1
|
Wang H, Han J, Zhang XA. Interplay of m6A RNA methylation and gut microbiota in modulating gut injury. Gut Microbes 2025; 17:2467213. [PMID: 39960310 PMCID: PMC11834532 DOI: 10.1080/19490976.2025.2467213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 12/12/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
The gut microbiota undergoes continuous variations among individuals and across their lifespan, shaped by diverse factors encompassing diet, age, lifestyle choices, medication intake, and disease states. These microbial inhabitants play a pivotal role in orchestrating physiological metabolic pathways through the production of metabolites like bile acids, choline, short-chain fatty acids, and neurotransmitters, thereby establishing a dynamic "gut-organ axis" with the host. The intricate interplay between the gut microbiota and the host is indispensable for gut health, and RNA N6-methyladenosine modification, a pivotal epigenetic mark on RNA, emerges as a key player in this process. M6A modification, the most prevalent internal modification of eukaryotic RNA, has garnered significant attention in the realm of RNA epigenetics. Recent findings underscore its potential to influence gut microbiota diversity and intestinal barrier function by modulating host gene expression patterns. Conversely, the gut microbiota, through its impact on the epigenetic landscape of host cells, may indirectly regulate the recruitment and activity of RNA m6A-modifying enzymes. This review endeavors to delve into the biological functions of m6A modification and its consequences on intestinal injury and disease pathogenesis, elucidating the partial possible mechanisms by which the gut microbiota and its metabolites maintain host intestinal health and homeostasis. Furthermore, it also explores the intricate crosstalk between them in intestinal injury, offering a novel perspective that deepens our understanding of the mechanisms underlying intestinal diseases.
Collapse
Affiliation(s)
- Haixia Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Juanjuan Han
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Xin-An Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
2
|
Gustafson KL, Rodriguez TR, McAdams ZL, Coghill LM, Ericsson AC, Franklin CL. Failure of colonization following gut microbiota transfer exacerbates DSS-induced colitis. Gut Microbes 2025; 17:2447815. [PMID: 39812347 PMCID: PMC11740679 DOI: 10.1080/19490976.2024.2447815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/16/2025] Open
Abstract
To study the impact of differing specific pathogen-free gut microbiomes (GMs) on a murine model of inflammatory bowel disease, selected GMs were transferred using embryo transfer (ET), cross-fostering (CF), and co-housing (CH). Prior work showed that the GM transfer method and the microbial composition of donor and recipient GMs can influence microbial colonization and disease phenotypes in dextran sodium sulfate-induced colitis. When a low richness GM was transferred to a recipient with a high richness GM via CH, the donor GM failed to successfully colonize, and a more severe disease phenotype resulted when compared to ET or CF, where colonization was successful. By comparing CH and gastric gavage for fecal material transfer, we isolated the microbial component of this effect and determined that differences in disease severity and survival were associated with microbial factors rather than the transfer method itself. Mice receiving a low richness GM via CH and gastric gavage exhibited greater disease severity and higher expression of pro-inflammatory immune mediators compared to those receiving a high richness GM. This study provides valuable insights into the role of GM composition and colonization in disease modulation.
Collapse
Affiliation(s)
- Kevin L. Gustafson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
| | - Trevor R. Rodriguez
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
| | - Zachary L. McAdams
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- Molecular Pathogenesis and Therapeutics Program, University of Missouri, Columbia, MO, USA
| | - Lyndon M. Coghill
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- University of Missouri Bioinformatics and Analytics Core, University of Missouri, Columbia, MO, USA
| | - Aaron C. Ericsson
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- University of Missouri College of Veterinary Medicine, Columbia, MO, USA
- University of Missouri Metagenomics Center, Columbia, MO, USA
| | - Craig L. Franklin
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, USA
- Comparative Medicine Program, University of Missouri, Columbia, MO, USA
- MU Mutant Mouse Resource and Research Center, University of Missouri, Columbia, MO, USA
- University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| |
Collapse
|
3
|
Wang XN, Liu JQ, Ji WL, Huo ZL, Liu LF, Zheng JY. Characterization of trimethylamine metabolic pathways using pseudo-targeted metabolomics. J Pharm Biomed Anal 2025; 258:116737. [PMID: 39919464 DOI: 10.1016/j.jpba.2025.116737] [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] [Revised: 01/20/2025] [Accepted: 02/03/2025] [Indexed: 02/09/2025]
Abstract
Trimethylamine (TMA) metabolism comprises choline-containing compounds' metabolization, TMA production and trimethylamine N-oxide (TMAO) generation. However, the presence of numerous compounds in the carnitine and phosphatidylcholine (PC) pool compositions complicates profiling work significantly. This study is aimed at developing an efficient method for profiling TMA metabolic pathways, including quantifying known compounds and semi-quantifying the differential metabolites in the carnitine and PC pool compositions. Pseudo-targeted metabolomics is applicable for characterization. Firstly, multivariate statistics were performed to identify valuable metabolites (variable importance in the projection >1) from quality control biological samples. Given that TMA metabolism involved in host-gut microbiota interaction, co-metabolites were defined as the intersections of valuable metabolites from different biological samples (serum, liver, and intestinal contents) and further screened. Finally, alterations in TMA metabolism were observed in dextran sulfate sodium-induced colitis, with semi-quantitative analysis for excavated co-metabolites including 11 PCs, 6 lyso-phosphatidylcholines, and 2 acylcarnitines and quantitative analysis for 10 known metabolites. The findings revealed increased TMA production and accumulation of choline-containing compounds in the gut during ulcerative colitis exacerbation. Correspondingly, the circulating level of TMAO was elevated in the colitis group. A comprehensive understanding of TMA metabolism can contribute to disease differential diagnoses and potential mechanism studies.
Collapse
Affiliation(s)
- Xin-Nan Wang
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, China; Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Jian-Qun Liu
- Key Laboratory of Modern Preparation of TCM, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, No. 818 Xingwan Road, Nanchang, Jiangxi 330004, China
| | - Wen-Liang Ji
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Zong-Li Huo
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Li-Fang Liu
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, China.
| | - Jia-Yi Zheng
- State Key Laboratory of Natural Medicines, Department of Chinese Medicines Analysis, School of Traditional Chinese Pharmacy, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, China.
| |
Collapse
|
4
|
Xia J, Wang Y, Li X, Liu L, Zhang P, Dai W, Luo P, Wang G, Li Y. The mechanism of perilla oil in regulating lipid metabolism. Food Chem 2025; 476:143318. [PMID: 39977980 DOI: 10.1016/j.foodchem.2025.143318] [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: 09/18/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/22/2025]
Abstract
Emerging science supports the role of lipid metabolism disorders in the occurrence and development of chronic diseases. Dietary intervention has been shown to be an effective strategy for regulating lipid metabolism. Recent studies showed that perilla is rich in various effective ingredients, including fatty acids, flavonoids, and phenolic acids. These ingredients exhibit a myriad of benefits, notably enhancing intestinal health and helping to manage metabolic diseases. Perilla oil stands out as a promising agent for regulating lipid metabolism, underscoring its potential for various health applications. This review introduces the active ingredients in perilla and provides a systematic overview of the mechanism by which perilla oil regulates lipid metabolism to expand its application value. Further research should focus on exploring the dose effect and absorption efficiency of perilla oil in clinical applications.
Collapse
Affiliation(s)
- Jiawei Xia
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China; Guizhou Rapeseed Institute, Guizhou Province Academy of Agricultural Sciences, No. 270-0061 Baiyun Road, Jinyang District, Guiyang, Guizhou 550008, China
| | - Yi Wang
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Xin Li
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Li Liu
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Pin Zhang
- Guizhou Rapeseed Institute, Guizhou Province Academy of Agricultural Sciences, No. 270-0061 Baiyun Road, Jinyang District, Guiyang, Guizhou 550008, China
| | - Wendong Dai
- Guizhou Rapeseed Institute, Guizhou Province Academy of Agricultural Sciences, No. 270-0061 Baiyun Road, Jinyang District, Guiyang, Guizhou 550008, China
| | - Peng Luo
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China
| | - Guoze Wang
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China.
| | - Yanhong Li
- The Affiliated Hospital of Guizhou Medical University, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, School of Public Health, Guizhou Medical University, Guiyang 561113, China.
| |
Collapse
|
5
|
Han L, Liu W, Yuan F, Liu Q, Cheng H, Jin X, Sun Y. Integration of microbiomics and metabolomics reveals energy metabolism imbalance in crucian carp (Carassius auratus) under saline-alkaline exposure. Comp Biochem Physiol C Toxicol Pharmacol 2025; 291:110145. [PMID: 39983937 DOI: 10.1016/j.cbpc.2025.110145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 02/23/2025]
Abstract
The ecological conditions of freshwater aquaculture are deteriorating by degrees in recent years. Consequently, the comprehensive utilization of saline-alkaline water has garnered increasing societal attention. Here, crucian carp (Carassius auratus) were exposed to 20, 40 mmol/L NaHCO3 for 30 days (T, F group). Metabolomic analyses were conducted using UPLC-QTOF/MS, complemented by biochemical and microbiology profiling to elucidate the damage of the saline environment to the intestinal microbial structure, which in turn interfered with the energy metabolism. It was observed that carbonate alkalinity (CA) exposure not only caused intestine oxidative stress but also changed the levels of several digestive enzymes, including α-amylase (AMS), chymotrypsin (CHY), lipase (LPS). Metabolomic analysis identified 22 different metabolites (DEMs) in T group and 77 DEMs in F group. MetaboAnalyst analysis indicated that these metabolites are primarily involved in energy-related pathways, including the citric acid cycle, galactose metabolism, and glycine, serine, and threonine metabolism. Intestinal microbial diversity and community composition were altered under carbonate alkalinity exposure, with increase in Proteobacteria abundance and decline in Firmicutes, abundance alongside enrichment of Sphingomonas. Herein, saline-alkaline stress disrupted the physiological homeostasis of the crucian carp intestine, leading to microbial dysbiosis and energy metabolic imbalance. This study provides a theoretical foundation for understanding the stress response of the crucian carp intestine and the role of the intestinal microbiome in host resilience under adverse environmental conditions.
Collapse
Affiliation(s)
- Lin Han
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences/Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; Department of Food Science and Engineering, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Wenzhi Liu
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences/Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; Department of Food Science and Engineering, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Fangying Yuan
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences/Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; Department of Chemical Engineering and Technology, College of Materials and Chemical Engineering, Harbin University of Science and Technology, Harbin 150080, China
| | - Qianwen Liu
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences/Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; Department of Food Science and Engineering, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Hongyu Cheng
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences/Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; Department of Food Science and Engineering, School of Food Science and Engineering, Dalian Ocean University, Dalian 116023, China
| | - Xiaofeng Jin
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences/Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; Department of Chemical Engineering and Technology, College of Materials and Chemical Engineering, Harbin University of Science and Technology, Harbin 150080, China
| | - Yanchun Sun
- Heilongjiang River Fisheries Research Institute of Chinese Academy of Fishery Sciences/Laboratory of Quality & Safety Risk Assessment for Aquatic Products (Harbin), Ministry of Agriculture and Rural Areas, Harbin 150070, China; Department of Food Science and Engineering, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China.
| |
Collapse
|
6
|
Ni X, Li J, Xiong H, Deng Z, Sun Y. Influence of fatty acid distribution on lipid metabolism and cognitive development in first-weaned mice. Food Res Int 2025; 209:116292. [PMID: 40253195 DOI: 10.1016/j.foodres.2025.116292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/18/2025] [Accepted: 03/13/2025] [Indexed: 04/21/2025]
Abstract
There are significant structural differences between breast milk fat and the fat found in existing infant formulas, and these differences may partly explain the observed variations in growth and development between breastfed and formula-fed infants. This study used mice compared three groups: a control group (mixed vegetable oil), an OPO group (vegetable oil added with OPO), and a human milk fat substitute (HMFS) group formulated to match the fatty acid composition of breast milk. Compared to the control group and OPO group, HMFS-fed mice exhibited reduced body fat content and improved cognitive abilities. Lipidomics studies revealed that these differences in HMFS mice were associated with downregulation of hepatic glycerolipids and upregulation of glycerophospholipids and sphingolipids, facilitating the delivery of long-chain polyunsaturated fatty acids to the brain. Molecular investigations confirmed that HMFS reduces body fat accumulation by inhibiting endogenous fatty acid synthesis and promoting fatty acid β-oxidation, while changes in hepatic lipid profiles result from lipid molecule synthesis and interconversion. Metataxonomic studies demonstrated that HMFS reshaped the gut microbiota, including upregulating Akkermansia and downregulating Desulfovibrio and the Firmicutes/Bacteroidetes ratio, with strong correlations observed between the change of gut microbiota and responded lipids in liver. Overall, the breast milk's unique fatty acid distribution promotes organismal growth by modulating hepatic lipid metabolism, systemic lipid circulation, and gut microbiota. These findings underscore the nutritional benefits of breast milk fat structure and provide insights for the development of next-generation infant formulas.
Collapse
Affiliation(s)
- Xinggang Ni
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Jing Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Hua Xiong
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China
| | - Yong Sun
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, Jiangxi 330047, China.
| |
Collapse
|
7
|
Zhang Q, Hutchison ER, Pan C, Warren MF, Keller MP, Attie AD, Lusis AJ, Rey FE. Systems genetics uncovers associations among host amylase locus, gut microbiome, and metabolic traits in mice. MICROBIOME 2025; 13:101. [PMID: 40259344 PMCID: PMC12012960 DOI: 10.1186/s40168-025-02093-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 03/16/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Population studies have revealed associations between host genetic and gut microbiome in humans and mice. However, the molecular bases for how host genetic variation impacts the gut microbial community and bacterial metabolic niches remain largely unknown. RESULTS We leveraged 90 inbred hyperlipidemic mouse strains from the hybrid mouse diversity panel (HMDP), previously studied for a variety of cardio-metabolic traits. Metagenomic analysis of cecal DNA followed by genome-wide association analysis identified genomic loci that were associated with microbial enterotypes in the gut. Among these, we detected a genetic locus surrounding multiple amylase genes that were associated with abundances of Firmicutes (Lachnospiraceae family) and Bacteroidetes (Muribaculaceae family) taxa encoding distinct starch and sugar degrading capabilities. The genetic variants at the amylase gene locus were associated with distinct gut microbial communities (enterotypes) with different predicted metabolic capacities for carbohydrate degradation. Mendelian randomization analysis revealed host phenotypes, including liver fibrosis and plasma HDL-cholesterol levels, that were associated with gut microbiome enterotypes. CONCLUSIONS This work reveals novel relationships among host genetic variation, gut microbial enterotypes, and host metabolic traits and supports the notion that variation of host amylase may represent a key determinant of gut microbiome in mice. Video Abstract.
Collapse
Affiliation(s)
- Qijun Zhang
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Evan R Hutchison
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Calvin Pan
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Matthew F Warren
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA.
