from the cecum intestinal content. For female mice, and to a lesser extent in males, we found that treatment with either d7-C12- or d7-C16-BAC led to decreased alpha diversity and differential composition of gut bacteria with notably decreased actinobacteria phylum. Lastly, through a targeted BA quantitation analysis, we observed decreases in secondary BAs in BAC-treated mice, which was more pronounced in the female mice. This finding is supported by decreases in bacteria known to metabolize primary BAs into secondary BAs, such as the families of Ruminococcaceae and Lachnospiraceae. Together, these data signify the potential impact of BACs on human health through disturbance of the gut microbiome and gut-liver interactions.
Collapse
Affiliation(s)
- Vanessa A Lopez
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, United States
| | - Joe J Lim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, United States
| | - Ryan P Seguin
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, United States
| | - Joseph L Dempsey
- Division of Gastroenterology, Department of Medicine, University of Washington, Seattle, WA 98195, United States
| | - Gabrielle Kunzman
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, United States
| | - Julia Y Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, United States
| | - Libin Xu
- Department of Medicinal Chemistry, University of Washington, Seattle, WA 98195, United States
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195, United States
| |
Collapse
|
26
|
Yang H. Gut Microbiota, Circulating Metabolites and Risk of Endometriosis: A Two-Step Mendelian Randomization Study. Pol J Microbiol 2024; 73:491-503. [PMID: 39670637 PMCID: PMC11639408 DOI: 10.33073/pjm-2024-041] [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: 06/30/2024] [Accepted: 09/19/2024] [Indexed: 12/14/2024] Open
Abstract
Epidemiological studies and animal models have suggested a possible link between gut microbiota (GM), circulating metabolites, and endometriosis (EMs) pathogenesis. However, whether these associations are causal or merely due to confounding factors remains unclear. We conducted a two-sample and two-step Mendelian randomization (MR) study to elucidate the potential causal relationship between GM and EMs, and the mediating role of circulating metabolites. Our MR analysis revealed that higher abundances of class Negativicutes, and order Selenomonadales, as well as genera Dialister, Enterorhabdus, Eubacterium xylanophilum group, Methanobrevibacter were associated with an increased risk of EMs (Odds Ratio (OR) range: 1.0019-1.0037). Conversely, higher abundances of genera Coprococcus 1 and Senegalimassilia were linked to reduced risk of EMs (OR range: 0.9964-0.9967). Additionally, elevated levels of circulating metabolites such as 1-eicosatrienoyl-glycerophosphocholine and 1-oleoylglycerophosphocholine were found to be associated with heightened risk of EMs (OR range: 2.21-3.16), while higher concentrations of 3-phenylpropionate and dihomo-linolenate were protective (OR range: 0.285-0.535). Two-step MR analysis indicated that specific microbial taxa, notably genus Enterorhabdus and order Selenomonadales, might function as mediators linking circulating metabolites to the risk of EMs. Our findings suggest a probable causal relationship between GM, circulating metabolites, and EMs, indicating that GM may mediate the influence of circulating metabolites on the pathophysiology of EMs. These results offer new leads for future mechanistic studies and could inform clinical translational research.
Collapse
Affiliation(s)
- Hua Yang
- Department of Gynecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
27
|
Papp PP, Hoffmann OI, Libisch B, Keresztény T, Gerőcs A, Posta K, Hiripi L, Hegyi A, Gócza E, Szőke Z, Olasz F. Effects of Polyvinyl Chloride (PVC) Microplastic Particles on Gut Microbiota Composition and Health Status in Rabbit Livestock. Int J Mol Sci 2024; 25:12646. [PMID: 39684357 DOI: 10.3390/ijms252312646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
The widespread use of polyvinyl chloride (PVC) and its entry into humans and livestock is of serious concern. In our study, we investigated the impact of PVC treatments on physiological, pathological, hormonal, and microbiota changes in female rabbits. Trend-like alterations in weight were observed in the spleen, liver, and kidney in both low (P1) and high dose (P2) PVC treatment groups. Histopathological examination revealed exfoliation of the intestinal mucosa in the treated groups compared to the control, and microplastic particles were penetrated and embedded in the spleen. Furthermore, both P1 and P2 showed increased 17-beta-estradiol (E2) hormone levels, indicating early sexual maturation. Moreover, the elevated tumor necrosis factor alpha (TNF-α) levels suggest inflammatory reactions associated with PVC treatment. Genus-level analyses of the gut microbiota in group P2 showed several genera with increased or decreased abundance. In conclusion, significant or trend-like correlations were demonstrated between the PVC content of feed and physiological, pathological, and microbiota parameters. To our knowledge, this is the first study to investigate the broad-spectrum effects of PVC microplastic exposure in rabbits. These results highlight the potential health risks associated with PVC microplastic exposure, warranting further investigations in both animals and humans.
Collapse
Affiliation(s)
- Péter P Papp
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Microbiology and Applied Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| | - Orsolya Ivett Hoffmann
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Animal Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| | - Balázs Libisch
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Microbiology and Applied Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| | - Tibor Keresztény
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Microbiology and Applied Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
- Doctoral School of Biological Sciences, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| | - Annamária Gerőcs
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Microbiology and Applied Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
- Doctoral School of Biology, ELTE Eötvös Loránd University, 1117 Budapest, Hungary
| | - Katalin Posta
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Microbiology and Applied Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| | - László Hiripi
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Animal Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
- Laboratory Animal Science Coordination Center, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
| | - Anna Hegyi
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Microbiology and Applied Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| | - Elen Gócza
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Animal Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| | - Zsuzsanna Szőke
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Animal Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| | - Ferenc Olasz
- Agribiotechnology and Precision Breeding for Food Security National Laboratory, Department of Microbiology and Applied Biotechnology, Institute of Genetics and Biotechnology, Hungarian University of Agriculture and Life Sciences, 2100 Gödöllő, Hungary
| |
Collapse
|
28
|
Joubran P, Roux FA, Serino M, Deschamps JY. Gut Microbiota Comparison in Rectal Swabs Versus Stool Samples in Cats with Kidney Stones. Microorganisms 2024; 12:2411. [PMID: 39770613 PMCID: PMC11677927 DOI: 10.3390/microorganisms12122411] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/17/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
To investigate the role of the intestinal bacterial microbiota in the pathogenesis of calcium oxalate nephrolithiasis in cats, a condition characterized by the formation of kidney stones, it is desirable to identify a sample collection method that accurately reflects the microbiota's composition. The objective of this study was to evaluate the impact of fecal sample collection methods on the intestinal microbiota composition in two cat populations: healthy cats and kidney stone-diseased cats. The study included eighteen cats from the same colony, comprising nine healthy cats and nine cats with spontaneously occurring presumed calcium oxalate kidney stones. Three fecal collection methods were compared: rectal swabs, the collection of fresh stool, and the collection of stool exposed to ambient air for 24 h. The bacterial microbiota was analyzed through the high-resolution sequencing of the V3-V4 region of the 16S rRNA gene. For all cats, within the same individual, a one-way PERMANOVA analysis showed a significant difference between the rectal swabs and fresh stool (p = 0.0003), as well as between the rectal swabs and stool exposed to ambient air for 24 h (p = 0.0003), but no significant difference was identified between the fresh stool and non-fresh stool (p = 0.0651). When comparing the two populations of cats, this study provides seemingly conflicting results. (1) A principal component analysis (PCA) comparison revealed a significant difference in the bacterial composition between the healthy cats and the cats with kidney stones only when the sample was a fresh fecal sample (p = 0.0037). This finding suggests that the intestinal bacteria involved in the pathogenesis of kidney stones in cats are luminal and strictly anaerobic bacteria. Consequently, exposure to ambient air results in a loss of information, preventing the identification of dysbiosis. For clinical studies, non-fresh stool samples provided by owners does not appear suitable for studying the gut microbiota of cats with kidney stones; fresh stool should be favored. (2) Interestingly, the rectal swabs alone highlighted significant differences in the proportion of major phyla between the two populations. These findings highlight the critical importance of carefully selecting fecal collection methods when studying feline gut microbiota. Combining rectal swabs and fresh stool sampling provides complementary insights, offering the most accurate understanding of the gut microbiota composition in the context of feline kidney stone pathogenesis.