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
8
|
Deng Y, Zhang Y, Xiao J, Cao Y, Ho CT, Lu M. Allicin Improves Diet-Induced Nonalcoholic Steatohepatitis and Gut Microbiota Dysbiosis in Mice via the Involvement of the Circadian Clock Gene Rev-erbα. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:9019-9032. [PMID: 40168418 DOI: 10.1021/acs.jafc.4c12566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Nonalcoholic Steatohepatitis (NASH) is a progressive liver disease characterized by inflammation and liver damage. Allicin, a bioactive compound derived from garlic, has demonstrated anti-inflammatory and antioxidant properties. This study explores the effects of allicin on NASH and gut microbiota dysbiosis induced by a high-fat, high-fructose diet (HFFD) in mice. Allicin supplementation significantly alleviated hepatic inflammation, improved glucose metabolism, and modulated the circadian rhythm gene Rev-erbα, which plays a critical role in regulating inflammation. The anti-inflammatory effects of allicin were diminished in Si-Rev-erbα-treated HepG2 cells, highlighting the importance of circadian regulation in mediating these effects. Allicin's anti-inflammatory effects were associated with increased levels of short-chain fatty acids (SCFAs) and the restoration of diurnal oscillations in proinflammatory cytokines and gut microbiota composition, particularly in genera, such as Akkermansia, Bacteroidetes, and Lactobacillus. These findings suggest that allicin could be a promising therapeutic approach for managing NASH, liver dysfunction, and related metabolic disorders through the modulation of circadian rhythms and the gut microbiome.
Collapse
Affiliation(s)
- Yupei Deng
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yiyi Zhang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, New Jersey 08901, United States
| | - Muwen Lu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
9
|
Cai M, Yang L, Zhong L, Xie S, Hu Y. Response patterns and community assembly processes of gut microbiota in grass carp subjected to various protein sources and their implications for growth and metabolism. FISH PHYSIOLOGY AND BIOCHEMISTRY 2025; 51:83. [PMID: 40238018 DOI: 10.1007/s10695-025-01498-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Feed nutrients are crucial in shaping the gut microbial community, especially for complex interactions. While much research focused on the impacts of dietary protein levels, exploration of protein sources remains insufficient. Accordingly, this study specifically investigated the effects of four protein sources [Clostridium autoethanolicum protein (CAP), cottonseed protein concentrate (CPC), Chlorella vulgaris meal (CVP), and Tenebrio molitor meal (TM)] replacing dietary soybean meal on microbial co-occurrence networks and key metabolic taxa. A 56-day feeding trial involved 1500 grass carp (20.00 g) fed five experimental diets, each incorporating one of the experimental protein sources. Results revealed that CPC and CVP diets improved the weight gain and specific growth rate, with the CPC group demonstrating the highest biomass gain and the CVP group exhibiting the best feed conversion ratio. Findings further indicated that SM-free diets enhanced intestinal immunity and barrier function while negatively impacting microbial diversity. Additional profiling revealed that each treatment exhibited distinct abundance profiles and unique species, with Firmicutes, Bacteroidota, and Proteobacteria as the dominant phyla and key genera such as Bacteroides, Erysipelatoclostridium, and Cetobacterium. Stochastic mechanisms drove the community assembly process, and prolonged SM-free diets led to simplified networks with increased generalists and specialists. Functional gene analysis highlighted roles in amino acid, carbohydrate, and lipid metabolism, underscoring the impact of protein sources on aquatic microbial communities and host-microbiome interactions. Overall, the study suggests the potential suitability of several protein sources as soybean meal substitutes, emphasizing the importance of further investigation into optimal inclusion levels for diverse proteins.
Collapse
Affiliation(s)
- Minglang Cai
- Fisheries College, Hunan Agricultural University, Changsha, China
| | - Linlin Yang
- Fisheries College, Hunan Agricultural University, Changsha, China
| | - Lei Zhong
- Fisheries College, Hunan Agricultural University, Changsha, China
| | - Shouqi Xie
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Yi Hu
- Fisheries College, Hunan Agricultural University, Changsha, China.
| |
Collapse
|
10
|
Fang X, Zhang Y, Huang X, Miao R, Zhang Y, Tian J. Gut microbiome research: Revealing the pathological mechanisms and treatment strategies of type 2 diabetes. Diabetes Obes Metab 2025. [PMID: 40230225 DOI: 10.1111/dom.16387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/19/2025] [Accepted: 03/23/2025] [Indexed: 04/16/2025]
Abstract
The high prevalence and disability rate of type 2 diabetes (T2D) caused a huge social burden to the world. Currently, new mechanisms and therapeutic approaches that may affect this disease are being sought. With in-depth research on the pathogenesis of T2D and growing advances in microbiome sequencing technology, the association between T2D and gut microbiota has been confirmed. The gut microbiota participates in the regulation of inflammation, intestinal permeability, short-chain fatty acid metabolism, branched-chain amino acid metabolism and bile acid metabolism, thereby affecting host glucose and lipid metabolism. Interventions focusing on the gut microbiota are gaining traction as a promising approach to T2D management. For example, dietary intervention, prebiotics and probiotics, faecal microbiota transplant and phage therapy. Meticulous experimental design and choice of analytical methods are crucial for obtaining accurate and meaningful results from microbiome studies. How to design gut microbiome research in T2D and choose different machine learning methods for data analysis are extremely critical to achieve personalized precision medicine.
Collapse
Affiliation(s)
- Xinyi Fang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Yanjiao Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyue Huang
- First Clinical Medical College, Changzhi Medical College, Shanxi, China
| | - Runyu Miao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Beijing, China
| | - Yuxin Zhang
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiaxing Tian
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
11
|
Shi S, Gan X, Qian Y, Cao Y, Wang Y, Shi C, Bi J, Yu Q, Han Q, Qu J, Li H. Metatranscriptomics reveals that plant tannins regulate the expression of intestinal antibiotic resistance genes in Qinghai voles (Neodon fuscus). ENVIRONMENTAL RESEARCH 2025; 271:121119. [PMID: 39952456 DOI: 10.1016/j.envres.2025.121119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025]
Abstract
Antibiotic resistance genes (ARGs) are a persistent harmful environmental pollutant, epidemic of ARGs thought to be a result of antibiotic misuse. Tannin acid (TA) is a natural plant compounds with bactericidal properties. Nowadays, TA is considered to be a potential replacement of antibiotics. However, the role of TA on ARGs is also not yet clear. To address this knowledge gap, we fed the model plateau animal Qinghai voles (Neodon fuscus) with different concentrations of TA. We used 16S rDNA sequencing for revealing total bacteria, 16S rRNA sequencing for revealing active bacteria, and metatranscriptomics (active function) sequencing for revealing ARGs and other functions. Our results showed that although TA reduced macrolide ARGs, TA group enriched 6-fold for tetracycline ARGs, 3-fold for multidrug ARGs, and 5-fold for aminoglycoside ARGs compared with control group. Moreover, TA reduced animal growth performance, and regulated gut microbiome more stable by improving microbial diversity. And TA promoted the production of short-chain fatty acids by gut microbes, such as lactate and acetate. This study reveals modulation of ARGs and gut microbiome by TA and also provides scientific value for the proper use of TA in feed and medical treatment.
Collapse
Affiliation(s)
- Shunqin Shi
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Xueying Gan
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Yuan Qian
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Yiming Cao
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Yu Wang
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Chenwei Shi
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Jie Bi
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Qiaoling Yu
- State Key Laboratory of Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, 730000, China
| | - Qian Han
- School of Public Health, Lanzhou University, Lanzhou, 730000, China
| | - Jiapeng Qu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, 810008, China
| | - Huan Li
- School of Public Health, Lanzhou University, Lanzhou, 730000, China; Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai, 810008, China; State Key Laboratory of Grassland Agro-ecosystems, Center for Grassland Microbiome, College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
12
|
Qu Q, Liu M, Hu Y, Huang G, Xuan Z, Lun J, Chen X, Lv W, Guo S. Modulatory effects of polyherbal mixture on the immuno-antioxidant capacity and intestinal health of chicks infected with Escherichia coli O78. Poult Sci 2025; 104:105156. [PMID: 40239311 DOI: 10.1016/j.psj.2025.105156] [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: 01/06/2025] [Revised: 04/02/2025] [Accepted: 04/11/2025] [Indexed: 04/18/2025] Open
Abstract
A total of 180 one-day-old white-feathered broiler chicks were selected and randomly divided into 4 treatments, namely the control group (CON), Escherichia coli groups (E. coli), 2 g/kg polyherbal mixture group (PHM2), and the 4 g/kg polyherbal mixture group (PHM4). The CON and E. coli groups were fed a basal diet, while the PHM2 and PHM4 groups were fed the basal diet supplemented with 2 g/kg and 4 g/kg PHM, respectively. Each group had 3 replicates, with 15 broilers per replicate. On day 17 of the experiment, broilers in the E. coli, PHM2, and PHM4 groups were intraperitoneally injected with 0.8 mL of 1 × 108 CFU/mL of E. coli O78. Broilers in the control group received an equivalent volume of saline. Chicks were euthanized 48 h postinjection for collecting serum, liver, spleen, jejunum, ileum, ileal mucosa, and cecal contents. Our results showed that PHM significantly reversed the weight loss and decreased the diarrhea rate and the mortality of chicks caused by E. coli infection (P < 0.05). In the serum of chicks infected with E. coli, PHM significantly enhanced the antioxidant capacity (P < 0.05), increased the levels of immunoglobulins and anti-inflammatory cytokines (P < 0.05), and decreased the concentrations of proinflammatory cytokines (P < 0.05). Meanwhile, PHM also promoted the mRNA expression of antioxidant-related genes and decreased the expression of proinflammatory cytokines and apoptosis-related genes in the liver, spleen, jejunum, and ileum (P < 0.05). In addition, PHM repaired the intestinal barrier and injury to further reduce the serum concentrations of d-lactate (DAO) and lipopolysaccharide (LPS) (P < 0.05). More importantly, PHM significantly regulated the composition of cecal microbiota, especially by up-regulating the relative abundance of beneficial bacteria, including Faecalibacterium, Bacteroides, Butyricicoccus, and Lactobacillus, and down-regulating the relative abundance of pathogenic bacteria, including Enterococcus, Escherichia, and Shigella (P < 0.05). These beneficial bacteria were significantly positively correlated with antioxidant capacity and intestinal barrier function, while pathogenic bacteria were significantly positively correlated with proinflammatory cytokines (P < 0.05). In conclusion, PHM may be a potential preventive strategy for E. coli-infected poultry, which is closely related to its modulation of gut microbiota.
Collapse
Affiliation(s)
- Qian Qu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Mengjie Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; School of Animal Science and Technology, Foshan University, Foshan, China
| | - Yifan Hu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Gengxiong Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Zhaoying Xuan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Jianchi Lun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaoli Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Weijie Lv
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; Guangdong Technology Research Center for Traditional Chinese Veterinary Medicine and Natural Medicine, Guangzhou, China
| | - Shining Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China; Guangdong Technology Research Center for Traditional Chinese Veterinary Medicine and Natural Medicine, Guangzhou, China.
| |
Collapse
|
13
|
Li Y, Zhang M, Zhang K, Niu H, Li H, Wu W. Ginsenosides modulate immunity via TLR4/MyD88/NF-κB pathway and gut microbiota. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156763. [PMID: 40252438 DOI: 10.1016/j.phymed.2025.156763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Ginsenosides, the primary active compounds in Panax ginseng C. A. Mey., are well known for their potent immunomodulatory effects. However, their precise mechanisms, particularly concerning the "intestinal-metabolism-immune axis", have yet to be fully elucidated. PURPOSE This study aims to investigate how ginsenosides protect immune function through the regulation of the gut-metabolism-immune axis. STUDY DESIGN A CTX-induced immunodeficient mouse model was established to assess the effects of ginsenosides on immune function, gut microbiota, and metabolic pathways. METHODS The immune organ indices (spleen and thymus), levels of immune cytokines (TNF-α, IFN-γ, IL-6, IL-1β), and immunoglobulins (IgM, IgA) were assessed. Intestinal microbial diversity was analyzed using 16S rRNA sequencing, and metabolomics was employed to identify disruptions in amino acid and lipid metabolic pathways. Spearman correlation analysis and Western blotting were conducted to explore the involvement of the TLR4/MyD88/NF-κB signaling pathway. RESULTS Ginsenosides significantly restored immune organ indices and enhanced cytokines and immunoglobulins. 16S rRNA sequencing revealed an increase in probiotic levels and a reduction in potentially harmful bacteria, thereby enhancing intestinal microbiota diversity. Metabolomics analysis showed that ginsenosides ameliorated CTX-induced metabolic disorders and stimulated the production of short-chain fatty acids (SCFAs) and bile acids. Western blot analysis confirmed the upregulation of TLR4, MyD88, and NF-κB p-p65 expression. CONCLUSION This study systematically elucidates the mechanism by which ginsenosides enhance immune function by regulating gut microbiota, restoring metabolic balance, and activating the TLR4/MyD88/NF-κB signaling pathway. These findings provide a molecular foundation for the potential use of ginsenosides in the prevention and treatment of immune-related diseases.
Collapse
Affiliation(s)
- Ying Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China; Shandong Academy of Chinese Medicine, Jinan, Shandong, 250014, China
| | - Meng Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Kaiyue Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Huazhou Niu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China
| | - Hui Li
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China.
| | - Wei Wu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China.
| |
Collapse
|
14
|
Gopalakrishnan V, Kumar C, Robertsen I, Morehouse C, Sparklin B, Khader S, Henry I, Johnson LK, Hertel JK, Christensen H, Sandbu R, Greasley PJ, Sellman BR, Åsberg A, Andersson S, Löfmark RJ, Hjelmesæth J, Karlsson C, Cohen TS. A multi-omics microbiome signature is associated with the benefits of gastric bypass surgery and is differentiated from diet induced weight loss through 2 years of follow-up. Mucosal Immunol 2025:S1933-0219(25)00040-6. [PMID: 40222615 DOI: 10.1016/j.mucimm.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/04/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
Roux-en-Y gastric bypass (GBP) surgery is an effective treatment for reducing body weight and correcting metabolic dysfunction in individuals with severe obesity. Herein, we characterize the differences between very low energy diet (VLED) and GBP induced weight loss by multi-omic analyses of microbiome and host features in a non-randomized, controlled, single-center study. Eighty-eight participants with severe obesity were recruited into two arms - GBP versus VLED with matching weight loss for 6 weeks and 2-years of follow-up. A dramatic shift in the distribution of gut microbial taxa and their functional capacity was seen in the GBP group at Week 2 after surgery and was sustained through 2 years. Multi-omic analyses were performed after 6 weeks of matching weight loss between the GBP and VLED groups, which pointed to microbiome derived metabolites such as indoxyl sulphate as characterizing the GBP group. We also identified an inverse association between Streptococcus parasanguinis (an oral commensal) and plasma levels of tryptophan and tyrosine. These data have important implications, as they reveal a significant robust restructuring of the microbiome away from a baseline dysbiotic state in the GBP group. Furthermore, multi-omics modelling points to potentially novel mechanistic insights at the intersection of the microbiome and host.
Collapse
Affiliation(s)
| | - Chanchal Kumar
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| | - Ida Robertsen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, PO 1068 Blindern, 0316 Oslo, Norway
| | - Christopher Morehouse
- Discovery Microbiome, Early Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, USA
| | - Ben Sparklin
- Discovery Microbiome, Early Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, USA
| | - Shameer Khader
- Data Science and Artificial Intelligence, Biopharmaceuticals R&D, AstraZeneca, USA.
| | - Ian Henry
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Line Kristin Johnson
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, P.O.Box 2168, 3103 Tønsberg, Norway
| | - Jens K Hertel
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, P.O.Box 2168, 3103 Tønsberg, Norway
| | - Hege Christensen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, PO 1068 Blindern, 0316 Oslo, Norway
| | - Rune Sandbu
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, P.O.Box 2168, 3103 Tønsberg, Norway
| | - Peter J Greasley
- Early Clinical Development, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Bret R Sellman
- Discovery Microbiome, Early Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, USA
| | - Anders Åsberg
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, PO 1068 Blindern, 0316 Oslo, Norway; Department of Transplantation Medicine, Oslo University Hospital, P.O.Box 4950 Nydalen 0424 Oslo, Norway
| | - Shalini Andersson
- Oligonucleotide Discovery, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Rasmus Jansson Löfmark
- Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Jøran Hjelmesæth
- Department of Endocrinology, Obesity and Nutrition, Vestfold Hospital Trust, P.O.Box 2168, 3103 Tønsberg, Norway; Department of Endocrinology, Morbid Obesity and Preventive Medicine, Institute of Clinical Medicine, University of Oslo, P.O. Box 1171, 0318 Oslo, Norway
| | - Cecilia Karlsson
- Late-stage Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden; Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Taylor S Cohen
- Late Vaccines and Immune Therapies, Biopharmaceuticals R&D, AstraZeneca, USA.
| |
Collapse
|
15
|
Wang L, He X, Zhang Z, Chen N. Distinct gut microbiota signatures in older people with sarcopenic obesity and sarcopenia without obesity. Clin Nutr 2025; 49:77-89. [PMID: 40252601 DOI: 10.1016/j.clnu.2025.04.004] [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: 06/11/2024] [Revised: 03/15/2025] [Accepted: 04/04/2025] [Indexed: 04/21/2025]
Abstract
BACKGROUND Previous evidence suggests that gut dysbiosis plays an important role in the development and progression of sarcopenia and sarcopenic obesity (SO), but evidence supporting this association is lacking. Thus, this study aimed to investigate the characteristics of gut microbiota in older people with sarcopenia and SO. METHODS A total of 1558 older adults (age ≥65 years) from a community-based cohort in Shanghai, China, underwent sarcopenia screening using the SARC-F questionnaire, with 351 participants completing further assessment. On the basis of the Asian Working Group for Sarcopenia 2019 and the World Health Organization obesity criteria, 60 participants were categorized into three groups: SO (n = 20), sarcopenia without obesity (Sar, n = 18), and controls (Con, n = 22). Gut microbiota composition was analyzed using 16S rRNA sequencing (V3-V4 regions). RESULTS Significant differences in the diversity and composition of the gut microbiota were observed in the Sar and SO groups. A reduction in alpha diversity (Chao1 and ACE indices) was found in the SO group. Beta diversity based on unweighted Unifrac PCoA was significantly different among the three groups. LEfSe analysis identified 39 taxa with significant differential abundances across groups. The Sar group exhibited enrichment of Christensenellaceae_R-7_group, Alistipes, Ruminococcus, Odoribacter, Prevotellaceae_UCG-001, Hungatella, Family_XIII_AD3011_group, Anaerotruncus, Ruminiclostridium, and Oxalobacter, along with their high taxonomic classifications. Meanwhile, Enterobacteriaceae, Allisonella, and Peptoclostridium were enriched in the SO group. Feature selection via Boruta algorithm identified five and four discriminatory taxa to construct random forest models, effectively distinguishing individuals with Sar and SO from Con. Key predictors for Sar included reduced Enterococcus, Enterobacter, and Hungatella and increased Odoribacter and Christensenellaceae_R-7_group. Conversely, SO was characterized by decreased Enterobacter, Alloprevotella, and Enterococcus and increased Allisonella. Five-fold cross-validation confirmed robust diagnostic efficacy, achieving AUCs of 0.860 (95 % CI: 0.786-0.996) for Sar and 0.826 (95 % CI: 0.735-0.970) for SO. CONCLUSION This study demonstrated that the gut microbiota of SO and Sar have distinct diversity and composition profiles. The results provide new insights into the role of gut microbiota in SO, highlighting its potential as a therapeutic target in this condition.
Collapse
Affiliation(s)
- Ling Wang
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China; School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Xiangfeng He
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Zhen Zhang
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Nan Chen
- Department of Rehabilitation, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, China; Department of Rehabilitation, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
16
|
Arenas-Montes J, Alcala-Diaz JF, Garcia-Fernandez H, Gutierrez-Mariscal FM, Lopez-Moreno A, Luque-Cordoba D, Arenas-de Larriva AP, Torres-Peña JD, Luque RM, Prodam F, Priego-Capote F, Delgado-Lista J, Lopez-Miranda J, Camargo A. A microbiota pattern associated with cardiovascular events in secondary prevention: the CORDIOPREV study. Eur Heart J 2025:ehaf181. [PMID: 40197788 DOI: 10.1093/eurheartj/ehaf181] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/21/2024] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND AND AIMS Preventing new cardiovascular events in patients with established cardiovascular disease (CVD) is a daunting task for clinicians. Intestinal microbiota may help identify patients at risk, thus improving the strategies of secondary prevention. The aim of this study was to evaluate the baseline differences between the gut microbiota from coronary heart disease (CHD) patients suffering new major adverse cardiovascular events (MACEs) in the following 7 years, compared with CHD patients who did not undergo new MACE in this period, and to build a score associated with the risk of suffering new MACE. METHODS Within the framework of the CORDIOPREV study, a clinical trial that involved 1002 patients with CHD, intestinal microbiota was examined in patients with available faecal samples (n = 679, 132 MACE), through 16S metagenomics on the Illumina MiSeq and Quiime2 software. Lipopolysaccharide (LPS) was measured using limulus amoebocyte lysate test. RESULTS Random survival forest identified 10 bacterial taxa with a higher predictive power for MACE incidence. Receiver operating characteristic curves yielded an area under the curve of 65.2% (59.1%-71.3%) in the training set and 68.6% (59.3%-77.9%) in the validation set. The intestinal microbiota risk score was associated with a MACE incidence hazard ratio of 2.01 (95% confidence interval 1.37-3.22). Lipopolysaccharide analysis showed a greater LPS post-prandial fold change in the MACE group (P = .005). CONCLUSIONS These results reinforce the relationship between intestinal microbiota and CVD and suggest that a microbiota profile is associated with MACE in CHD patients, in addition to higher endotoxaemia.
Collapse
Affiliation(s)
- Javier Arenas-Montes
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Juan F Alcala-Diaz
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Helena Garcia-Fernandez
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Francisco M Gutierrez-Mariscal
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Alejandro Lopez-Moreno
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Diego Luque-Cordoba
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- Department of Analytical Chemistry, Annex Marie Curie Building, Campus of Rabanales, University of Cordoba, Cordoba 14071, Spain
- Consortium for Biomedical Research in Frailty & Healthy Ageing, CIBERFES, Carlos III Institute of Health, Madrid 28029, Spain
| | - Antonio P Arenas-de Larriva
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Jose D Torres-Peña
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Raul M Luque
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba 14071, Spain
| | - Flavia Prodam
- Department of Health Sciences, Unit of Endocrinology, Università del Piemonte Orientale, Novara 28100, Italy
| | - Feliciano Priego-Capote
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- Department of Analytical Chemistry, Annex Marie Curie Building, Campus of Rabanales, University of Cordoba, Cordoba 14071, Spain
- Consortium for Biomedical Research in Frailty & Healthy Ageing, CIBERFES, Carlos III Institute of Health, Madrid 28029, Spain
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Antonio Camargo
- Lipids and Atherosclerosis Unit, Department of Internal Medicine, Hospital Universitario Reina Sofía, Cordoba 14004, Spain
- Department of Medical and Surgical Sciences, Universidad de Cordoba, Cordoba 14004, Spain
- Maimonides Institute for Biomedical Research in Cordoba (IMIBIC), Cordoba 14004, Spain
- CIBER Fisiopatologia Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
17
|
Zhang Y, Si L, Shu X, Qiu C, Wan X, Li H, Ma S, Jin X, Wei Z, Hu H. Gut microbiota contributes to protection against porcine deltacoronavirus infection in piglets by modulating intestinal barrier and microbiome. MICROBIOME 2025; 13:93. [PMID: 40189556 PMCID: PMC11974153 DOI: 10.1186/s40168-025-02092-z] [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: 04/16/2024] [Accepted: 03/14/2025] [Indexed: 04/09/2025]
Abstract
BACKGROUND Gut microbiota plays a critical role in counteracting enteric viral infection. Our previous study demonstrated that infection of porcine deltacoronavirus (PDCoV) disturbs gut microbiota and causes intestinal damage and inflammation in piglets. However, the influence of gut microbiota on PDCoV infection remains unclear. RESULTS Firstly, the relationship between gut microbiota and disease severity of PDCoV infection was evaluated using 8-day-old and 90-day-old pigs. The composition of gut microbiota was significantly altered in 8-day-old piglets after PDCoV infection, leading to severe diarrhea and intestinal damage. In contrast, PDCoV infection barely affected the 90-day-old pigs. Moreover, the diversity (richness and evenness) of microbiota in 90-day-old pigs was much higher compared to the 8-day-old piglets, suggesting the gut microbiota is possibly associated with the severity of PDCoV infection. Subsequently, transplanting the fecal microbiota from the 90-day-old pigs to the 3-day-old piglets alleviated clinical signs of PDCoV infection, modulated the diversity and composition of gut microbiota, and maintained the physical and chemical barrier of intestines. Additionally, metabolomic analysis revealed that the fecal microbiota transplantation (FMT) treatment upregulated the swine intestinal arginine biosynthesis, FMT significantly inhibited the inflammatory response in piglet intestine by modulating the TLR4/MyD88/NF-κB signaling pathway. CONCLUSIONS PDCoV infection altered the structure and composition of the gut microbiota in neonatal pigs. FMT treatment mitigated the clinical signs of PDCoV infection in the piglets by modulating the gut microbiota composition and intestinal barrier, downregulating the inflammatory response. The preventive effect of FMT provides novel targets for the development of therapeutics against enteropathogenic coronaviruses. Video Abstract.
Collapse
Affiliation(s)
- Yunfei Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Lulu Si
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Xiangli Shu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Congrui Qiu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Xianhua Wan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
| | - Haiyan Li
- College of Sport, Yan'an University, Yanan, 716000, People's Republic of China
| | - Shijie Ma
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
- Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou, 450046, People's Republic of China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, People's Republic of China
| | - Xiaohui Jin
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China
- Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou, 450046, People's Republic of China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, People's Republic of China
| | - Zhanyong Wei
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China.
- Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou, 450046, People's Republic of China.
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, People's Republic of China.
- Longhu Laboratory of Henan Province, Zhengzhou, 450046, People's Republic of China.
| | - Hui Hu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, People's Republic of China.
- Henan Province Key Laboratory for Animal Food Pathogens Surveillance, Zhengzhou, 450046, People's Republic of China.
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou, 450046, People's Republic of China.
- Longhu Laboratory of Henan Province, Zhengzhou, 450046, People's Republic of China.
| |
Collapse
|
18
|
Wang Y, Li Y, Lin Y, Cao C, Chen D, Huang X, Li C, Xu H, Lai H, Chen H, Zhou Y. Roles of the gut microbiota in hepatocellular carcinoma: from the gut dysbiosis to the intratumoral microbiota. Cell Death Discov 2025; 11:140. [PMID: 40185720 PMCID: PMC11971373 DOI: 10.1038/s41420-025-02413-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/23/2025] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is closely linked to alterations in the gut microbiota. This dysbiosis is characterized by significant changes in the microbial population, which correlate with the progression of HCC. Gut dysbiosis ultimately promotes HCC development in several ways: it damages the integrity of the gut-vascular barrier (GVB), alters the tumor microenvironment (TME), and even affects the intratumoral microbiota. Subsequently, intratumoral microbiota present a characteristic profile and play an essential role in HCC progression mainly by causing DNA damage, mediating tumor-related signaling pathways, altering the TME, promoting HCC metastasis, or through other mechanisms. Both gut microbiota and intratumoral microbiota have dual effects on HCC progression; a comprehensive understanding of their complex biological roles will provide a theoretical foundation for potential clinical applications in HCC treatment.
Collapse
Affiliation(s)
- Yiqin Wang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yongqiang Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yong Lin
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Chuangyu Cao
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Dongcheng Chen
- Department of Gastroenterology and Hepatology, Baiyun Hospital of Guangzhou First People's Hospital (The Second People's Hospital of Baiyun District), Guangzhou, China
| | - Xianguang Huang
- Department of Gastroenterology and Hepatology, Baiyun Hospital of Guangzhou First People's Hospital (The Second People's Hospital of Baiyun District), Guangzhou, China
| | - Canhua Li
- Department of Gastroenterology and Hepatology, Baiyun Hospital of Guangzhou First People's Hospital (The Second People's Hospital of Baiyun District), Guangzhou, China
| | - Haoming Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huasheng Lai
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Huiting Chen
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
| | - Yongjian Zhou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
| |
Collapse
|
19
|
Larnder AH, Manges AR, Murphy RA. The estrobolome: Estrogen-metabolizing pathways of the gut microbiome and their relation to breast cancer. Int J Cancer 2025. [PMID: 40177842 DOI: 10.1002/ijc.35427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
Increasing evidence links the gut microbiome to carcinogenesis. Disruptions in estrogen regulation by the estrobolome-gut microbiota with estrogen-related functions-may promote breast cancer. However, precise information on estrobolome targets and their underlying mechanisms is limited. This review identifies relevant targets for measuring the estrobolome, focusing on enzymes and microbial taxa involved in processing estrogens, precursors, metabolites, and phytoestrogens, to facilitate the exploration of potential links to breast cancer. Evidence from breast cancer case-control studies is synthesized to assess alignment with these targets, highlight gaps in the evidence, and suggest new paths forward. Findings from case-control studies were heterogeneous and showed limited alignment with estrobolome targets, with only Escherichia coli and Roseburia inulinivorans identified as differentially abundant and functionally relevant between cases and controls. The lack of compelling evidence for estrobolome-specific mechanisms may reflect measurement challenges or may suggest that broader ecological changes in the microbiome, which influence a network of interacting mechanisms, are more influential for carcinogenesis. To clarify the estrobolome's role in breast cancer, future research should use advanced sequencing techniques and methods such as metabolomics and transcriptomics, while considering clinical and behavioral factors that may modify estrobolome mechanisms.
Collapse
Affiliation(s)
- Ashley H Larnder
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Amee R Manges
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
- British Columbia Centre for Disease Control, Vancouver, Canada
| | - Rachel A Murphy
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
- Cancer Control Research, BC Cancer, Vancouver, Canada
| |
Collapse
|
20
|
Roessler J, Zimmermann F, Heidecker B, Landmesser U, Haghikia A. Gut microbiota-related modulation of immune mechanisms in post-infarction remodelling and heart failure. ESC Heart Fail 2025; 12:942-954. [PMID: 39385474 PMCID: PMC11911630 DOI: 10.1002/ehf2.14991] [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: 05/03/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 10/12/2024] Open
Abstract
The immune system has long been recognized as a key driver in the progression of heart failure (HF). However, clinical trials targeting immune effectors have consistently failed to improve patient outcome across different HF aetiologies. The activation of the immune system in HF is complex, involving a broad network of pro-inflammatory and immune-modulating components, which complicates the identification of specific immune pathways suitable for therapeutic targeting. Increasing attention has been devoted to identifying gut microbial pathways that affect cardiac remodelling and metabolism and, thereby impacting the development of HF. In particular, gut microbiota-derived metabolites, absorbed by the host and transported to the peripheral circulation, can act as signalling molecules, influencing metabolism and immune homeostasis. Recent reports suggest that the gut microbiota plays a crucial role in modulating immune processes involved in HF. Here, we summarize recent advances in understanding the contributory role of gut microbiota in (auto-)immune pathways that critically determine the progression or alleviation of HF. We also thoroughly discuss potential gut microbiota-based intervention strategies to treat or decelerate HF progression.