Collapse
Affiliation(s)
- Patrick Joubran
- NP3, Nutrition, PathoPhysiology and Pharmacology Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44307 Nantes, France; (P.J.); (F.A.R.)
| | - Françoise A. Roux
- NP3, Nutrition, PathoPhysiology and Pharmacology Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44307 Nantes, France; (P.J.); (F.A.R.)
- Emergency and Critical Care Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44307 Nantes, France
| | - Matteo Serino
- IRSD, Institut de Recherche en Santé Digestive, Institut National de la Santé et de la Recherche Médicale (INSERM) U1220, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Ecole Nationale Vétérinaire de Toulouse (ENVT), Université de Toulouse III-Paul Sabatier (UPS), CS 60039, 31024 Toulouse, France;
| | - Jack-Yves Deschamps
- NP3, Nutrition, PathoPhysiology and Pharmacology Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44307 Nantes, France; (P.J.); (F.A.R.)
- Emergency and Critical Care Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44307 Nantes, France
| |
Collapse
|
29
|
Yang H. The causality between gut microbiota and endometriosis: a bidirectional Mendelian randomization study. Front Med (Lausanne) 2024; 11:1434582. [PMID: 39650192 PMCID: PMC11621931 DOI: 10.3389/fmed.2024.1434582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 11/13/2024] [Indexed: 12/11/2024] Open
Abstract
Background Observational studies and animal experiments had suggested a potential relationship between gut microbiota abundance and pathogenesis of endometriosis (EMs), but the relevance of this relationship remains to be clarified. Methods We perform a two-sample bidirectional Mendelian randomization (MR) analysis to explore whether there is a causal correlation between the abundance of the gut microbiota and EMs and the direction of causality. Genome-wide association study (GWAS) data ukb-d-N80, finn-b-N14-EM, and MiBinGen were selected. Inverse variance weighted (IVW), weighted median, and MR Egger are selected for causal inference. The Cochran Q test, Egger intercept test, and leave-one-out analysis are performed for sensitivity analyses. Results In the primary outcome, we find that a higher abundance of class Negativicutes, genus Dialister, genus Enterorhabdus, genus Eubacterium xylanophilum group, genus Methanobrevibacter and order Selenomonadales predict a higher risk of EMs, and a higher abundance of genus Coprococcus and genus Senegalimassilia predict a lower risk of EMs. During verifiable outcomes, we find that a higher abundance of phylum Cyanobacteria, genus Ruminococcaceae UCG002, and genus Coprococcus 3 predict a higher risk of EMs, and a higher abundance of genus Flavonifracto, genus Bifidobacterium, and genus Rikenellaceae RC9 predict a lower risk of EMs. In primary reverse MR analysis, we find that EMs predict a lower abundance of the genus Eubacterium fissicatena group, genus Prevotella7, genus Butyricicoccus, family Lactobacillaceae, and a higher abundance of genus Ruminococcaceae UCG009. In verifiable reverse MR analysis, we find that EMs predict a lower abundance of the genus Ruminococcaceae UCG004 and a higher abundance of the genus Howardella. Conclusion Our study implies a mutual causality between gut microbiota abundance and the pathogenesis of EMs, which may provide a novel direction for EMs diagnosis, prevention, and treatment, may promote future functional or clinical analysis.
Collapse
Affiliation(s)
- Hua Yang
- Department of Gynecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
30
|
Biennier S, Fontaine M, Duquenoy A, Schwintner C, Doré J, Corvaia N. Narrative Review: Advancing Dysbiosis Treatment in Onco-Hematology with Microbiome-Based Therapeutic Approach. Microorganisms 2024; 12:2256. [PMID: 39597645 PMCID: PMC11596191 DOI: 10.3390/microorganisms12112256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
This review explores the complex relationship between gut dysbiosis and hematological malignancies, focusing on graft-versus-host disease (GvHD) in allogeneic hematopoietic stem cell transplantation (allo-HSCT) recipients. We discuss how alterations in microbial diversity and composition can influence disease development, progression, and treatment outcomes in blood cancers. The mechanisms by which the gut microbiota impacts these conditions are examined, including modulation of immune responses, production of metabolites, and effects on intestinal barrier function. Recent advances in microbiome-based therapies for treating and preventing GvHD are highlighted, with emphasis on full ecosystem standardized donor-derived products. Overall, this review underscores the growing importance of microbiome research in hematology-oncology and its potential to complement existing treatments and improve outcomes for thousands of patients worldwide.
Collapse
Affiliation(s)
- Salomé Biennier
- MaaT Pharma, 69007 Lyon, France; (S.B.); (A.D.); (C.S.); (N.C.)
| | | | - Aurore Duquenoy
- MaaT Pharma, 69007 Lyon, France; (S.B.); (A.D.); (C.S.); (N.C.)
| | | | - Joël Doré
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS, 78350 Jouy-en-Josas, France;
| | | |
Collapse
|
31
|
Schmid SM, Tolbert MK. Harnessing the microbiome: probiotics, antibiotics and their role in canine and feline gastrointestinal disease. Vet Rec 2024; 195:13-25. [PMID: 39545593 DOI: 10.1002/vetr.4915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Unfavourable alterations of the host microbial environment, known as dysbiosis, have been identified in many canine and feline gastrointestinal (GI) diseases. As a result, normalisation of microbial composition and function has become an important therapeutic target. Given the complex and individualistic interplay between the resident microbiota, host and environment, a multimodal approach is often necessary when addressing dysbiosis in dogs and cats with GI disease. Systemic antibiotics are often empirically used to treat acute and chronic GI diseases. However, with modern genomic techniques demonstrating the profound negative effect antibiotics can have on the GI microbiota and the rapid emergence of resistant bacteria globally, there has been an increased focus on identifying antibiotic alternatives for use in small animal practice. Biotics, such as prebiotics, probiotics and synbiotics, are of growing interest due to their potential supportive effect on the microbiota. This article reviews the evidence for the use of biotics in canine and feline GI disease, highlighting how judicious use of antibiotics and targeted probiotic supplementation can enhance patient outcomes by promoting a balanced gut microbial environment.