Collapse
Affiliation(s)
- Johann Roessler
- University Hospital St Josef‐Hospital Bochum, Cardiology and RhythmologyRuhr University BochumBochumGermany
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
| | - Friederike Zimmermann
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
| | - Bettina Heidecker
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
- Berlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
- Berlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
- Friede Springe‐Cardiovascular Prevention Center at Charité, Charité‐Universitätsmedizin, Berlin Institute of Health (BIH)BerlinGermany
| | - Arash Haghikia
- University Hospital St Josef‐Hospital Bochum, Cardiology and RhythmologyRuhr University BochumBochumGermany
- Department of Cardiology, Angiology and Intensive CareDeutsches Herzzentrum der Charité (DHZC), Campus Benjamin FranklinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), Partner site BerlinBerlinGermany
- Berlin Institute of Health at Charité – Universitätsmedizin BerlinBerlinGermany
- Friede Springe‐Cardiovascular Prevention Center at Charité, Charité‐Universitätsmedizin, Berlin Institute of Health (BIH)BerlinGermany
| |
Collapse
|
21
|
Trukovich JJ. From reactions to reflection: A recursive framework for the evolution of cognition and complexity. Biosystems 2025; 250:105408. [PMID: 39892697 DOI: 10.1016/j.biosystems.2025.105408] [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: 10/22/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
This paper presents a comprehensive framework that traces the evolution of consciousness through a continuum of recursive processes spanning reaction, temporogenesis, symbiogenesis, and cognogenesis. By integrating biological cooperation, temporal structuring, and self-referential processing, our model provides a novel perspective on how complexity emerges and scales across evolutionary time. Reaction is established as the foundational mechanism that enables adaptive responses to environmental stimuli, which, through recursive refinement, transitions into temporogenesis-the synchronization of internal processes with external temporal rhythms. Symbiogenesis further enhances this process by fostering cooperative interactions at multiple biological levels, facilitating the emergence of higher-order cognitive functions. Cognogenesis represents the culmination of these recursive processes, where self-awareness and intentionality arise through iterative feedback loops. Our framework offers a biologically grounded pathway to addressing the "hard problem" of consciousness by proposing that subjective experience emerges as a result of progressively complex recursive interactions rather than as a static or isolated phenomenon. In comparing our approach with established theories such as Integrated Information Theory, Global Workspace Theory, and enactive cognition, we highlight its unique contributions in situating consciousness within a broader evolutionary and biological context. This work aims to provide a foundational model that bridges the gap between reaction and reflection, offering empirical avenues for further exploration in neuroscience, evolutionary biology, and artificial intelligence.
Collapse
|
22
|
Dai S, Long J, Han W, Zhang L, Chen B. Alleviative effect of probiotics and prebiotics on dry eye in type 2 diabetic mice through the gut-eye axis. Ocul Surf 2025; 36:244-260. [PMID: 39922458 DOI: 10.1016/j.jtos.2025.02.004] [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: 02/05/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
Diabetes Mellitus (DM) is a metabolic disease that manifests as a state of "chronic low-grade inflammation". Patients with DM have a disorder of intestinal flora. There is a discernible correlation between this disorder of intestinal flora and the onset and progression of eye diseases, which offers novel insights into treating eye diseases through the modulation of intestinal flora. Here, we demonstrated that a high-fat diet and streptozotocin injection-induced intestinal microbiota dysbiosis can lead to dry eye-like manifestations in T2DM mice. Probiotic and prebiotic treatments not only alleviated intestinal inflammation and barrier disruption, but also mitigated damage to the lacrimal barrier and suppressed immune cell infiltration and inflammatory responses. Additional mechanism investigation found that probiotics and prebiotics inhibited the TLR4/NF-κB signaling pathway and its downstream pro-inflammatory products both in the lacrimal gland and colon. 16S RNA sequencing identified a reduction in the bacterial genera Akkermansia and Lactobacillus in the fecal samples of DM mice. By contrast, treatment with probiotics and prebiotics led to a reshaping of the intestinal microbial community and a reduction in bile acid metabolites, such as taurocholic acid and deoxycholic acid. Our current study demonstrates that probiotic and prebiotic treatments can ameliorate dry eye-like symptoms and associated pathological changes in T2DM mice. Moreover, we proved that a high-fat diet and STZ-induced microbiota dysbiosis were involved in diabetic dry eye through the gut-eye axis.
Collapse
Affiliation(s)
- Shirui Dai
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| | - Jianfeng Long
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| | - Wentao Han
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| | - Liwei Zhang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| | - Baihua Chen
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan, 410011, China.
| |
Collapse
|
23
|
Baker JT, Deng Z, Gormley AR, Kim SW. Impacts of non-starch polysaccharide sources with enzymes influencing intestinal mucosa-associated microbiota and mucosal immunity of nursery pigs on growth and carcass traits at market weight. J Anim Sci Biotechnol 2025; 16:47. [PMID: 40165296 PMCID: PMC11959798 DOI: 10.1186/s40104-025-01170-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 01/20/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND This study investigated the effects of different non-starch polysaccharide (NSP) sources with NSP degrading enzymes (NSPases) and the influence on the mucosa-associated microbiota and intestinal immunity of nursery pigs, on growth performance and carcass traits at market weight. METHODS One hundred and sixty newly weaned pigs at 7.0 ± 0.3 kg body weight (BW) were allotted in a 2 × 2 factorial with NSP sources and NSPases serving as factors. The 4 dietary treatments were: DDGS, corn distillers' dried grains with solubles as source of NSP; DDGS + NSPases (DDGS +), DDGS with xylanase at 0.01%, 3,000 U/kg of feed and β-mannanase at 0.05%, 400 U/kg of feed; SHWB, soybean hulls and wheat bran replacing corn DDGS as the source of NSP; SHWB with NSPases (SHWB +), SHWB with xylanase at 0.01%, 3,000 U/kg of feed and β-mannanase at 0.05%, 400 U/kg of feed. Pigs were fed for 37 d and housed in groups of 4 pigs per pen. At d 37, the median body weight pig in each pen was euthanized for sampling to analyze intestinal health parameters. Remaining pigs were fed a common diet for subsequent phases to evaluate the carryover effect on growth and carcass traits. RESULTS The SHWB decreased (P < 0.05) the relative abundance of Helicobacter, tended to increase (P = 0.074) the relative abundance of Lactobacillus, increased (P < 0.05) immunoglobulin G (IgG) in the jejunal mucosa, tended to increase (P = 0.096) the villus height (VH) in the jejunum, and tended to improve ADG (P = 0.099) and feed efficiency (P = 0.068) during phase 1 compared to DDGS treatment. Supplementation of NSPases increased (P < 0.05) Shannon index of diversity, increased the relative abundance of Streptococcus and Acinetobacter, and tended to increase (P = 0.082) dry matter digestibility. The BW of pigs fed SHWB was more uniform (P < 0.05) at the end of the 120 d study. Additionally, hot carcass weight of pigs fed SHWB tended to be more uniform (P = 0.089) than DDGS treatment. CONCLUSION Soybean hulls and wheat bran replacing DDGS in nursery diets improved uniformity of pigs at market weight, which might be attributed to beneficial modulation of the mucosa-associated microbiota and enhanced intestinal morphology during the nursery phase. Supplementation of NSPases had beneficial effects on the intestinal mucosa-associated microbiota, digestibility, and intestinal immunity in SHWB treatment, whereas no carryover effects were overserved at market weight.
Collapse
Affiliation(s)
- Jonathan T Baker
- Department of Animal Science, North Carolina State University, 116 Polk Hall, Campus Box 7621, Raleigh, NC, 27695, USA
| | - Zixiao Deng
- Department of Animal Science, North Carolina State University, 116 Polk Hall, Campus Box 7621, Raleigh, NC, 27695, USA
| | - Alexa R Gormley
- Department of Animal Science, North Carolina State University, 116 Polk Hall, Campus Box 7621, Raleigh, NC, 27695, USA
| | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, 116 Polk Hall, Campus Box 7621, Raleigh, NC, 27695, USA.
| |
Collapse
|
24
|
Marin J, Bertoye PA, Birgy A, Dziri S, Lescat M. Validation of rectal swabbing for total and aerobic gut microbiota study. Microbiol Spectr 2025; 13:e0182324. [PMID: 39969225 PMCID: PMC11960100 DOI: 10.1128/spectrum.01823-24] [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: 07/24/2024] [Accepted: 01/17/2025] [Indexed: 02/20/2025] Open
Abstract
In microbiota research, whole stool sampling is the conventional approach but can be problematic or infeasible for certain patients. This study aims to validate the use of rectal swabbing as an alternative method for microbiota analysis and determine optimal storage conditions suitable for various clinical settings, including intensive care units. We evaluated different sampling techniques and storage temperatures. Our findings indicated that rectal swabs yield microbiota diversity comparable to whole stool samples. Notably, storage conditions significantly impacted microbiota profiles, with increased E. coli and Enterococcus sp. quantifications observed at room temperature (RT). Consequently, we recommend immediate refrigeration of rectal swabs to reliably assess aerobic and total microbiota, particularly for patients requiring urgent care, such as antibiotic treatment. IMPORTANCE We developed a pragmatic approach to study total and aerobic gut microbiota, applicable in numerous clinical units, such as intensive care or emergency units, where whole stool sampling is often impractical. This approach employs ESwab devices, which are already commonly used in hospitals.
Collapse
Affiliation(s)
- Julie Marin
- Université Sorbonne Paris Nord and Université Paris Cité, Inserm, IAME, Bobigny, France
| | - Paul Albin Bertoye
- Université Sorbonne Paris Nord and Université Paris Cité, Inserm, IAME, Bobigny, France
- Service Microbiologie, AP-HP, Hôpital Avicenne, Bobigny, France
| | - Andre Birgy
- Université Sorbonne Paris Nord and Université Paris Cité, Inserm, IAME, Bobigny, France
- Service Microbiologie, AP-HP, Hôpital Robert-Debré, Paris, France
| | - Samira Dziri
- Service Microbiologie, AP-HP, Hôpital Avicenne, Bobigny, France
| | - Mathilde Lescat
- Université Sorbonne Paris Nord and Université Paris Cité, Inserm, IAME, Bobigny, France
- Service Microbiologie, AP-HP, Hôpital Avicenne, Bobigny, France
- Université Paris Cité, Inserm, Institut Cochin, Paris, France
| |
Collapse
|
25
|
Abdi R, Datta S, Zawar A, Kafle P. Evaluation of extended-spectrum β-lactamase producing bacteria in feces of shelter dogs as a biomarker for altered gut microbial taxa and functional profiles. Front Microbiol 2025; 16:1556442. [PMID: 40196031 PMCID: PMC11975251 DOI: 10.3389/fmicb.2025.1556442] [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: 01/06/2025] [Accepted: 02/19/2025] [Indexed: 04/09/2025] Open
Abstract
Background The USA is home to 83-88 million dogs, with 3-7 million living in shelters. Shelter dogs move through the supply chain from their geographical origin to adoptive homes, with possible exposure to pathogens and shift in their gut microbiota. However, research in this area is limited. This study examined the effects of intestinal colonization by ESBL bacteria on gut taxa abundance, diversity, and functions in 52 shelter dogs of various ages, sexes, and fertility statuses. Methodology We isolated fecal DNA, sequenced their 16S, processed the sequences using DADA2, identified taxa profiles in each dog by Phyloseq, and analyzed Chao1, Shannon, and Simpson alpha diversity by ggplot2 and Wilcoxon test. We analyzed beta diversity using Bray-Curtis dissimilarity matrix from the vegan package. Differential abundance of taxa, gut microbiome functions, and differential abundance of microbiome functions were analyzed using DESeq2, PICRUSt2, and ALDEx2, respectively, with Wilcoxon rank and Kruskal-Wallis tests for comparisons between dog groups. Results Firmicutes (69.3%), Bacteroidota (13.5%), Actinobacteriota (6.77%), Proteobacteria (5.54%), and Fusobacteriota (4.75%) were the major phyla in the gut of shelter dogs. ESBL bacteria colonized dogs had reduced gut microbiota alpha diversity than non-colonized dogs. The abundance levels of the following phyla (Proteobacteria, Deferribacterota, Bacteroidota, Fusobacteriota, and Spirochaetota), class (Gammaproteobacteria, Bacteroidia, Deferribacteres, Brachyspirae, and Fusobacteria), and families (Enterobacteriaceae, Peptostreptococcaceae, Lactobacillaceae, Lachnospiraceae, Prevotellaceae, and Peptostreptococcaceae) were significantly (p < 0.05) varied between the two dog groups. Further stratified analysis by age, sex, and spaying/neutering status influenced the abundance of taxa in ESBL bacteria colonized dogs, indicating these covariates act as effect modifiers. Most gut metabolic and biosynthetic pathways were downregulated in ESBL bacteria colonized dogs compared to non-colonized dogs. However, alpha-linolenic acid metabolism and shigellosis, fluorobenzoate degradation, allantoin degradation, toluene degradation, glycol degradation, fatty acid and beta-oxidation, and glyoxylate metabolism bypass pathways were increased in dogs colonized by ESBL bacteria. Conclusion Colonization by ESBL bacteria marks altered gut microbiota. Dog's demography and fertility status modify the alterations, indicating host factors and ESBL bacteria interplay to shape gut microbiota. ESBL bacteria or other factors reprogram gut microbiome functions through down and upregulating multiple metabolic and biosynthesis pathways to promote ESBL bacteria colonization.