Collapse
Affiliation(s)
- Sarah M Schmid
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Tennessee, USA
| | - M Katherine Tolbert
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, College of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
32
|
Zhu M, Zhang T, Xu D, Zhou B, Wang K, Liao C, Cheng Z, Li P, Chen C. Impact of fermented wine lees on gut microbiota and metabolic responses in Guanling crossbred cattle. BMC Microbiol 2024; 24:421. [PMID: 39438796 PMCID: PMC11495091 DOI: 10.1186/s12866-024-03583-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND The addition of wine lees to diets can make up for the deficiencies caused by traditional forages in beef cattle farming. However, the effects of different wine lees ratios on average daily weight, gastrointestinal microbial community structure and metabolites in Guanling crossbred cattle have been rarely studied. This study assessed the effects of feeds containing wine lees on weight gain, gastrointestinal microbial community structure, and metabolites in Guanling crossbred cattle and elucidated the metabolic responses induced by wine lees. Eighteen cows were randomly assigned to receive fed concentrate (C group), feed containing 15% wine lees (group A), or feed containing 30% wine lees (group B) for 60 days. RESULTS The average daily weight gain of group A and group B increased by 76.75% and 57.65%, respectively, compared with group C. Microbial community analysis showed that wine lees increased the abundance of Prevotella_1 in the rumen, decreased the abundance of Ruminococcaceae UCG 011 and Lachnospiraceae_FCS020_group in the rumen, and increased the abundance of Tyzzerella_4, Family_Xlll_AD3011_group, Granulicella, and Eisenbergiella in the cecum. Metabolomics analyses showed that wine lees decreased the concentrations of indole-3-ethanol in the rumen, and complexity cecal metabolism. Notably, linoleic acid metabolism was significantly enriched in both the rumen and cecum. Mantel test analyses indicated that the adverse effects of WL were reduced by stimulating the metabolism of linoleic acid, α-linolenic acid, and tryptophan, and these changes were mediated by intestinal microorganisms. The Guanling cattle cecum was enriched for several unfavorable metabolic pathways when wine lees concentrations reached 30%, which increased the likelihood of intestinal lesions. CONCLUSION This study shows that WL supplementation alters gut microbiota and metabolic pathways, improving cattle growth and health. Moderate WL levels (15%) enhance gut health and beneficial pathways (e.g., linoleic and alpha-linolenic acid metabolism). However, higher WL inclusion (30%) may activate adverse pathways, raising the risk of intestinal damage. To maximize benefits and minimize risks, WL levels should be carefully managed.
Collapse
Affiliation(s)
- Mingming Zhu
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Tiantian Zhang
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Duhan Xu
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Bijun Zhou
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Kaigong Wang
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Chaosheng Liao
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Zhentao Cheng
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Ping Li
- College of Animal Science, Guizhou University, Guiyang, 550025, China
| | - Chao Chen
- College of Animal Science, Guizhou University, Guiyang, 550025, China.
| |
Collapse
|
33
|
Li J, Sun Z, Sun F, Lai Y, Yi X, Wang Z, Yuan J, Hu Y, Pan A, Pan XF, Zheng Y, Chen D. Gut antibiotic resistome during pregnancy associates with the risk of gestational diabetes mellitus: New evidence from a prospective nested case-control study. JOURNAL OF HAZARDOUS MATERIALS 2024; 478:135434. [PMID: 39146585 DOI: 10.1016/j.jhazmat.2024.135434] [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: 12/24/2023] [Revised: 07/24/2024] [Accepted: 08/04/2024] [Indexed: 08/17/2024]
Abstract
Antibiotic resistome has emerged as a global threat to public health. However, gestational antibiotic resistome and potential link with adverse pregnancy outcomes remains poorly understood. Our study reports for the first time an association between gut antibiotic resistome during early pregnancy and the risk of gestational diabetes mellitus (GDM) based on a prospective nested case-control cohort including 120 cases and 120 matched controls. A total of 214 antibiotic resistance gene (ARG) subtypes belonging to 17 ARG types were identified in > 10 % fecal samples collected during each trimester. The data revealed dynamic profiles of gut antibiotic resistome through pregnancy, and significant positive associations between selected features (i.e., ARG abundances and a GDM-ARG score which is a new feature characterizing the association between ARGs and GDM) of gut antibiotic resistome during early pregnancy and GDM risk as well as selected endogenous metabolites. The findings demonstrate ubiquitous presence of ARGs in pregnant women and suggest it could constitute an important risk factor for the development of GDM.
Collapse
Affiliation(s)
- Jing Li
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China; School of Public Health, Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Zhonghan Sun
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Fudan University, 200433, Shanghai, China
| | - Fengjiang Sun
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China
| | - Yuwei Lai
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xinzhu Yi
- Institute of Ecological Sciences, School of Life Sciences, South China Normal University, Guangzhou 510631, Guangdong, China
| | - Zhang Wang
- Institute of Ecological Sciences, School of Life Sciences, South China Normal University, Guangzhou 510631, Guangdong, China
| | - Jiaying Yuan
- Department of Science and Education, Shuangliu Maternal and Child Health Hospital, Chengdu 610200, Sichuan, China
| | - Yayi Hu
- Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - An Pan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
| | - Xiong-Fei Pan
- Shuangliu Institute of Women's and Children's Health, Shuangliu Maternal and Child Health Hospital, Chengdu 610041, Sichuan, China; Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & National Medical Products Administration Key Laboratory for Technical Research on Drug Products In Vitro and In Vivo Correlation, West China Second University Hospital, Sichuan University, Shuangliu Maternal and Child Health Hospital, Chengdu 610041, Sichuan, China.
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Human Phenome Institute, Fudan University, 200433, Shanghai, China.
| | - Da Chen
- College of Environment and Climate, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China.
| |
Collapse
|
34
|
Kang JW, Vemuganti V, Kuehn JF, Ulland TK, Rey FE, Bendlin BB. Gut microbial metabolism in Alzheimer's disease and related dementias. Neurotherapeutics 2024; 21:e00470. [PMID: 39462700 PMCID: PMC11585892 DOI: 10.1016/j.neurot.2024.e00470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024] Open
Abstract
Multiple studies over the last decade have established that Alzheimer's disease and related dementias (ADRD) are associated with changes in the gut microbiome. These alterations in organismal composition result in changes in the abundances of functions encoded by the microbial community, including metabolic capabilities, which likely impact host disease mechanisms. Gut microbes access dietary components and other molecules made by the host and produce metabolites that can enter circulation and cross the blood-brain barrier (BBB). In recent years, several microbial metabolites have been associated with or have been shown to influence host pathways relevant to ADRD pathology. These include short chain fatty acids, secondary bile acids, tryptophan derivatives (such as kynurenine, serotonin, tryptamine, and indoles), and trimethylamine/trimethylamine N-oxide. Notably, some of these metabolites cross the BBB and can have various effects on the brain, including modulating the release of neurotransmitters and neuronal function, inducing oxidative stress and inflammation, and impacting synaptic function. Microbial metabolites can also impact the central nervous system through immune, enteroendocrine, and enteric nervous system pathways, these perturbations in turn impact the gut barrier function and peripheral immune responses, as well as the BBB integrity, neuronal homeostasis and neurogenesis, and glial cell maturation and activation. This review examines the evidence supporting the notion that ADRD is influenced by gut microbiota and its metabolites. The potential therapeutic advantages of microbial metabolites for preventing and treating ADRD are also discussed, highlighting their potential role in developing new treatments.
Collapse
Affiliation(s)
- Jea Woo Kang
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Vaibhav Vemuganti
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jessamine F Kuehn
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Tyler K Ulland
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
35
|
Zare D, Aryaee H, Mirdamadi S, Shirkhan F. The Benefits and Applications of Lactobacillus plantarum in Food and Health: A Narrative Review. IRANIAN JOURNAL OF PUBLIC HEALTH 2024; 53:2201-2213. [PMID: 39544869 PMCID: PMC11557752 DOI: 10.18502/ijph.v53i10.16698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/18/2024] [Indexed: 11/17/2024]
Abstract
Lactobacillus plantarum is a type of gram-positive lactic acid bacteria. This bacterium is considered a safe probiotic and, many applications and benefits including prolonging food shelf-life, enhancing antioxidant activity, improving food flavor characteristics and antimicrobial activities in the food industry, and application as a potential starter for dairy products have been attributed to it. Various studies have also emphasized its health-giving properties. As a result, the features and wide application of this bacterium, as well as the safety of L. plantarum and its strains, have made it a popular probiotic in the food and medical industries. Thus, in the present study keywords including L. plantarum and Lactiplantibacillus plantarum along with application, benefits, food, health, anti-oxidant, anti-diabetic, anti-obesity, anti-inflammatory, antiviral, and anti-depression were searched in databases of PubMed, Scopus, Web of Science, Sience direct and Google Scholar with no time restriction. Then, important features, benefits, and uses of L. plantarum were categorized and discussed. The ability of L. plantarum on the food such as prolonging food shelf-life, enhancing antioxidant activity, improving food flavor characteristics and antimicrobial activities in the food industry, and as a potential starter for dairy products is effective. In addition, several studies have emphasized of L. plantarum health-giving properties.
Collapse
Affiliation(s)
- Davood Zare
- Department of Biotechnology, Iranian Research Organization for Science & Technology (IROST), Tehran, Iran
| | - Hadis Aryaee
- Department of Biotechnology, Iranian Research Organization for Science & Technology (IROST), Tehran, Iran
| | - Saeed Mirdamadi
- Department of Biotechnology, Iranian Research Organization for Science & Technology (IROST), Tehran, Iran
| | - Faezeh Shirkhan
- Department of Food Science and Technology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
36
|
Guan J, Ramírez GA, Eng C, Oakley B. Microbiome resilience of three-toed box turtles ( Terrapene carolina triunguis) in response to rising temperatures. Front Vet Sci 2024; 11:1276436. [PMID: 39286599 PMCID: PMC11402899 DOI: 10.3389/fvets.2024.1276436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
The gastrointestinal (GI) microbiome of chelonians (testudines) plays an important role in their metabolism, nutrition, and overall health but the GI microbiome of three-toed box turtles (Terrapene carolina triunguis) has yet to be characterized. How the GI microbiome responds to rapidly rising environmental temperatures has also not been studied extensively in ectotherms, specifically chelonians. In this study, twenty (20) T.c.triunguis were split into control and experimental groups. The experimental group experienced 4.5°C increases every two weeks while the control group stayed at a constant ambient temperature (24°C) through the entirety of the experiment. Before each temperature increase, all turtles had cloacal swab samples taken. These samples underwent DNA extraction followed by 16S rRNA gene sequencing and microbial community analyses. Differences in diversity at the community level in the controls compared to the experimental groups were not statistically significant, indicating microbiome resilience to rapid temperature changes in T.c.triunguis, although some differentially abundant lineages were identified. Interestingly, an amplicon sequence variant belonging to the Erysipelothrix spp. was exclusively enriched in the highest temperature group relative to controls. Overall, our work suggests that there may be an innate robustness to rapid temperature swings in the microbiome of T.c.triunguis which are native to temperate North America. Despite this resilience, Erysipelothrix spp. was enriched at the highest temperature. Phylogenetic analysis of this amplicon variant showed that it is a close relative of Erysipelothrix rhusiopathiae, a pathogen of zoonotic importance associated with both wildlife and livestock.
Collapse
Affiliation(s)
- Jimmy Guan
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, United States
| | - Gustavo A Ramírez
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, United States
- Department of Biological Sciences, California State University Los Angeles, Los Angeles, CA, United States
| | - Curtis Eng
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, United States
| | - Brian Oakley
- College of Veterinary Medicine, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
37
|
Russell AC, Kenna MA, Huynh AV, Rice AM. Microbial DNA extraction method for avian feces and preen oil from diverse species. Ecol Evol 2024; 14:e70220. [PMID: 39224152 PMCID: PMC11368492 DOI: 10.1002/ece3.70220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 08/07/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
As DNA sequencing technology continues to rapidly improve, studies investigating the microbial communities of host organisms (i.e., microbiota) are becoming not only more popular but also more financially accessible. Across many taxa, microbiomes can have important impacts on organismal health and fitness. To evaluate the microbial community composition of a particular microbiome, microbial DNA must be successfully extracted. Fecal samples are often easy to collect and are a good source of gut microbial DNA. Additionally, interest in the avian preen gland microbiome is rapidly growing, due to the importance of preen oil for many aspects of avian life. Microbial DNA extractions from avian fecal and preen oil samples present multiple challenges, however. Here, we describe a modified PrepMan Ultra Sample Preparation Reagent microbial DNA extraction method that is less expensive than other commonly used methodologies and is highly effective for both fecal and preen oil samples collected from a broad range of avian species. We expect our method will facilitate microbial DNA extractions from multiple avian microbiome reservoirs, which have previously proved difficult and expensive. Our method therefore increases the feasibility of future studies of avian host microbiomes.
Collapse
Affiliation(s)
- Austin C. Russell
- Department of Biological SciencesLehigh UniversityBethlehemPennsylvaniaUSA
| | - Margaret A. Kenna
- Department of Biological SciencesLehigh UniversityBethlehemPennsylvaniaUSA
| | - Alex Van Huynh
- Department of BiologyDeSales UniversityCenter ValleyPennsylvaniaUSA
| | - Amber M. Rice
- Department of Biological SciencesLehigh UniversityBethlehemPennsylvaniaUSA
| |
Collapse
|
38
|
Jia H, Dong N. Effects of bile acid metabolism on intestinal health of livestock and poultry. J Anim Physiol Anim Nutr (Berl) 2024; 108:1258-1269. [PMID: 38649786 DOI: 10.1111/jpn.13969] [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: 09/01/2022] [Revised: 01/27/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
Bile acids are synthesised in the liver and are essential amphiphilic steroids for maintaining the balance of cholesterol and energy metabolism in livestock and poultry. They can be used as novel feed additives to promote fat utilisation in the diet and the absorption of fat-soluble substances in the feed to improve livestock performance and enhance carcass quality. With the development of understanding of intestinal health, the balance of bile acid metabolism is closely related to the composition and growth of livestock intestinal microbiota, inflammatory response, and metabolic diseases. This paper systematically reviews the effects of bile acid metabolism on gut health and gut microbiology in livestock. In addition, our paper summarised the role of bile acid metabolism in performance and disease control.