Collapse
Affiliation(s)
- Reta Abdi
- Biomedical Sciences College of Veterinary Medicine, Long Island University, Brookville, NY, United States
| | - Srinka Datta
- GeneSpectrum Life Sciences LLP, Pune, Maharashtra, India
| | | | - Pratap Kafle
- Shreiber School of Veterinary Medicine, Rowan University, Mullica Hill, NJ, United States
| |
Collapse
|
26
|
Beyoğlu D, Idle JR. The Microbiome and Metabolic Dysfunction-Associated Steatotic Liver Disease. Int J Mol Sci 2025; 26:2882. [PMID: 40243472 PMCID: PMC11988851 DOI: 10.3390/ijms26072882] [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: 02/17/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a condition wherein excessive fat accumulates in the liver, leading to inflammation and potential liver damage. In this narrative review, we evaluate the tissue microbiota, how they arise and their constituent microbes, and the role of the intestinal and hepatic microbiota in MASLD. The history of bacteriophages (phages) and their occurrence in the microbiota, their part in the potential causation of MASLD, and conversely, "phage therapy" for antibiotic resistance, obesity, and MASLD, are all described. The microbiota metabolism of bile acids and dietary tryptophan and histidine is defined, together with the impacts of their individual metabolites on MASLD pathogenesis. Both periodontitis and intestinal microbiota dysbiosis may cause MASLD, and how individual microorganisms and their metabolites are involved in these processes is discussed. Novel treatment opportunities for MASLD involving the microbiota exist and include fecal microbiota transplantation, probiotics, prebiotics, synbiotics, tryptophan dietary supplements, intermittent fasting, and phages or their holins and endolysins. Although FDA is yet to approve phage therapy in clinical use, there are multiple FDA-approved clinical trials, and this may represent a new horizon for the future treatment of MASLD.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
| | - Jeffrey R. Idle
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
- Department of Biomedical Research, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
27
|
Díez-Madueño K, Montero I, Fernández-Gosende M, Martínez-Álvarez N, Hidalgo-Cantabrana C, de la Cueva Dobao P, Coto-Segura P. Compositional and Functional Profile of Gut Microbiota in a Cohort of Adult Spanish Patients with Atopic Dermatitis Using Metagenomics: A Cross-Sectional Study. Dermatitis 2025. [PMID: 40111891 DOI: 10.1089/derm.2024.0536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Background: The role of gut dysbiosis in the pathophysiology of atopic dermatitis (AD) through immune system (IS) imbalance is a novel line of investigation currently under discussion. This study aimed to characterize compare the composition and functional profile of the gut microbiota (GM) between adults with AD and healthy individuals. Methods: Observational cross-sectional study, where fecal samples from 70 adults (38 patients and 32 controls) were analyzed using metagenomics and bioinformatics. Results: Differences between the GM of patients with AD and healthy individuals were demonstrated. Reduced microbial diversity was found in subjects with AD. Bacterial species with lower abundance primarily belonged to the families Ruminococcaceae, Akkermansiaceae, and Methanobacteriaceae. Several microbial metabolic pathways were found to be decreased in patients with AD, including amino acid biosynthesis, vitamin biosynthesis, fatty acids and lipids biosynthesis, and energy metabolism. Conclusion: Adults with AD exhibited a distinct GM compared to healthy individuals. Changes were demonstrated both compositionally and functionally. Further investigation is mandatory to elucidate the potential link and causal relationship between gut dysbiosis and AD, which may be crucial for a deeper understanding of the disease's pathophysiology and the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Kevin Díez-Madueño
- From the Dermatology Department, Hospital Universitario Infanta Leonor, Madrid, Spain
- Complutense University of Madrid, Madrid, Spain
| | | | | | | | | | - Pablo de la Cueva Dobao
- From the Dermatology Department, Hospital Universitario Infanta Leonor, Madrid, Spain
- Complutense University of Madrid, Madrid, Spain
| | - Pablo Coto-Segura
- Dermatology Department, Hospital Vital Álvarez Buylla, Mieres, Spain
| |
Collapse
|
28
|
Ruiz-Moreno AJ, Del Castillo-Izquierdo Á, Tamargo-Rubio I, Fu J. MicrobeRX: a tool for enzymatic-reaction-based metabolite prediction in the gut microbiome. MICROBIOME 2025; 13:78. [PMID: 40108657 PMCID: PMC11921629 DOI: 10.1186/s40168-025-02070-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 02/23/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND The gut microbiome functions as a metabolic organ, producing numerous enzymes that influence host health; however, their substrates and metabolites remain largely unknown. RESULTS We present MicrobeRX, an enzyme-based metabolite prediction tool that employs 5487 human reactions and 4030 unique microbial reactions from 6286 genome-scale models, as well as 3650 drug metabolic reactions from the DrugBank database (v.5.1.12). MicrobeRX includes additional analysis modules for metabolite visualization and enzymatic and taxonomic analyses. When we applied MicrobeRX to 1083 orally administered drugs that have been approved in at least one jurisdiction at some point in time (DrugBank), it predicted metabolites with physicochemical properties and structures similar to metabolites found in biosamples (from MiMeDB). It also outperformed another existing metabolite prediction tool (BioTransformer 3.0) in terms of predictive potential, molecular diversity, reduction of redundant predictions, and enzyme annotation. CONCLUSIONS Our analysis revealed both unique and overlapping metabolic capabilities in human and microbial metabolism and chemo- and taxa-specific microbial biotransformations. MicrobeRX bridges the genomic and chemical spaces of the gut microbiome, making it a valuable tool for unlocking the chemical potential of the gut microbiome in human health, the food and pharmaceutical industries, and environmental safety. Video Abstract.
Collapse
Affiliation(s)
- Angel J Ruiz-Moreno
- Department of Genetics, University Medical Center Groningen, Groningen, 9713GZ, The Netherlands.
- Department of Pediatrics, University Medical Center Groningen, Groningen, 9713GZ, The Netherlands.
| | - Ángela Del Castillo-Izquierdo
- Department of Genetics, University Medical Center Groningen, Groningen, 9713GZ, The Netherlands
- Department of Medical Microbiology, University Medical Center Groningen, Groningen, 9713GZ, The Netherlands
| | - Isabel Tamargo-Rubio
- Department of Genetics, University Medical Center Groningen, Groningen, 9713GZ, The Netherlands
| | - Jingyuan Fu
- Department of Genetics, University Medical Center Groningen, Groningen, 9713GZ, The Netherlands.
- Department of Pediatrics, University Medical Center Groningen, Groningen, 9713GZ, The Netherlands.
| |
Collapse
|
29
|
Zhu D, Li S, Xu Z, Kulyar MF, Bai X, Wang Y, Wang B, Khateeb E, Deng D, Wang L, Chen Y, Guo A, Shen Y. Comparative analysis of gut microbiota in healthy and diarrheic foals. Microbiol Spectr 2025:e0087124. [PMID: 40105330 DOI: 10.1128/spectrum.00871-24] [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: 04/05/2024] [Accepted: 02/01/2025] [Indexed: 03/20/2025] Open
Abstract
Diarrhea presents a substantial risk of high morbidity and mortality among foals. Although studies have shown connections between gut microbiota and several gastrointestinal diseases, there is still inadequate information on gut microbial alterations in foals during diarrhea. In this study, we conducted 16S rRNA and ITS gene amplicon sequencing to investigate gut bacterial and fungal differences between healthy and diarrheic foals. The results unveiled significant reductions in gut bacterial and fungal diversities among foals experiencing diarrhea, accompanied by notable shifts in the composition of gut microbial communities. A considerable decrease was observed in the relative abundance of 30 bacterial and 34 fungal genera. Moreover, two bacterial and eight fungal genera were utterly undetectable in the gut microbiota of diarrheic foals. Some decreased genera, such as Bifidobacterium and Saccharomyces, were deemed beneficial and recognized as probiotics. The study revealed significant alterations in foals' gut bacterial and fungal communities during diarrhea, which enriched our comprehension of gut microbial dynamics in foals across varying health statuses. These findings offer valuable insights for managing diarrhea through gut microbiota modulation, suggesting that probiotics may be superior to antibiotics in preventing and controlling foal diarrhea.IMPORTANCEThis research advances the understanding of gut bacterial and fungal dynamics in foals, highlighting gut microbiota dysbiosis as a potential contributor to foal diarrhea. Additionally, we observed that many altered bacteria and fungi were downregulated during diarrhea, including some probiotic strains. Consequently, our findings provide evidence that probiotics may offer superior efficacy compared with antibiotics as potential candidates for preventing and treating foal diarrhea.
Collapse
Affiliation(s)
- Di Zhu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Siyu Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Zhixiang Xu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Md F Kulyar
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Xu Bai
- China Horse Industry Association, Beijing, China
| | - Yu Wang
- China Horse Industry Association, Beijing, China
| | - Boya Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Emaan Khateeb
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Dandan Deng
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Lidan Wang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yuji Chen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Aizhen Guo
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yaoqin Shen
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
30
|
Saad MJA, Santos A. The Microbiota and Evolution of Obesity. Endocr Rev 2025; 46:300-316. [PMID: 39673174 PMCID: PMC11894537 DOI: 10.1210/endrev/bnae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/03/2024] [Accepted: 12/12/2024] [Indexed: 12/16/2024]
Abstract
Obesity is a major global concern and is generally attributed to a combination of genetic and environmental factors. Several hypotheses have been proposed to explain the evolutionary origins of obesity epidemic, including thrifty and drifty genotypes, and changes in thermogenesis. Here, we put forward the hypothesis of metaflammation, which proposes that due to intense selection pressures exerted by environmental pathogens, specific genes that help develop a robust defense mechanism against infectious diseases have had evolutionary advantages and that this may contribute to obesity in modern times due to connections between the immune and energy storage systems. Indeed, incorporating the genetic variations of gut microbiota into the complex genetic framework of obesity makes it more polygenic than previously believed. Thus, uncovering the evolutionary origins of obesity requires a multifaceted approach that considers the complexity of human history, the unique genetic makeup of different populations, and the influence of gut microbiome on host genetics.
Collapse
Affiliation(s)
- Mario J A Saad
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, CEP 13083-887 Campinas, SP, Brazil
| | - Andrey Santos
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, CEP 13083-887 Campinas, SP, Brazil
| |
Collapse
|
31
|
Hao S, Sun W, Wei P, Wu H, Lu W, He Y. Supplementation with Rare Earth-Chitosan Chelate Improves Tibia Quality, Disease Resistance Capacity, and Performance in Nursery Pigs. Int J Mol Sci 2025; 26:2409. [PMID: 40141053 PMCID: PMC11942057 DOI: 10.3390/ijms26062409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/16/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
The aim of this study was to investigate the effects on the tibia, liver, and gut, and on performance, when supplementing nursery pigs with different levels of rare earth-chitosan chelate (RECC). A total of 80 piglets, weaned at 7.67 ± 0.09 kg, were randomly assigned to groups RECC0 (RECC, 0 mg/kg diet), RECC200 (RECC, 200 mg/kg diet), RECC400 (RECC, 400 mg/kg diet), and RECC600 (RECC, 600 mg/kg diet), with four replicates in each group and five pigs per replicate during a 28 d experiment. Samples of the left hind tibia, serum, and feces were collected for analysis. The results indicated that, compared to pigs from group RECC0, pigs from group RECC200 presented with the following: a longer trabecular perimeter (p < 0.05), a larger trabecular area (p < 0.01), a higher trabecular number (p < 0.05), a smaller degree of trabecular separation (p < 0.01), and a lower number of osteoclasts (p < 0.01) in the tibia; higher abundances of beneficial fecal bacteria such as g_Prevotellaceae_NK3B31_group, g_UCG_005, g_Rikenellaceae_RC9_gut_group, g_Acetitomaculum, g_Glutamicibacter, g_Frisingicoccus, and g_Alistipes; higher (p < 0.01) serum levels of IgM, IgA, IgG, and IL-10; a lower (p < 0.01) serum concentration of TNF-α; a higher (p < 0.05) average daily gain and feed conversion ratio; and a lower (p < 0.01) incidence of diarrhea. The dietary addition of RECC contributes to improvements in tibia quality, gut health, and performance in nursery pigs.
Collapse
Affiliation(s)
- Shaobin Hao
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (S.H.); (W.S.); (P.W.); (W.L.)
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Wenchen Sun
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (S.H.); (W.S.); (P.W.); (W.L.)
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Panting Wei
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (S.H.); (W.S.); (P.W.); (W.L.)
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Huadong Wu
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Wei Lu
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (S.H.); (W.S.); (P.W.); (W.L.)
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| | - Yuyong He
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Engineering Research Center of Feed Development, Jiangxi Agricultural University, Nanchang 330045, China; (S.H.); (W.S.); (P.W.); (W.L.)
- College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China;
| |
Collapse
|
32
|
Zheng T, Meng C, Lv Z, Wu C, Zhou X, Mao W. The Critical Role of Faecalibacterium prausnitzii in Cardiovascular Diseases. Rev Cardiovasc Med 2025; 26:26740. [PMID: 40160596 PMCID: PMC11951488 DOI: 10.31083/rcm26740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 04/02/2025] Open
Abstract
Due to the continued aging of the global population, cardiovascular diseases (CVDs) remain the main cause of death worldwide, with millions of fatalities from diseases, including stroke and coronary artery disease, reported annually. Thus, novel therapeutic approaches and targets are urgently required for diagnosing and treating CVDs. Recent studies emphasize the vital part of gut microbiota in both CVD prevention and management. Among these, Faecalibacterium prausnitzii (F. prausnitzii) has emerged as a promising probiotic capable of improving intestinal health. Although preliminary investigations demonstrate that F. prausnitzii positively enhances cardiovascular health, research specifically connecting this strain to CVD outcomes remains limited. Based on current research and assessment of possible clinical applications, this paper aimed to investigate the positive effects on cardiovascular health using F. prausnitzii and its metabolites. Targeting gut flora is expected to become a mainstay in CVD treatment as research develops.
Collapse
Affiliation(s)
- Tiantian Zheng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Chenchen Meng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Zhengtian Lv
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
| | - Chenxia Wu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, 310053 Hangzhou, Zhejiang, China
- Department of Cardiology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, 310030 Hangzhou, Zhejiang, China
| | - Xinbin Zhou
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), 310006 Hangzhou, Zhejiang, China
| | - Wei Mao
- Department of Cardiology, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, 310030 Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Integrative Chinese and Western Medicine for Diagnosis and Treatment of Circulatory Diseases, 310030 Hangzhou, Zhejiang, China
| |
Collapse
|
33
|
Lan F, Wang X, Zhou Q, Li X, Jin J, Zhang W, Wen C, Wu G, Li G, Yan Y, Yang N, Sun C. Deciphering the coordinated roles of the host genome, duodenal mucosal genes, and microbiota in regulating complex traits in chickens. MICROBIOME 2025; 13:62. [PMID: 40025569 PMCID: PMC11871680 DOI: 10.1186/s40168-025-02054-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 02/01/2025] [Indexed: 03/04/2025]
Abstract
BACKGROUND The complex interactions between host genetics and the gut microbiome are well documented. However, the specific impacts of gene expression patterns and microbial composition on each other remain to be further explored. RESULTS Here, we investigated this complex interplay in a sizable population of 705 hens, employing integrative analyses to examine the relationships among the host genome, mucosal gene expression, and gut microbiota. Specific microbial taxa, such as the cecal family Christensenellaceae, which showed a heritability of 0.365, were strongly correlated with host genomic variants. We proposed a novel concept of regulatability ( r b 2 ), which was derived from h2, to quantify the cumulative effects of gene expression on the given phenotypes. The duodenal mucosal transcriptome emerged as a potent influencer of duodenal microbial taxa, with much higher r b 2 values (0.17 ± 0.01, mean ± SE) than h2 values (0.02 ± 0.00). A comparative analysis of chickens and humans revealed similar average microbiability values of genes (0.18 vs. 0.20) and significant differences in average r b 2 values of microbes (0.17 vs. 0.04). Besides, cis ( h cis 2 ) and trans heritability ( h trans 2 ) were estimated to assess the effects of genetic variations inside and outside the cis window of the gene on its expression. Higher h trans 2 values than h cis 2 values and a greater prevalence of trans-regulated genes than cis-regulated genes underscored the significant role of loci outside the cis window in shaping gene expression levels. Furthermore, our exploration of the regulatory effects of duodenal mucosal genes and the microbiota on 18 complex traits enhanced our understanding of the regulatory mechanisms, in which the CHST14 gene and its regulatory relationships with Lactobacillus salivarius jointly facilitated the deposition of abdominal fat by modulating the concentration of bile salt hydrolase, and further triglycerides, total cholesterol, and free fatty acids absorption and metabolism. CONCLUSIONS Our findings highlighted a novel concept of r b 2 to quantify the phenotypic variance attributed to gene expression and emphasize the superior role of intestinal mucosal gene expressions over host genomic variations in elucidating host‒microbe interactions for complex traits. This understanding could assist in devising strategies to modulate host-microbe interactions, ultimately improving economic traits in chickens.
Collapse
Affiliation(s)
- Fangren Lan
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiqiong Wang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Qianqian Zhou
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiaochang Li
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jiaming Jin
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Wenxin Zhang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Chaoliang Wen
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Guiqin Wu
- Beijing Engineering Research Centre of Layer, Beijing, 101206, China
| | - Guangqi Li
- Beijing Engineering Research Centre of Layer, Beijing, 101206, China
| | - Yiyuan Yan
- Beijing Engineering Research Centre of Layer, Beijing, 101206, China
| | - Ning Yang
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China.
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China.
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| | - Congjiao Sun
- State Key Laboratory of Animal Biotech Breeding and Frontier Science Center of Molecular Design Breeding, China Agricultural University, Beijing, 100193, China.
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, China Agricultural University, Beijing, 100193, China.