Collapse
Affiliation(s)
- Hongpeng Jia
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| | - Na Dong
- The Laboratory of Molecular Nutrition and Immunity, Institute of Animal Nutrition, Northeast Agricultural University, Harbin, China
| |
Collapse
|
39
|
Wu WC, Pan YF, Zhou WD, Liao YQ, Peng MW, Luo GY, Xin GY, Peng YN, An T, Li B, Luo H, Barrs VR, Beatty JA, Holmes EC, Zhao W, Shi M, Shu Y. Meta-transcriptomic analysis of companion animal infectomes reveals their diversity and potential roles in animal and human disease. mSphere 2024; 9:e0043924. [PMID: 39012105 PMCID: PMC11351045 DOI: 10.1128/msphere.00439-24] [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: 05/22/2024] [Accepted: 06/28/2024] [Indexed: 07/17/2024] Open
Abstract
Companion animals such as cats and dogs harbor diverse microbial communities that can potentially impact human health due to close and frequent contact. To better characterize their total infectomes and assess zoonotic risks, we characterized the overall infectomes of companion animals (cats and dogs) and evaluated their potential zoonotic risks. Meta-transcriptomic analyses were performed on 239 samples from cats and dogs collected across China, identifying 24 viral species, 270 bacterial genera, and two fungal genera. Differences in the overall microbiome and infectome composition were compared across different animal species (cats or dogs), sampling sites (rectal or oropharyngeal), and health status (healthy or diseased). Diversity analyses revealed that viral abundance was generally higher in diseased animals compared to healthy ones, while differences in microbial composition were mainly driven by sampling site, followed by animal species and health status. Disease association analyses validated the pathogenicity of known pathogens and suggested potential pathogenic roles of previously undescribed bacteria and newly discovered viruses. Cross-species transmission analyses identified seven pathogens shared between cats and dogs, such as alphacoronavirus 1, which was detected in both oropharyngeal and rectal swabs albeit with differential pathogenicity. Further analyses showed that some viruses, like alphacoronavirus 1, harbored multiple lineages exhibiting distinct pathogenicity, tissue, or host preferences. Ultimately, a systematic evolutionary screening identified 27 potential zoonotic pathogens in this sample set, with far more bacterial than viral species, implying potential health threats to humans. Overall, our meta-transcriptomic analysis reveals a landscape of actively transcribing microorganisms in major companion animals, highlighting key pathogens, those with the potential for cross-species transmission, and possible zoonotic threats. IMPORTANCE This study provides a comprehensive characterization of the entire community of infectious microbes (viruses, bacteria, and fungi) in companion animals like cats and dogs, termed the "infectome." By analyzing hundreds of samples from across China, the researchers identified numerous known and novel pathogens, including 27 potential zoonotic agents that could pose health risks to both animals and humans. Notably, some of these zoonotic pathogens were detected even in apparently healthy pets, highlighting the importance of surveillance. The study also revealed key microbial factors associated with respiratory and gastrointestinal diseases in pets, as well as potential cross-species transmission events between cats and dogs. Overall, this work sheds light on the complex microbial landscapes of companion animals and their potential impacts on animal and human health, underscoring the need for monitoring and management of these infectious agents.
Collapse
Affiliation(s)
- Wei-Chen Wu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yuan-Fei Pan
- Ministry of Education Key Laboratory of Biodiversity Science and Ecological Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Wu-Di Zhou
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yu-Qi Liao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Min-Wu Peng
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Geng-Yan Luo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Gen-Yang Xin
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Ya-Ni Peng
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Tongqing An
- State Key Laboratory of Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Bo Li
- Ministry of Education Key Laboratory of Biodiversity Science and Ecological Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Ministry of Education Key Laboratory for Ecosecurity of Southwest China, Yunnan Key Laboratory of Plant Reproductive Adaptation and Evolutionary, Ecology and Centre for Invasion Biology, Institute of Biodiversity, School of Ecology and Environmental Science, Yunnan University, Kunming, China
| | - Huanle Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Vanessa R. Barrs
- Department of Veterinary Clinical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR, China
- Centre for Animal Health and Welfare, City University of Hong Kong, Hong Kong SAR, China
| | - Julia A. Beatty
- Department of Veterinary Clinical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Hong Kong SAR, China
- Centre for Animal Health and Welfare, City University of Hong Kong, Hong Kong SAR, China
| | - Edward C. Holmes
- Sydney Institute for Infectious Diseases, School of Medical Sciences, The University of Sydney, Sydney, New South Wales, Australia
- Laboratory of Data Discovery for Health Limited, Hong Kong SAR, China
| | - Wenjing Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Mang Shi
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
- Key Laboratory of Pathogen Infection Prevention and Control (MOE), State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
40
|
Lai S, Wang H, Bork P, Chen WH, Zhao XM. Long-read sequencing reveals extensive gut phageome structural variations driven by genetic exchange with bacterial hosts. SCIENCE ADVANCES 2024; 10:eadn3316. [PMID: 39141729 PMCID: PMC11323893 DOI: 10.1126/sciadv.adn3316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 07/10/2024] [Indexed: 08/16/2024]
Abstract
Genetic variations are instrumental for unraveling phage evolution and deciphering their functional implications. Here, we explore the underlying fine-scale genetic variations in the gut phageome, especially structural variations (SVs). By using virome-enriched long-read metagenomic sequencing across 91 individuals, we identified a total of 14,438 nonredundant phage SVs and revealed their prevalence within the human gut phageome. These SVs are mainly enriched in genes involved in recombination, DNA methylation, and antibiotic resistance. Notably, a substantial fraction of phage SV sequences share close homology with bacterial fragments, with most SVs enriched for horizontal gene transfer (HGT) mechanism. Further investigations showed that these SV sequences were genetic exchanged between specific phage-bacteria pairs, particularly between phages and their respective bacterial hosts. Temperate phages exhibit a higher frequency of genetic exchange with bacterial chromosomes and then virulent phages. Collectively, our findings provide insights into the genetic landscape of the human gut phageome.
Collapse
Affiliation(s)
- Senying Lai
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Huarui Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Peer Bork
- European Molecular Biology Laboratory, Structural and Computational Biology Unit, Heidelberg, Germany
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany
- Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany
| | - Wei-Hua Chen
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
- College of Life Science, Henan Normal University, Xinxiang, Henan, China
| | - Xing-Ming Zhao
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Ding J, Cui X, Wang X, Zhai F, Wang L, Zhu L. Multi-omics analysis of gut microbiota and metabolites reveals contrasting profiles in domestic pigs and wild boars across urban environments. Front Microbiol 2024; 15:1450306. [PMID: 39193431 PMCID: PMC11347354 DOI: 10.3389/fmicb.2024.1450306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
The gut microbiota plays a crucial role in host health and metabolism. This study explores the differences in gut microbiota and metabolites between domestic pigs (DP) and wild boars (WB) in urban environments. We analyzed gut microbial composition, metabolic profiles, virome composition, antibiotic resistance genes (ARGs), and human pathogenic bacteria (HPB) in both DP and WB. Our results revealed that DP exhibited a higher Firmicutes/Bacteroidetes ratio and were enriched in bacterial genera associated with domestication and modern feeding practices. Metabolomic analysis showed distinct profiles, with WB significantly enriched in the Pantothenate and CoA biosynthesis pathway, highlighting dietary and environmental influences on host metabolism. Additionally, DP had a distinct gut virome composition, particularly enriched in lytic phages of the Chaseviridae family. ARG analysis indicated a higher abundance of tetracycline resistance genes in DP, likely due to antibiotic use in pig farms. Furthermore, variations in HPB composition underscored potential health risks associated with contact with pig feces. These findings provide valuable insights into the microbial ecology of domestic pigs and wild boars, emphasizing the importance of these comparisons in identifying zoonotic pathogen transmission pathways and managing antibiotic resistance. Continued research in this area is essential for developing effective strategies to mitigate public health risks and promote sustainable livestock management practices.
Collapse
Affiliation(s)
- Jingjing Ding
- Jiangsu Academy of Forestry, Nanjing, China
- Jiangsu Yangzhou Urban Forest Ecosystem National Observation and Research Station, Yangzhou, China
| | - Xinyuan Cui
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuan Wang
- Jiangsu Academy of Forestry, Nanjing, China
- Jiangsu Yangzhou Urban Forest Ecosystem National Observation and Research Station, Yangzhou, China
| | - Feifei Zhai
- Jiangsu Wildlife Protection Station, Nanjing, China
| | - Lei Wang
- Jiangsu Academy of Forestry, Nanjing, China
- Jiangsu Yangzhou Urban Forest Ecosystem National Observation and Research Station, Yangzhou, China
| | - Lifeng Zhu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
42
|
Deleu S, Jacobs I, Vazquez Castellanos JF, Verstockt S, Trindade de Carvalho B, Subotić A, Verstockt B, Arnauts K, Deprez L, Vissers E, Lenfant M, Vandermeulen G, De Hertogh G, Verbeke K, Matteoli G, Huys GRB, Thevelein JM, Raes J, Vermeire S. Effect of Mutant and Engineered High-Acetate-Producing Saccharomyces cerevisiae var. boulardii Strains in Dextran Sodium Sulphate-Induced Colitis. Nutrients 2024; 16:2668. [PMID: 39203805 PMCID: PMC11357622 DOI: 10.3390/nu16162668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
Acetate-producing Saccharomyces cerevisiae var. boulardii strains could exert improved effects on ulcerative colitis, which here, was preclinically evaluated in an acute dextran sodium sulphate induced model of colitis. Nine-week-old female mice were divided into 12 groups, receiving either drinking water or 2.75% dextran sodium sulphate for 7 days, combined with a daily gavage of various treatments with different levels of acetate accumulation: sham control (phosphate buffered saline, no acetate), non-probiotic control (Baker's yeast, no acetate), probiotic control (Enterol®, transient acetate), and additionally several Saccharomyces cerevisiae var. boulardii strains with respectively no, high, and extra-high acetate accumulation. Disease activity was monitored daily, and feces samples were collected at different timepoints. On day 14, the mice were sacrificed, upon which blood and colonic tissue were collected for analysis. Disease activity in inflamed mice was lower when treated with the high-acetate-producing strain compared to sham and non-probiotic controls. The non-acetate-producing strain showed higher disease activity compared to the acetate-producing strains. Accordingly, higher histologic inflammation was observed in non- or transient-acetate-producing strains compared to the sham control, whereas this increase was not observed for high- and extra-high-acetate-producing strains upon induction of inflammation. These anti-inflammatory findings were confirmed by transcriptomic analysis of differentially expressed genes. Moreover, only the strain with the highest acetate production was superior in maintaining a stable gut microbial alpha-diversity upon inflammation. These findings support new possibilities for acetate-mediated management of inflammation in inflammatory bowel disease by administrating high-acetate-producing Saccharomyces cerevisae var. boulardii strains.
Collapse
Affiliation(s)
- Sara Deleu
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Inge Jacobs
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Jorge F. Vazquez Castellanos
- VIB-KU Leuven Center for Microbiology, 3001 Leuven, Belgium (G.R.B.H.)
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Sare Verstockt
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | | | - Ana Subotić
- NovelYeast bv, Bio-Incubator BIO4, Gaston Geenslaan 3, Leuven-Heverlee, 3001 Leuven, Belgium
| | - Bram Verstockt
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
- Department of Gastroenterology and Hepatology, UZ Leuven, KU Leuven, 3000 Leuven, Belgium
| | - Kaline Arnauts
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Lowie Deprez
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Eva Vissers
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Matthias Lenfant
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
- Department of Gastroenterology and Hepatology, UZ Leuven, KU Leuven, 3000 Leuven, Belgium
| | - Greet Vandermeulen
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Gert De Hertogh
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
- Laboratory of Morphology and Molecular Pathology, UZ Leuven, 3000 Leuven, Belgium
| | - Kristin Verbeke
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Gianluca Matteoli
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
| | - Geert R. B. Huys
- VIB-KU Leuven Center for Microbiology, 3001 Leuven, Belgium (G.R.B.H.)
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Johan M. Thevelein
- NovelYeast bv, Bio-Incubator BIO4, Gaston Geenslaan 3, Leuven-Heverlee, 3001 Leuven, Belgium
| | - Jeroen Raes
- VIB-KU Leuven Center for Microbiology, 3001 Leuven, Belgium (G.R.B.H.)
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, 3000 Leuven, Belgium
| | - Séverine Vermeire
- TARGID, Department of Chronic Diseases and Metabolism (CHROMETA), KU Leuven, 3000 Leuven, Belgium; (S.D.); (E.V.)
- Department of Gastroenterology and Hepatology, UZ Leuven, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
43
|
Endo HM, Bandeca SCS, Olchanheski LR, Schemczssen-Graeff Z, Pileggi M. Probiotics and the reduction of SARS-CoV-2 infection through regulation of host cell calcium dynamics. Life Sci 2024; 350:122784. [PMID: 38848939 DOI: 10.1016/j.lfs.2024.122784] [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: 01/16/2024] [Revised: 05/21/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Calcium is a secondary messenger that interacts with several cellular proteins, regulates various physiological processes, and plays a role in diseases such as viral infections. Next-generation probiotics and live biotherapeutic products are linked to the regulation of intracellular calcium levels. Some viruses can manipulate calcium channels, pumps, and membrane receptors to alter calcium influx and promote virion production and release. In this study, we examined the use of bacteria for the prevention and treatment of viral diseases, such as coronavirus of 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Vaccination programs have helped reduce disease severity; however, there is still a lack of well-recognized drug regimens for the clinical management of COVID-19. SARS-CoV-2 interacts with the host cell calcium (Ca2+), manipulates proteins, and disrupts Ca2+ homeostasis. This article explores how viruses exploit, create, or exacerbate calcium imbalances, and the potential role of probiotics in mitigating viral infections by modulating calcium signaling. Pharmacological strategies have been developed to prevent viral replication and block the calcium channels that serve as viral receptors. Alternatively, probiotics may interact with cellular calcium influx, such as Lactobacillus spp. The interaction between Akkermansia muciniphila and cellular calcium homeostasis is evident. A scientific basis for using probiotics to manipulate calcium channel activity needs to be established for the treatment and prevention of viral diseases while maintaining calcium homeostasis. In this review article, we discuss how intracellular calcium signaling can affect viral replication and explore the potential therapeutic benefits of probiotics.