- Department of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
34
|
Marin J, Ghalayini M, Kaoudji Y, Dziri S, Zylberfajn C, Blaise L, Hoogvorst A, Charpentier S, Chaillou V, Beauchamp S, Donneger S, Barget N, Touvier M, Nahon P, Amathieu R, Lescat M. Comprehensive toolkit integrating lifestyle and clinical questionnaires with gut microbiota profiling via rectal swabs: application in intensive care cirrhotic patients. J Med Microbiol 2025; 74:001964. [PMID: 40047237 PMCID: PMC11936375 DOI: 10.1099/jmm.0.001964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/19/2024] [Indexed: 03/27/2025] Open
Abstract
Introduction. The study of gut microbiota is now an essential dimension in many clinical studies. For instance, microbiota diversity investigation can help us to better manage cirrhotic patients by the identification of markers of severity and the identification of possible sources of pathogens.Hypothesis/Gap Statement. Conducting clinical research on gut microbiota for fragile patients in intensive care units, such as cirrhotic patients, poses significant challenges.Aim. In this study, we developed a comprehensive toolkit for investigating gut microbiota in fragile patients using rectal swabbing combined with straightforward lifestyle and clinical questionnaires.Methodology . We applied this prospective approach to 49 well-phenotyped cirrhotic patients as a function of their compensation status (compensated patients with outpatients' recruitment vs decompensated patients in intensive care units).Results . Our results, consistent with the literature, showed that liver function impairment is associated with lower microbiota diversity. Additionally, we monitored aerobic microbiota in decompensated cirrhotic patients, observing the invasion of extended spectrum beta-lactamase (ESBL)-producing Escherichia coli in the gut's aerobic microbiota prior to severe infection caused by these pathogens.Conclusion. We propose this pragmatic methodology for larger cohort studies, aiming to enhance the monitoring of immunocompromised patients by using microbiota analysis as a predictive tool for the severity of associated pathologies and the identification of agents responsible for severe infections.
Collapse
Affiliation(s)
- Julie Marin
- Université Sorbonne Paris Nord and Université Paris Cité, Inserm, IAME, F-93000 Bobigny, France
| | - Mohamed Ghalayini
- Université Sorbonne Paris Nord and Centre hospitalier de Gonesse, Gonesse, France
| | - Younes Kaoudji
- Université Sorbonne Paris Nord and Université Paris Cité, Inserm, IAME, F-93000 Bobigny, France
| | - Samira Dziri
- Service Microbiologie, AP-HP, Hôpitaux Universitaires de Paris Seine Saint Denis France, Paris, France
| | - Cecile Zylberfajn
- Service de Réanimation, AP-HP, Hôpitaux Universitaires de Paris Seine Saint Denis, Paris, France
| | - Lorraine Blaise
- Université Sorbonne Paris Nord and Service d’Hépatologie, AP-HP, Hôpitaux Universitaires de Paris Seine Saint Denis, Paris, France
| | - Astrid Hoogvorst
- Service de Réanimation, AP-HP, Hôpitaux Universitaires de Paris Seine Saint Denis, Paris, France
| | - Stephane Charpentier
- Service de Réanimation, AP-HP, Hôpitaux Universitaires de Paris Seine Saint Denis, Paris, France
| | - Virginie Chaillou
- Service Microbiologie, AP-HP, Hôpitaux Universitaires de Paris Seine Saint Denis France, Paris, France
| | - Sylvie Beauchamp
- Service Microbiologie, AP-HP, Hôpitaux Universitaires de Paris Seine Saint Denis France, Paris, France
| | - Séverine Donneger
- Centre de Ressources biologiques, AP-HP, Hôpitaux Universitaires de Paris Seine Saint Denis, Paris, France
| | - Nathalie Barget
- Centre de Ressources biologiques, AP-HP, Hôpitaux Universitaires de Paris Seine Saint Denis, Paris, France
| | - Mathilde Touvier
- Université Sorbonne Paris Nord and Université Paris Cité, Inserm, EREN, F-93000 Bobigny, France
| | - Pierre Nahon
- Université Sorbonne Paris Nord and Service d’Hépatologie, AP-HP, Hôpitaux Universitaires de Paris Seine Saint Denis, Paris, France
| | - Roland Amathieu
- Université Sorbonne Paris Nord and Centre hospitalier de Gonesse, Gonesse, France
| | - Mathilde Lescat
- Université Sorbonne Paris Nord and Université Paris Cité, Inserm, IAME, F-93000 Bobigny, France
- Université Paris Cité, Inserm, Institut Cochin, F-75014 Paris, France
- Bacteriology Unit, Microbiology and Infectious Diseases Department, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France
- CNR-LE Charbon, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France and Aix Marseille Université, INSERM, SSA, MCT, Marseille, France
| |
Collapse
|
35
|
Kim G, Kim S, Lee W, Shin H. The impact of coffee on gut microbial structure based on in vitro fecal incubation system. Food Sci Biotechnol 2025; 34:971-979. [PMID: 39974865 PMCID: PMC11832990 DOI: 10.1007/s10068-024-01717-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 02/21/2025] Open
Abstract
Coffee is globally popular beverage, renowned for its taste and stimulating properties. This study aims to explore the impact of two different types of coffee, depending on extraction methods, on the gut microbiota. Fecal samples from healthy donors (n = 20) were cultured with or without coffee using in vitro fecal incubation. Both coffee-treated groups exhibited lower microbial diversity and greater structural differences in their communities compared to the control. Notably, the Bifidobacterium genus was overrepresented in the instant coffee (IC)-treated groups, whereas the Blautia genus was underrepresented in both coffee-treated groups. Additionally, genes for TCA cycle and vitamin B6 metabolism were more prevalent in coffee-treated groups than in the control. However, the precursor pathways leading to the TCA cycle differed between the DC- and IC-treated groups, reflecting the distinct chemical compositions of each coffee type. These findings demonstrate that extraction method of coffee significantly affects its impacts on gut microbial structure. Supplementary Information The online version contains supplementary material available at 10.1007/s10068-024-01717-7.
Collapse
Affiliation(s)
- Gyungcheon Kim
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, College of Life Science, Sejong University, Seoul, 05006 Republic of Korea
| | - Seongok Kim
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, College of Life Science, Sejong University, Seoul, 05006 Republic of Korea
| | - WonJune Lee
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, College of Life Science, Sejong University, Seoul, 05006 Republic of Korea
| | - Hakdong Shin
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, College of Life Science, Sejong University, Seoul, 05006 Republic of Korea
| |
Collapse
|
36
|
Sun Y, Yao J, Gao R, Hao J, Liu Y, Liu S. Interactions of non-starch polysaccharides with the gut microbiota and the effect of non-starch polysaccharides with different structures on the metabolism of the gut microbiota: A review. Int J Biol Macromol 2025; 296:139664. [PMID: 39798752 DOI: 10.1016/j.ijbiomac.2025.139664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Humans consume large amounts of non-starch polysaccharides(NPs) daily. Some NPs, not absorbed by the body, proceed to the intestines. An increasing number of studies reveal a close relationship between NPs and gut microbiota(GM) that impact the human body. This review not only describes in detail the structures of several common NPs and their effects on GM, but also elucidates the degradation mechanisms of NPs in the intestine. The purpose of this review is to elucidate how NPs interact with GM in the intestine, which can provide valuable information for further studies of NPs.
Collapse
Affiliation(s)
- Yujiao Sun
- Natural Food Macromolecule Research Center, School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, PR China; Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an 710021, PR China.
| | - Jiaxuan Yao
- Natural Food Macromolecule Research Center, School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, PR China
| | - Running Gao
- Natural Food Macromolecule Research Center, School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, PR China
| | - Junyu Hao
- Natural Food Macromolecule Research Center, School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, PR China
| | - Yang Liu
- Shaanxi Academy of Traditional Chinese Medicine, Xi'an 710003, China
| | - Shuai Liu
- Shaanxi Academy of Traditional Chinese Medicine, Xi'an 710003, China.
| |
Collapse
|
37
|
Manfredi JN, Gupta SK, Vyavahare S, Deak F, Lu X, Buddha L, Wankhade U, Lohakare J, Isales C, Fulzele S. Gut microbiota dysbiosis in Alzheimer's disease (AD): Insights from human clinical studies and the mouse AD models. Physiol Behav 2025; 290:114778. [PMID: 39672482 DOI: 10.1016/j.physbeh.2024.114778] [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: 08/20/2024] [Revised: 11/19/2024] [Accepted: 12/07/2024] [Indexed: 12/15/2024]
Abstract
Alzheimer's Disease (AD) is a debilitating neurocognitive disorder with an unclear underlying mechanism. Recent studies have implicated gut microbiota dysbiosis with the onset and progression of AD. The connection between gut microbiota and AD can significantly affect the prevention and treatment of AD patients. This systematic review summarizes primary outcomes of human and mouse AD models concerning gut microbiota alterations. A systematic literature search in February through March 2023 was conducted on PubMed, Embase, and Web of Science. We identified 711 as potential manuscripts of which 672 were excluded because of irrelevance to the identified search criteria. Primary outcomes include microbiota compositions of control and AD models in humans and mice. In total, 39 studies were included (19 mouse and 20 human studies), published between 2017 and 2023. We included studies involving well-established mice models of AD (5xFAD, 3xTg-AD, APP/PS1, Tg2576, and APPPS2) which harbor mutations and genes that drive the formation of Aß plaques. All human studies were included on those with AD or mild cognitive impairment. Among alterations in gut microbiota, most studies found a decreased abundance of the phyla Firmicutes and Bifidobacteria, a genus of the phylum Actinomycetota. An increased abundance of the phyla Bacteroidetes and Proteobacteria were identified in animal and human studies. Studies indicated that gut microbiota alter the pathogenesis of AD through its impact on neuroinflammation and permeability of the gastrointestinal tract. The ensuing increase in blood-brain barrier permeability may accelerate Aβ penetrance and formation of neuritic plaques that align with the amyloid hypothesis of AD pathogenesis. Further studies should assess the relationship between gut microbiota and AD progression and therapy preserving beneficial gut microbiota.
Collapse
Affiliation(s)
- John N Manfredi
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sonu Kumar Gupta
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sagar Vyavahare
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Ferenc Deak
- Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA
| | - Xinyun Lu
- Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA
| | - Lasya Buddha
- Arkansas Children's Nutrition Center, Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Umesh Wankhade
- Arkansas Children's Nutrition Center, Department of Pediatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Jayant Lohakare
- College of Agriculture, Food, and Natural Resources, Prairie View A&M University, Prairie View, TX 77446, USA
| | - Carlos Isales
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA; Centre for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Sadanand Fulzele
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, USA; Deptment of Neuroscience & Regenerative Medicine, Augusta, GA 30912, USA; College of Agriculture, Food, and Natural Resources, Prairie View A&M University, Prairie View, TX 77446, USA; Centre for Healthy Aging, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, GA, USA; Department of Orthopedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA.
| |
Collapse
|
38
|
Patki A, Kunjimoideen K, Sawankar S, Tyagi R, Hegde V, Budi J. Expert Opinion on the Use of Probiotics in Recurrent Pregnancy Loss. Cureus 2025; 17:e81056. [PMID: 40271290 PMCID: PMC12015142 DOI: 10.7759/cureus.81056] [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] [Accepted: 03/15/2025] [Indexed: 04/25/2025] Open
Abstract
Recurrent pregnancy loss (RPL) involves multiple consecutive miscarriages in early pregnancy, affecting a significant number of Indian women and placing substantial physical and emotional stress on expecting couples. This expert consensus aims to highlight probiotics as a promising option for enhancing fertility and supporting successful pregnancy outcomes, offering hope to individuals and couples affected by RPL. A group of fourteen experts with diverse expertise in gynecology, obstetrics, and fertility from across India gathered between June 29 and June 30, 2024. According to the experts, advanced maternal age emerges as an independent risk factor for miscarriage, with increased risks among older Indian women. The major contributors to RPL include thyroid disease and polycystic ovarian disease. Experts emphasize that the vaginal microbiome dysbiosis, characterized by the reduced dominance of Lactobacilli, is associated with adverse pregnancy outcome, such as preterm birth, early pregnancy loss, and increased events of RPL. Oral probiotic supplementation, particularly strains like L. acidophilus and L. rhamnosus, may improve embryo implantation, reduce miscarriage risk, and support pregnancy maintenance. A healthy lifestyle choice and minimal use of antibiotics are important in creating a positive reproductive outcome. The present expert opinion supports the potential benefits of probiotics, particularly Lactobacillus species, in managing RPL and improving reproductive outcomes. By promoting a balanced microbiota, reducing inflammation, and modulating immune responses, probiotics may play a critical role in enhancing reproductive success.
Collapse
Affiliation(s)
- Ameet Patki
- Obstetrics and Gynecology, Indian Society for Assisted Reproduction (ISAR), Mumbai, IND
| | - K Kunjimoideen
- Obstetrics and Gynecology, Asian Reproductive Medicine Centre, Kochi, IND
| | - Sheetal Sawankar
- Obstetrics and Gynecology, Avisa IVF and Fertility Center, Mumbai, IND
| | - Rajul Tyagi
- Obstetrics and Gynecology, Javitri Hospital and Test Tube Baby Centre, Lucknow, IND
| | - Vandana Hegde
- Obstetrics and Gynecology, Hegde Fertility, Hyderabad, IND
| | - Jyoti Budi
- Obstetrics and Gynecology, Ferty9 Fertility Center, Hyderabad, IND
| |
Collapse
|
39
|
Chen Y, Luo Q, Wu H, Wang Q, Zhang Y. Amomum longiligulare polysaccharide 1 supplementation promotes the proliferation of jejunal epithelial cells in piglets by regulating jejunal metabolites. Int J Biol Macromol 2025; 306:141366. [PMID: 39993675 DOI: 10.1016/j.ijbiomac.2025.141366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/08/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Although natural polysaccharides often have growth-promoting effects on animals, little is known about how polysaccharides act when they are administered as feed additives. This work shows that Amomum longiligulare polysaccharide 1 (ALP1) improves the growth performance of piglets by promoting the proliferation of jejunal epithelial cells. ALP1 improves the growth performance of piglets, increasing the average daily gain by 32.71 % and reducing the feed-to-gain (F/G) ratio by 21.93 %. The gut microbiota is an important regulatory target of polysaccharides. The results of jejunal microbiota transplantation trials indicate that the jejunal microbiota from ALP1-fed piglets exhibits better growth performance and that the F/G ratio is reduced by 12.72 %. Furthermore, 16S rDNA sequencing and nontargeted metabolomic analyses reveal that ALP1 supplementation can increase the abundance of Lactobacillus in the jejuna of piglets, resulting in a high abundance of 11Z-eicosenoic acid (EA). In addition, EA increases the villus height-to-crypt depth (VH/CD) ratio in the jejunum by 27.41 %, potentially increasing epithelial cell proliferation. These results suggest that oral ALP1 supplementation promotes growth by modulating the composition of the jejunal microbiota and its associated metabolites.
Collapse
Affiliation(s)
- Yun Chen
- Sanya Institute of Breeding and Multiplication, School of Tropical Agriculture and Forestry, Hainan University, Sanya 572025, PR China.
| | - Qiyuan Luo
- Sanya Institute of Breeding and Multiplication, School of Tropical Agriculture and Forestry, Hainan University, Sanya 572025, PR China
| | - Haowen Wu
- Sanya Institute of Breeding and Multiplication, School of Tropical Agriculture and Forestry, Hainan University, Sanya 572025, PR China
| | - Quanjiang Wang
- Sanya Institute of Breeding and Multiplication, School of Tropical Agriculture and Forestry, Hainan University, Sanya 572025, PR China
| | - Yue Zhang
- Sanya Institute of Breeding and Multiplication, School of Tropical Agriculture and Forestry, Hainan University, Sanya 572025, PR China
| |
Collapse
|
40
|
Atabieke F, Aierken A, Aierken M, Rehaman M, Zhang QQ, Li J, Xia Y, Aizezi Y, Dilixiati D, Gao HL, Zhang ZQ. Investigating casual association among gut microbiome and esophageal cancer: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41563. [PMID: 39993127 PMCID: PMC11856886 DOI: 10.1097/md.0000000000041563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
The gut microbiota has been strongly linked to gastrointestinal cancer, but the relationship between gut microbiota and esophageal cancer (EC) is still not fully understood. We conducted a 2-sample Mendelian randomization (MR) study to unveil the potential impact of intestinal microorganisms on EC in East Asian populations. In order to delve deeper into the potential causal relationship between gut microbiota and EC, we conducted a 2-sample MR analysis, utilizing 211 single nucleotide polymorphisms associated with gut microbiota, sourced from the largest genome-wide association study on gut microbiota, for our analysis. To estimate the causal relationship, we employed the inverse variance weighting method. In addition, to assess the potential influence of pleiotropy, we used MR-Egger regression in our analysis. Among the 10 specific bacterial taxa identified using the inverse variance weighting as being associated with the risk of EC, we observed a positive association between family Bacteroidaceae (P = .04), genus Bacteroides (P = .04), genus Bilophila (P = .02), genus Candidatus Soleaferrea (P = .02) and the EC, while family Victivallaceae (P = .03), genus Eubacterium coprostanoligenes (P = .01), genus Catenibacterium (P = .01), genus Coprococcus2 (P = .01), unknowngenus.id.959 (P = .02) and unknowngenus.id.1868 (P = .01) may be associated with a reduced risk of EC. Our MR analysis indicate a probable association between gut microbiota and the development and advancement of EC. These findings offer novel perspectives on the possible application of targeted gut bacteria for the prevention and management of EC.
Collapse
Affiliation(s)
- Falide Atabieke
- The Second Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | | | - Munire Aierken
- Department of Disinfection and Vector Biocontrol, Disease Prevention and Control Center, Urumqi, China
| | - Mayinuer Rehaman
- Department of Physiology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Qi-Qi Zhang
- The Second Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jian Li
- The Second Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yu Xia
- The Second Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yierzhati Aizezi
- Center of Critical Care Medicine, First Affiliated Hospital of Xinjiang Medical University, Urumqi
| | - Diliyaer Dilixiati
- Department of Urology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Hong-Liang Gao
- The Second Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Zhi-Qiang Zhang
- The Second Department of Gastroenterology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
41
|
Chen X, Wei J, Zhang L, Wang H, Zhang Y, Li Z, Wang X, Liu L, Zhang Y, Zhang T. Association between plasma short-chain fatty acids and inflammation in human immunodeficiency virus-associated neurocognitive disorder: a pilot study. Lipids Health Dis 2025; 24:66. [PMID: 39984934 PMCID: PMC11846350 DOI: 10.1186/s12944-025-02477-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND AND AIMS Short-chain fatty acids (SCFAs), key metabolites produced by gut microbiota, have neuroprotective effects in neurodegenerative diseases by modulating immune responses. However, their role in human immunodeficiency virus (HIV)-associated neurocognitive disorder (HAND) remains largely unexplored. METHODS We recruited HAND patients, HIV Control, and healthy controls (HC). Plasma SCFAs and SCFA-producing gut microbiota were quantified via gas chromatography-mass spectrometry and fecal metagenomic analysis. Inflammatory cytokine levels were measured using liquid chromatography. Receiver operating characteristic (ROC) curves were generated to evaluate the predictive accuracy of SCFAs for HAND. RESULTS Plasma SCFAs were significantly reduced in HAND patients, correlating with a decrease in SCFA-producing gut bacteria, such as Prevotella and its related species. Reduced SCFAs were positively correlated with pro-inflammatory cytokines and cognitive impairment, while being negatively correlated with anti-inflammatory cytokines. ROC curve analysis demonstrated that several SCFAs exhibited strong predictive accuracy for HAND status. CONCLUSIONS SCFAs may influence cognitive function by modulating inflammatory responses, and identifies plasma SCFAs as potential biomarkers and therapeutic targets for HAND. Further investigation is needed to delineate the mechanisms that SCFAs influence HAND pathology.
Collapse
Affiliation(s)
- Xue Chen
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Youan Hospital, Beijing Institute of Hepatology, Capital Medical University, Beijing, China
| | - Jiaqi Wei
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Ling Zhang
- Beijing Youan Hospital, Beijing Institute of Hepatology, Capital Medical University, Beijing, China
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hu Wang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yang Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Zhen Li
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xia Wang
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Lifeng Liu
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yulin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China.
| | - Tong Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
42
|
Amillano-Cisneros JM, Fuentes-Valencia MA, Leyva-Morales JB, Savín-Amador M, Márquez-Pacheco H, Bastidas-Bastidas PDJ, Leyva-Camacho L, De la Torre-Espinosa ZY, Badilla-Medina CN. Effects of Microorganisms in Fish Aquaculture from a Sustainable Approach: A Review. Microorganisms 2025; 13:485. [PMID: 40142378 PMCID: PMC11945242 DOI: 10.3390/microorganisms13030485] [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: 01/25/2025] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025] Open
Abstract
Aquaculture is the fastest-growing food production sector. However, it faces significant challenges, including demand from a growing global population, which is estimated to reach 10.4 billion by the year 2100, disease outbreaks, environmental impacts, and the overuse of antibiotics. To address these issues, sustainable alternatives such as the use of microorganisms (probiotics, bacteriophages, and genetically modified microorganisms) have gained attention. This review examines the effects of these microorganisms on fish aquaculture, focusing on their potential to improve growth, health, and disease resistance while reducing environmental impacts. Probiotics, particularly lactic acid bacteria and yeasts, have been shown to enhance immune responses, digestive enzyme activity, and nutrient absorption in fish. Bacteriophages offer a promising alternative to antibiotics for controlling bacterial pathogens, with studies demonstrating their efficacy in reducing mortality rates in infected fish. Additionally, genetically modified microorganisms (GMMs) have been explored for their ability to produce beneficial compounds, such as enzymes and antimicrobial peptides, which can improve fish health and reduce the need for chemical treatments. Despite their potential, challenges such as regulatory hurdles, public acceptance, and environmental risks must be addressed. This review highlights the importance of further research to optimize the use of microorganisms in aquaculture and underscores their role in promoting sustainable practices. By integrating these biological tools, the aquaculture industry can move towards a more sustainable and environmentally friendly future.
Collapse
Affiliation(s)
- Jesús Mateo Amillano-Cisneros
- Ingeniería en Agrobiotecnología, Universidad Politécnica del Mar y la Sierra (UPMYS), La Cruz 82700, Mexico
- Maestría en Biotecnología Agropecuaria, Universidad Politécnica del Mar y la Sierra (UPMYS), La Cruz 82700, Mexico
- Ingeniería en Producción Animal, Universidad Politécnica del Mar y la Sierra (UPMYS), La Cruz 82700, Mexico
| | - María Anel Fuentes-Valencia
- Ingeniería en Agrobiotecnología, Universidad Politécnica del Mar y la Sierra (UPMYS), La Cruz 82700, Mexico
- Maestría en Biotecnología Agropecuaria, Universidad Politécnica del Mar y la Sierra (UPMYS), La Cruz 82700, Mexico
- Ingeniería en Producción Animal, Universidad Politécnica del Mar y la Sierra (UPMYS), La Cruz 82700, Mexico
| | - José Belisario Leyva-Morales
- Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Pachuca 42184, Mexico
- Centro de Investigación en Recursos Naturales y Sustentabilidad (CIRENYS), Universidad Bernardo O’Higgins, Avenida Viel 1497, Santiago de Chile 8370993, Chile
| | - Macario Savín-Amador
- Coordinación de Ingenierías, Universidad Tecnológica de La Paz, La Paz 23088, Mexico
| | - Henri Márquez-Pacheco
- Ingeniería en Agrobiotecnología, Universidad Politécnica del Mar y la Sierra (UPMYS), La Cruz 82700, Mexico
| | | | - Lucía Leyva-Camacho
- Departamento de Salud-Licenciatura en Ciencias Biomédicas, Universidad Autónoma de Occidente, Guasave 81044, Mexico
| | | | - César Noé Badilla-Medina
- Ingeniería en Producción Animal, Universidad Politécnica del Mar y la Sierra (UPMYS), La Cruz 82700, Mexico
| |
Collapse
|
43
|
Sánchez-Martínez E, Rondeau LE, Garrido-Romero M, da Luz BB, Haas DA, Yuen G, Hall P, Dang R, Wang XY, Moreno-Serna L, López-Sanz C, Nuñez-Borque E, Garrido-Arandia M, Diaz-Perales A, Carrasco YR, Koenig JF, Walker TD, Jordana M, Verdu EF, Surette MG, Ojeda P, Vega F, Blanco C, Shreffler WG, Patil SU, Moreno FJ, Jiménez-Saiz R, Caminero A. Microbial metabolism of food allergens determines the severity of IgE-mediated anaphylaxis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638013. [PMID: 40027733 PMCID: PMC11870547 DOI: 10.1101/2025.02.17.638013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Anaphylaxis is an acute, potentially life-threatening reaction, often triggered by foods and largely mediated by IgE. A critically important aspect of anaphylaxis pertains to the factors that modulate its severity. The human microbiota is known to influence oral tolerance, but the microbial mechanisms directly involved in IgE-mediated anaphylaxis remain unknown. Here, we demonstrate that human saliva harbors peanut-degrading bacteria that metabolize immunodominant allergens (Ara h 1 and Ara h 2) and alter IgE binding. Additionally, we provide in vivo evidence showing that oral bacteria metabolize peanut allergens, influencing systemic allergen exposure and the severity of anaphylaxis. Finally, in a clinical study, we observe that common peanut-degrading bacteria, such as Rothia, from the oral cavity, are more abundant in peanut-allergic patients who exhibit better tolerance to allergen exposure. Altogether, these results demonstrate the role of the human microbiota in modulating IgE-mediated reactions through allergen metabolism. These findings reveal a novel microbial mechanism with potential to prevent, or reduce, the severity of IgE-mediated anaphylaxis.
Collapse
Affiliation(s)
- Elisa Sánchez-Martínez
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Liam E. Rondeau
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Manuel Garrido-Romero
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Bioactivity and Food Analysis, Instituto de Investigación en Ciencias de la Alimentación (CIAL), CSIC-UAM, CEI, Madrid, Spain
| | - Bruna Barbosa da Luz
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Dominic A. Haas
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Gavin Yuen
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Peter Hall
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Rebecca Dang
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Xuan-Yu Wang
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Lucía Moreno-Serna
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Celia López-Sanz
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Emilio Nuñez-Borque
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Maria Garrido-Arandia
- Centre for Plant Biotechnology and Genomics (CBGP), Universidad Politécnica de Madrid (UPM-INIA), Madrid, Spain
| | - Araceli Diaz-Perales
- Centre for Plant Biotechnology and Genomics (CBGP), Universidad Politécnica de Madrid (UPM-INIA), Madrid, Spain
| | - Yolanda R. Carrasco
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
| | - Joshua F.E. Koenig
- Department of Medicine, McMaster Immunology Research Centre (MIRC), Schroeder Allergy and Immunology Research Institute (SAIRI), McMaster University, Hamilton, ON, Canada
| | - Tina D. Walker
- Department of Medicine, McMaster Immunology Research Centre (MIRC), Schroeder Allergy and Immunology Research Institute (SAIRI), McMaster University, Hamilton, ON, Canada
| | - Manel Jordana
- Department of Medicine, McMaster Immunology Research Centre (MIRC), Schroeder Allergy and Immunology Research Institute (SAIRI), McMaster University, Hamilton, ON, Canada
| | - Elena F. Verdu
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Michael G. Surette
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| | - Pedro Ojeda
- Clínica de Asma y Alergia Dres. Ojeda, Madrid, Spain
| | - Francisco Vega
- Department of Allergy, Hospital Universitario de La Princesa, IIS-Princesa, Madrid, Spain
| | - Carlos Blanco
- Department of Allergy, Hospital Universitario de La Princesa, IIS-Princesa, Madrid, Spain
| | - Wayne G. Shreffler
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sarita U. Patil
- Food Allergy Center and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - F. Javier Moreno
- Department of Bioactivity and Food Analysis, Instituto de Investigación en Ciencias de la Alimentación (CIAL), CSIC-UAM, CEI, Madrid, Spain
| | - Rodrigo Jiménez-Saiz
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Medicine, McMaster Immunology Research Centre (MIRC), Schroeder Allergy and Immunology Research Institute (SAIRI), McMaster University, Hamilton, ON, Canada
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria (UFV), Madrid, Spain
| | - Alberto Caminero
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
44
|
Li X, Cao L, Li J, Li Z, Ma H, Cheng S, Xu H, Zhao Y. Orally Administrated Inulin-Modified Nanozymes for CT-Guided IBD Theranostics. Int J Nanomedicine 2025; 20:2119-2131. [PMID: 39990289 PMCID: PMC11846537 DOI: 10.2147/ijn.s497558] [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: 10/17/2024] [Accepted: 02/06/2025] [Indexed: 02/25/2025] Open
Abstract
Background Inflammatory bowel disease (IBD) is a chronic inflammatory bowel disease with no clinical cure. Excessive production of reactive oxygen species (ROS) at the inflammatory sites leads to the onset and progression of IBD. And the current non-invasive imaging methods are not ideal for the diagnosis and monitoring of IBD. Methods Herein, we developed inulin (IN)-coated cerium oxide nanoparticles (CeO2@IN NPs) for treatment and monitoring of IBD guided by computed tomography (CT). The physicochemical properties, ROS scavenging ability and CT imaging capabilities of CeO2@IN were investigated in vitro. Moreover, the therapeutic and targeted inflammation imaging effects of CeO2@IN were validated in dextran sulfate sodium (DSS)-induced colitis model. Results CeO2@IN with catalase (CAT) and superoxide dismutase (SOD) capabilities effectively scavenged ROS, thus protecting the cells against oxidative stress. In colitis model mice, orally administered CeO2@IN successfully traversed the gastrointestinal tract to reach the colon under the protection of IN, and effectively reduced intestinal inflammation, thereby maintaining the intestinal epithelial integrity. Notably, CeO2@IN performed better than conventional CT contrast agents for gastrointestinal tract imaging, particularly in detecting the inflamed areas in the colon. In addition, CeO2@IN exhibited excellent biocompatibility in vitro and in vivo. Conclusion The study provided a novel integrated diagnostic and therapeutic tool for the treatment and monitoring of IBD, presenting great potential as a clinical application for IBD.