Collapse
Affiliation(s)
- Hugo Massami Endo
- Environmental Microbiology Laboratory, Life Sciences and Health Institute, Structural and Molecular Biology, and Genetics Department, Ponta Grossa State University, Ponta Grossa, Brazil
| | | | - Luiz Ricardo Olchanheski
- Environmental Microbiology Laboratory, Life Sciences and Health Institute, Structural and Molecular Biology, and Genetics Department, Ponta Grossa State University, Ponta Grossa, Brazil
| | - Zelinda Schemczssen-Graeff
- Comparative Immunology Laboratory, Department of Microbiology, Parasitology and Pathology, Federal University of Paraná, Curitiba, Brazil
| | - Marcos Pileggi
- Environmental Microbiology Laboratory, Life Sciences and Health Institute, Structural and Molecular Biology, and Genetics Department, Ponta Grossa State University, Ponta Grossa, Brazil.
| |
Collapse
|
44
|
Sadeghi J, Zaib F, Heath DD. Genetic architecture and correlations between the gut microbiome and gut gene transcription in Chinook salmon (Oncorhynchus tshawytscha). Heredity (Edinb) 2024; 133:54-66. [PMID: 38822131 PMCID: PMC11222526 DOI: 10.1038/s41437-024-00692-3] [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: 02/05/2024] [Revised: 05/12/2024] [Accepted: 05/14/2024] [Indexed: 06/02/2024] Open
Abstract
Population divergence through selection can drive local adaptation in natural populations which has implications for the effective restoration of declining and extirpated populations. However, adaptation to local environmental conditions is complicated when both the host and its associated microbiomes must respond via co-evolutionary change. Nevertheless, for adaptation to occur through selection, variation in both host and microbiome traits should include additive genetic effects. Here we focus on host immune function and quantify factors affecting variation in gut immune gene transcription and gut bacterial community composition in early life-stage Chinook salmon (Oncorhynchus tshawytscha). Specifically, we utilized a replicated factorial breeding design to determine the genetic architecture (sire, dam and sire-by-dam interaction) of gut immune gene transcription and microbiome composition. Furthermore, we explored correlations between host gut gene transcription and microbiota composition. Gene transcription was quantified using nanofluidic qPCR arrays (22 target genes) and microbiota composition using 16 S rRNA gene (V5-V6) amplicon sequencing. We discovered limited but significant genetic architecture in gut microbiota composition and transcriptional profiles. We also identified significant correlations between gut gene transcription and microbiota composition, highlighting potential mechanisms for functional interactions between the two. Overall, this study provides support for the co-evolution of host immune function and their gut microbiota in Chinook salmon, a species recognized as locally adapted. Thus, the inclusion of immune gene transcription profile and gut microbiome composition as factors in the development of conservation and commercial rearing practices may provide new and more effective approaches to captive rearing.
Collapse
Affiliation(s)
- Javad Sadeghi
- Great Lakes Institute for Environmental Research, University of Windsor, Windsor, ON, Canada
- Department of Physical & Environmental Sciences, University of Toronto-Scarborough, Toronto, ON, Canada
| | - Farwa Zaib
- Great Lakes Institute for Environmental Research, University of Windsor, Windsor, ON, Canada
| | - Daniel D Heath
- Great Lakes Institute for Environmental Research, University of Windsor, Windsor, ON, Canada.
- Department of Integrative Biology, University of Windsor, Ontario, ON, Canada.
| |
Collapse
|
45
|
Cheng G, Liu Y, Guo R, Wang H, Zhang W, Wang Y. Molecular mechanisms of gut microbiota in diabetic nephropathy. Diabetes Res Clin Pract 2024; 213:111726. [PMID: 38844054 DOI: 10.1016/j.diabres.2024.111726] [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: 03/14/2024] [Revised: 05/10/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024]
Abstract
Diabetic nephropathy is a common complication of diabetes and a considerable contributor to end-stage renal disease. Evidence indicates that glucose dysregulation and lipid metabolism comprise a pivotal pathogenic mechanism in diabetic nephropathy. However, current treatment outcomes are limited, as they only provide symptomatic relief without preventing disease progression. The gut microbiota is a group of microorganisms that inhabit the human intestinal tract and play a crucial role in maintaining host energy balance, metabolism, and immune activity. Patients with diabetic nephropathy exhibit altered gut microbiota, suggesting its potential involvement in the onset and progression of the disease. However, how a perturbed microbiota induces and promotes diabetic nephropathy remains unelucidated. This article summarizes the evidence of the impact of gut microbiota on the progression of diabetic nephropathy, with a particular focus on the molecular mechanisms involved, aiming to provide new insights into the treatment of diabetic nephropathy.
Collapse
Affiliation(s)
- Gang Cheng
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China.
| | - YuLin Liu
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China.
| | - Rong Guo
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China.
| | - Huinan Wang
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730000, China.
| | - Wenjun Zhang
- Department of Nephrology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
| | - Yingying Wang
- Department of Nephrology, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
| |
Collapse
|
46
|
Bisle E, Varadarajan S, Kolassa IT. Vitamin-mediated interaction between the gut microbiome and mitochondria in depression: A systematic review-based integrated perspective. Brain Behav Immun Health 2024; 38:100790. [PMID: 38974216 PMCID: PMC11225645 DOI: 10.1016/j.bbih.2024.100790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/08/2024] [Accepted: 05/02/2024] [Indexed: 07/09/2024] Open
Abstract
Depression is one of the world's most prevalent mental disorders and its treatment remains suboptimal. Depression is a systemic disease with highly complex biological mechanisms. Emerging evidence points towards the involvement of mitochondria, microbiome and vitamins in its pathophysiology. Mitochondrial energy production was shown to be lowered in patients with depression. Mitochondrial energy production depends on vitamins, which are available from food, but are also synthesized by the gut microbiota. Several studies reported altered vitamin levels as well as changes in the gut microbiome composition and its vitamin metabolism in patients with depression. Therefore, the question of a connection between mitochondria and gut microbiome and vitamins influencing the mental health arises. This review aims to systematically investigate a combination of the topics - depression, mitochondria, microbiome, and vitamins - to generate an overview of a novel yet extremely complex and interconnected research field. A systematic literature search yielded 34 articles, and the results were summarized and bundled to develop this new integrative perspective on mitochondrial function mediated by the microbiome and microbiome-derived vitamins in depression. Furthermore, by discussing the research gaps this review aims to encourage innovative research approaches to better understand the biology of depression, which could result in optimized therapeutic approaches.