Collapse
Affiliation(s)
- Xinwen Li
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Lin Cao
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Jianmin Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Zhengyang Li
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Hongyu Ma
- Image Center, Cangzhou Integrated Traditional and Western Medicine Hospital, Cangzhou, 061000, People’s Republic of China
| | - Shifeng Cheng
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Hongyi Xu
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| | - Yang Zhao
- Department of Radiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, People’s Republic of China
| |
Collapse
|
45
|
Wang Z, Tian L, Jiang Y, Ning L, Zhu X, Chen X, Xuan B, Zhou Y, Ding J, Ma Y, Zhao Y, Huang X, Hu M, Fang JY, Shen N, Cao Z, Chen H, Wang X, Hong J. Synergistic role of gut-microbial L-ornithine in enhancing ustekinumab efficacy for Crohn's disease. Cell Metab 2025:S1550-4131(25)00007-5. [PMID: 39978335 DOI: 10.1016/j.cmet.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/27/2024] [Accepted: 01/08/2025] [Indexed: 02/22/2025]
Abstract
The role of the intestinal microbiome in Crohn's disease (CD) treatment remains poorly understood. This study investigates microbe-host interactions in CD patients undergoing ustekinumab (UST) therapy. Fecal metagenome, metabolome, and host transcriptome data from 85 CD patients were analyzed using multi-omics integration and mediation analysis. Our findings reveal significant microbiome-metabolite-host interactions. Specifically, Faecalibacterium prausnitzii was linked to altered L-ornithine biosynthesis, resulting in higher L-ornithine levels in patients before UST therapy. In vivo and in vitro studies demonstrated that microbiome-derived L-ornithine enhances UST treatment sensitivity in CD by disrupting the host IL-23 receptor signaling and inhibiting Th17 cell stabilization through the IL-12RB1/TYK2/STAT3 axis. L-ornithine significantly enhances the therapeutic efficacy of UST in CD patients, as demonstrated in a prospective clinical trial. These findings suggest that targeting specific microbe-host metabolic pathways may improve the efficacy of inflammatory bowel disease (IBD) treatments.
Collapse
Affiliation(s)
- Zhenyu Wang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Li Tian
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yi Jiang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Lijun Ning
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Xiaoqiang Zhu
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Xuejie Chen
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Baoqin Xuan
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Yilu Zhou
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Jinmei Ding
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Yanru Ma
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Ying Zhao
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Xiaowen Huang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Muni Hu
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Jing-Yuan Fang
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China
| | - Nan Shen
- Department of Infectious Disease, Shanghai Children's Medical Center, National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Dongfang Rd. 1678, Shanghai 200127, China
| | - Zhijun Cao
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China.
| | - Haoyan Chen
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China.
| | - Xiaoyan Wang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China.
| | - Jie Hong
- Division of Gastroenterology and Hepatology, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Middle Shandong Road, Shanghai 200001, China.
| |
Collapse
|
46
|
Gutierrez MW, van Tilburg Bernardes E, Ren E, Kalbfleisch KN, Day M, Lameu EL, Glatthardt T, Mercer EM, Sharma S, Zhang H, Al-Azawy A, Chleilat F, Hirota SA, Reimer RA, Arrieta MC. Early-life gut mycobiome core species modulate metabolic health in mice. Nat Commun 2025; 16:1467. [PMID: 39922818 PMCID: PMC11807121 DOI: 10.1038/s41467-025-56743-8] [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: 02/12/2024] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
The gut microbiome causally contributes to obesity; however, the role of fungi remains understudied. We previously identified three core species of the infant gut mycobiome (Rhodotorula mucilaginosa, Malassezia restricta and Candida albicans) that correlated with body mass index, however their causal contributions to obesity development are unknown. Here we show the effects of early-life colonization by these fungal species on metabolic health in gnotobiotic mice fed standard (SD) or high-fat-high-sucrose (HFHS) diets. Each species resulted in bacterial microbiome compositional and functional differences. R. mucilaginosa and M. restricta increased adiposity in mice fed SD, while only R. mucilaginosa exacerbated metabolic disease. In contrast, C. albicans resulted in leanness and resistance to diet-induced obesity. Intestinal nutrient transporter expression was unaffected by the presence of fungi in jejunal enteroids, yet the immune landscape in white adipose tissue was distinctly impacted by each fungal species, suggesting that these phenotypes may be a result of fungal immune regulation. This work revealed that three common fungal colonizers have distinct causal influences on obesity and metabolic inflammation and justifies the consideration of fungi in microbiome research on host metabolism.
Collapse
Affiliation(s)
- Mackenzie W Gutierrez
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Erik van Tilburg Bernardes
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Ellen Ren
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Kristen N Kalbfleisch
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Madeline Day
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Ewandson Luiz Lameu
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Thaís Glatthardt
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Emily M Mercer
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Sunita Sharma
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Hong Zhang
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Ali Al-Azawy
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Faye Chleilat
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Simon A Hirota
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Raylene A Reimer
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | - Marie-Claire Arrieta
- Department of Pediatrics, University of Calgary, Calgary, AB, Canada.
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada.
- International Microbiome Centre, Snyder Institute, University of Calgary, Calgary, AB, Canada.
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
47
|
Han J, Wang M, Zhou S, Wang Z, Duan D, Li M, Li X, Xin W, Li X. The Joint Contribution of Host Genetics and Probiotics to Pig Growth Performance. Microorganisms 2025; 13:358. [PMID: 40005725 PMCID: PMC11857988 DOI: 10.3390/microorganisms13020358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/03/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Intestinal probiotics significantly regulate the growth performance of their host, with their composition being influenced by various factors. While many studies have explored how gut microbiota composition affects growth traits such as body weight and BMI, the research on probiotics influenced by host genetic factors, and their subsequent impact on host growth performance, remains limited. To address this research gap, we collected fecal and tissue samples, as well as phenotypic data, from 193 Yunong black pigs at 280 days of age. We then sequenced and genotyped all 193 subjects using the 50K SNP BeadChip, yielding a comprehensive dataset for genetic and microbiome analyses. We then employed microbiome-wide association studies (MWAS), a meta-analysis, and microbiome-wide genetic association studies (MGWASs) to examine the relationship between host genetics, gut microbiota, and growth performance. Four key microbial taxa, namely Coprococcus, Blautia, Ruminococcaceae, and RF16, were identified as being significantly associated with body weight and BMI. The MGWAS analysis revealed that both Coprococcus and Ruminococcaceae were significantly associated with host genomic variations. A total of four important single nucleotide polymorphisms (SNPs) were mapped to two chromosomal regions, corresponding to three candidate genes. Among them, the candidate genes INPP4B, SCOC, and PABPC4L were identified as being related to the abundance of key microbes. This study provides new insights into the joint contributions of host genetics and probiotics to host growth traits, offering theoretical guidance and data support for the development of efficient and targeted breeding strategies.
Collapse
Affiliation(s)
- Jinyi Han
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Mingyu Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Shenping Zhou
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| | - Zhenyu Wang
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| | - Dongdong Duan
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| | - Mengyu Li
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| | - Xiuling Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
| | - Wenshui Xin
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| | - Xinjian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| |
Collapse
|
48
|
He J, Zhang F, Fang M, Zhang Y, Zhu C, Xiang S, Yu D, Wu H, Shu Y. Alteration of intestinal microbiota-intestinal barrier interaction interferes with intestinal health after microcystin-LR exposure in Lithobates catesbeianus tadpoles. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 279:107249. [PMID: 39826206 DOI: 10.1016/j.aquatox.2025.107249] [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: 07/27/2024] [Revised: 01/13/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
There remains uncertainty regarding the influence of microcystin-leucine arginine (MC-LR) on amphibian intestinal health, specifically how MC-LR interferes with intestinal microbiota following exposure to environmental concentrations. In this study, Lithobates catesbeianus tadpoles were exposed to varying MC-LR concentrations (0, 0.5, and 2 µg/L) over a 30-day period. The aim was to investigate how altered interactions between tadpole intestinal microbiota and the intestinal barrier influence intestinal health following MC-LR exposure. Following exposure to the MC-LR at low ambient concentrations, tadpole intestinal tissue was damaged. It had increased permeability, reduced pathogen inhibition capacity, and impaired digestive function. Additionally, there was a significant increase in lipopolysaccharide content and upregulation of downstream response genes, including TLR4, MyD88, and NF-κB, within the intestinal tissue. Therefore, eosinophils' count and pro-inflammatory cytokines' expression increased. In addition, MC-LR exposure induced oxidative stress and mitochondrial structural damage by increasing the levels of reactive oxygen species in intestinal tissue. CytoC and Bax transcription, as well as caspase 9 and caspase 3 activities, increased significantly. Significant downregulation of Bcl-2 transcription promoted apoptosis in tadpole intestinal cells. MC-LR exposure disrupted intestinal microbiota and metabolism in tadpoles. Correlation analysis revealed a strong association between intestinal microbiota and oxidative stress, inflammation, immunity, and tissue damage in the intestine. Conclusively, this study provides the first demonstration that MC-LR significantly affects amphibian intestinal microbiota, highlighting tadpoles' susceptibility to environmental risks posed by MC-LR.
Collapse
Affiliation(s)
- Jun He
- Department of Pathology, Wannan Medical College, Wuhu, Anhui 241002, China; Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Affiliated Middle School, Anhui Normal University, Wuhu 241002, China
| | - Fengqi Zhang
- Department of Pathology, Wannan Medical College, Wuhu, Anhui 241002, China
| | - Minglan Fang
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Affiliated Middle School, Anhui Normal University, Wuhu 241002, China
| | - Yuchen Zhang
- Department of Pathology, Wannan Medical College, Wuhu, Anhui 241002, China
| | - Changjing Zhu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Affiliated Middle School, Anhui Normal University, Wuhu 241002, China
| | - Shangfei Xiang
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Affiliated Middle School, Anhui Normal University, Wuhu 241002, China
| | - Desheng Yu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Affiliated Middle School, Anhui Normal University, Wuhu 241002, China
| | - Hailong Wu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Affiliated Middle School, Anhui Normal University, Wuhu 241002, China.
| | - Yilin Shu
- Collaborative Innovation Center of Recovery and Reconstruction of Degraded Ecosystem in Wanjiang Basin Co-founded by Anhui Province and Ministry of Education, School of Ecology and Environment, Affiliated Middle School, Anhui Normal University, Wuhu 241002, China.
| |
Collapse
|
49
|
Zhou F, Shang BH, Liu CW, Fang WW, Wen S, Zeng HZ, Huang JA, Liu ZH. Comparative study of the anti-obesity effects of white tea and dark tea: Insights from microbiome and metabolomics. Food Res Int 2025; 202:115666. [PMID: 39967141 DOI: 10.1016/j.foodres.2025.115666] [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: 09/30/2024] [Revised: 12/11/2024] [Accepted: 01/01/2025] [Indexed: 02/20/2025]
Abstract
The anti-obesity effects of tea and its functional components have been extensively studied. However, the protective effects of different types of tea against obesity induced by a high-fat diet (HFD) and the underlying mechanisms remain unclear. This study systematically compared the effects of white tea and dark tea on obese rats and explored their mechanisms. The results indicated that dark tea extracts (DT) higher concentrations of theabrownins and gallic acid, while white tea extracts (WT) contained abundant levels of polyphenols and amino acids. Moreover, both WT and DT effectively improved obesity-related symptoms, including weight loss, reduced fat accumulation, improved dyslipidemia, and alleviated liver and colon damage. Specifically, WT primarily functioned by inhibiting white fat accumulation and enhancing UCP1 expression in brown fat, leading to more significant weight loss. Conversely, DT increased both the quantity and uniform distribution of colonic goblet cells and elevated the expression levels of tight junction proteins in obese rats, thereby providing better protection for the intestinal barrier. Furthermore, 16S rRNA sequencing revealed that WT and DT regulated gut microbiota imbalances, restored microbiota diversity, inhibited the growth of potentially harmful bacteria, and promoted the proliferation of beneficial bacteria. Metabolomics analyses demonstrated that WT and DT increased the concentration of short-chain fatty acids in the feces of obese rats, regulated the biosynthesis of phenylalanine, tyrosine, and tryptophan, as well as the biosynthesis pathways of valine, leucine, and isoleucine, while decreasing the levels of these amino acids in feces. In conclusion, this study provides new evidence supporting the idea that tea can mitigate HFD-induced obesity through the regulation of gut microbiota and alteration of fecal metabolite profiles.
Collapse
Affiliation(s)
- Fang Zhou
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, PR China; School of Chemistry and Environmental Sciences, Xiangnan University, Chenzhou, Hunan 423000, PR China; National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, Hunan Agricultural University, Changsha, Hunan 410128, PR China; Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Hunan 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Hunan 410128, PR China
| | - Bo-Hao Shang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, PR China; National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, Hunan Agricultural University, Changsha, Hunan 410128, PR China; Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Hunan 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Hunan 410128, PR China
| | - Chang-Wei Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, PR China; National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, Hunan Agricultural University, Changsha, Hunan 410128, PR China; Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Hunan 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Hunan 410128, PR China
| | - Wen-Wen Fang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, PR China; National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, Hunan Agricultural University, Changsha, Hunan 410128, PR China; Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Hunan 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Hunan 410128, PR China
| | - Shuai Wen
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, PR China; National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, Hunan Agricultural University, Changsha, Hunan 410128, PR China; Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Hunan 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Hunan 410128, PR China
| | - Hong-Zhe Zeng
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, PR China; National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, Hunan Agricultural University, Changsha, Hunan 410128, PR China; Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Hunan 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Hunan 410128, PR China
| | - Jian-An Huang
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, PR China; National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, Hunan Agricultural University, Changsha, Hunan 410128, PR China; Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Hunan 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Hunan 410128, PR China.
| | - Zhong-Hua Liu
- Key Laboratory of Tea Science of Ministry of Education, Hunan Agricultural University, Changsha, Hunan 410128, PR China; National Research Center of Engineering Technology for Utilization of Functional Ingredients from Botanicals, Hunan Agricultural University, Changsha, Hunan 410128, PR China; Co-Innovation Center of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Hunan 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Hunan 410128, PR China.
| |
Collapse
|
50
|
Lan T, Cai M, Wang S, Lu Y, Tang Z, Tang Q, Gao J, Xu Y, Peng X, Sun Z. Effects of adding niacinamide to diets with normal and low protein levels on the immunity, antioxidant, and intestinal microbiota in growing-finishing pigs. J Nutr Biochem 2025; 136:109809. [PMID: 39549857 DOI: 10.1016/j.jnutbio.2024.109809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/03/2024] [Accepted: 11/11/2024] [Indexed: 11/18/2024]
Abstract
This study aimed to investigate the effects of nicotinamide (NAM) applied to diets with different crude protein levels on immune function, antioxidant capacity, and intestinal flora in growing-finishing pigs. Forty barrows (37.0±1.0 kg) were randomly allocated to one of four dietary treatments (n=10 per group). The diets in the two phases consisted of a basal diet with 30 mg/kg NAM, a basal diet with 360 mg/kg NAM, a low-protein diet with 30 mg/kg NAM, and a low-protein diet with 360 mg/kg NAM. The results showed that dietary addition of 360 mg/kg NAM decreased IL-12, malondialdehyde, IgG and IgM contents in the plasma and increased total superoxide dismutase activity and total antioxidant capacity in the colonic mucosa (P < .05). Supplementing the diet with 360 mg/kg NAM increased mRNA expression of the nucleotide-binding oligomerization domain containing 2 and nuclear factor erythroid 2-related factor 2 and protein expression of nuclear factor kappa-B and toll-like receptor 4 in the colonic mucosa (P < .05). The concentrations of acetic acid and butyric acid in the colonic contents and the abundance of Actinobacteriota in the colon at the phylum level were significantly decreased by feeding low-protein diets (P < .05). Additionally, the addition of 360 mg/kg NAM to diets increased (P < .05) the Sobs, Ace, and Chao indices of colonic microorganisms in pigs. Overall, the rational use of NAM can improve inflammatory status, enhance antioxidant capacity and intestinal barrier function, and increase colonic microbial diversity in growing-finishing pigs.
Collapse
Affiliation(s)
- Tianyi Lan
- Center for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Meiya Cai
- Center for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Sishen Wang
- Center for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yingying Lu
- Center for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Zhiru Tang
- Center for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Qingsong Tang
- Center for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Jingchun Gao
- Center for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Yetong Xu
- Center for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Xie Peng
- Center for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Zhihong Sun
- Center for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China.
| |
Collapse
|