Collapse
Affiliation(s)
- Ellen Bisle
- Department of Clinical & Biological Psychology, Institute of Psychology & Education, Ulm University, Albert-Einstein-Allee 47, 89081, Ulm, Germany
| | - Suchithra Varadarajan
- Department of Clinical & Biological Psychology, Institute of Psychology & Education, Ulm University, Albert-Einstein-Allee 47, 89081, Ulm, Germany
| | - Iris-Tatjana Kolassa
- Department of Clinical & Biological Psychology, Institute of Psychology & Education, Ulm University, Albert-Einstein-Allee 47, 89081, Ulm, Germany
| |
Collapse
|
47
|
Yang Y, Chi L, Hsiao YC, Lu K. Sex-specific effects of gut microbiome on shaping bile acid metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601003. [PMID: 38979196 PMCID: PMC11230406 DOI: 10.1101/2024.06.27.601003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Gut microbiome is a group of microorganisms that plays important roles in contributing to health and diseases. These bacterial compositions have been demonstrated to impact bile acids (BAs) profiles, either by directly metabolizing primary BAs to secondary BAs or indirect ways through host metabolism by influencing BAs synthesis, transportation and conjugation in liver. It has been observed sexually dimorphic gut microbiome and bile acids composition, with variations in expression levels of bile acid metabolizing genes in the liver. However, associations between sex-specific differences in gut microbiome and BAs profiles are not well understood. This study aimed to investigate whether gut microbiome could influence BAs profiles in host in a sexspecific manner. We transplanted cecum feces of male and female C57BL/6 mice to male mice and measured BAs concentrations in feces, serum and liver samples 7 days after fecal transplantation. We found different BAs profiles between mice with male and female gut microbiome, including altering levels and proportions of secondary BAs. We also observed varied expression levels of genes related to bile acid metabolism in the liver and distal ileum. Our results highlight sex-specific effects of gut microbiome on shaping bile acid metabolism through gut bacteria and regulation of host genes.
Collapse
Affiliation(s)
- Yifei Yang
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Liang Chi
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Yun-Chung Hsiao
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, 27599, United States
| |
Collapse
|
48
|
Semenova N, Garashchenko N, Kolesnikov S, Darenskaya M, Kolesnikova L. Gut Microbiome Interactions with Oxidative Stress: Mechanisms and Consequences for Health. PATHOPHYSIOLOGY 2024; 31:309-330. [PMID: 39051221 PMCID: PMC11270257 DOI: 10.3390/pathophysiology31030023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/27/2024] Open
Abstract
Understanding how gut flora interacts with oxidative stress has been the subject of significant research in recent years. There is much evidence demonstrating the existence of the microbiome-oxidative stress interaction. However, the biochemical basis of this interaction is still unclear. In this narrative review, possible pathways of the gut microbiota and oxidative stress interaction are presented, among which genetic underpinnings play an important role. Trimethylamine-N-oxide, mitochondria, short-chain fatty acids, and melatonin also appear to play roles. Moreover, the relationship between oxidative stress and the gut microbiome in obesity, metabolic syndrome, chronic ethanol consumption, dietary supplements, and medications is considered. An investigation of the correlation between bacterial community features and OS parameter changes under normal and pathological conditions might provide information for the determination of new research methods. Furthermore, such research could contribute to establishing a foundation for determining the linkers in the microbiome-OS association.
Collapse
Affiliation(s)
- Natalya Semenova
- Scientific Centre for Family Health and Human Reproduction Problems, 664003 Irkutsk, Russia; (N.G.); (S.K.); (M.D.); (L.K.)
| | | | | | | | | |
Collapse
|
49
|
Breczko WJ, Bubak J, Miszczak M. The Importance of Intestinal Microbiota and Dysbiosis in the Context of the Development of Intestinal Lymphoma in Dogs and Cats. Cancers (Basel) 2024; 16:2255. [PMID: 38927960 PMCID: PMC11202240 DOI: 10.3390/cancers16122255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Recent advancements have significantly enhanced our understanding of the crucial role animal microbiomes play in veterinary medicine. Their importance in the complex intestinal environment spans immune modulation, metabolic homeostasis, and the pathogenesis of chronic diseases. Dysbiosis, a microbial imbalance, can lead to a range of diseases affecting both individual organs and the entire organism. Microbial disruption triggers inflammatory responses in the intestinal mucosa and disturbs immune homeostasis, increasing susceptibility to toxins and their metabolites. These dynamics contribute to the development of intestinal lymphoma, necessitating rigorous investigation into the role of microbiota in tumorigenesis. The principles explored in this study extend beyond veterinary medicine to encompass broader human health concerns. There are remarkable parallels between the subtypes of lymphoproliferative disorders in animals and humans, particularly Hodgkin's lymphoma and non-Hodgkin's lymphoma. Understanding the etiology of a cancer of the lymphatic system formation is critical for developing both preventive strategies and therapeutic interventions, with the potential to significantly improve patient outcomes. The aim of this study is to discuss the optimal composition of the microbiome in dogs and cats and the potential alterations in the microbiota during the development of intestinal lesions, particularly intestinal lymphoma. Molecular and cellular analyses are also incorporated to detect inflammatory changes and carcinogenesis. A review of the literature on the connections between the gut microbiome and the development of lymphomas in dogs and cats is presented, along with potential diagnostic approaches for these cancers.
Collapse
Affiliation(s)
- Wioleta Jadwiga Breczko
- EZA Student Science Club, The Faculty of Veterinary Medicine in Wroclaw, Wrocław University of Environmental and Life Sciences, 31 Norwida St., 50-375 Wrocław, Poland
| | - Joanna Bubak
- Department of Pathology, Division of Pathomorphology and Veterinary Forensics, The Faculty of Veterinary Medicine in Wrocław, Wrocław University of Environmental and Life Sciences, 31 Norwida St., 50-375 Wrocław, Poland;
| | - Marta Miszczak
- Department of Epizootiology and Clinic of Birds and Exotic Animals, Division of Infectious Diseases and Veterinary Administration, The Faculty of Veterinary Medicine in Wroclaw, Wrocław University of Environmental and Life Sciences, Grunwaldzki Sq. 45, 50-366 Wrocław, Poland;
| |
Collapse
|
50
|
Gong Z, Xue Q, Luo Y, Yu B, Hua B, Liu Z. The interplay between the microbiota and opioid in the treatment of neuropathic pain. Front Microbiol 2024; 15:1390046. [PMID: 38919504 PMCID: PMC11197152 DOI: 10.3389/fmicb.2024.1390046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/24/2024] [Indexed: 06/27/2024] Open
Abstract
Neuropathic pain (NP) is characterized by its complex and multifactorial nature and limited responses to opioid therapy; NP is associated with risks of drug resistance, addiction, difficulty in treatment cessation, and psychological disorders. Emerging research on gut microbiota and their metabolites has demonstrated their effectiveness in alleviating NP and augmenting opioid-based pain management, concurrently mitigating the adverse effects of opioids. This review addresses the following key points: (1) the current advances in gut microbiota research and the challenges in using opioids to treat NP, (2) the reciprocal effects and benefits of gut microbiota on NP, and (3) the interaction between opioids with gut microbiota, as well as the benefits of gut microbiota in opioid-based treatment of NP. Through various intricate mechanisms, gut microbiota influences the onset and progression of NP, ultimately enhancing the efficacy of opioids in the management of NP. These insights pave the way for further pragmatic clinical research, ultimately enhancing the efficacy of opioid-based pain management.
Collapse
Affiliation(s)
- Zexiong Gong
- Department of Anesthesiology, Health Science Center, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Qingsheng Xue
- Department of Anesthesiology, School of Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yan Luo
- Department of Anesthesiology, School of Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Buwei Yu
- Department of Anesthesiology, School of Medicine, Ruijin Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Bo Hua
- Department of Anesthesiology, Health Science Center, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zhiheng Liu
- Department of Anesthesiology, Health Science Center, Shenzhen Second People's Hospital/The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|