1
|
Fumagalli A, Castells-Nobau A, Trivedi D, Garre-Olmo J, Puig J, Ramos R, Ramió-Torrentà L, Pérez-Brocal V, Moya A, Swann J, Martin-Garcia E, Maldonado R, Fernández-Real JM, Mayneris-Perxachs J. Archaea methanogens are associated with cognitive performance through the shaping of gut microbiota, butyrate and histidine metabolism. Gut Microbes 2025; 17:2455506. [PMID: 39910065 PMCID: PMC11810085 DOI: 10.1080/19490976.2025.2455506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/28/2024] [Accepted: 01/13/2025] [Indexed: 02/07/2025] Open
Abstract
The relationship between bacteria, cognitive function and obesity is well established, yet the role of archaeal species remains underexplored. We used shotgun metagenomics and neuropsychological tests to identify microbial species associated with cognition in a discovery cohort (IRONMET, n = 125). Interestingly, methanogen archaeas exhibited the strongest positive associations with cognition, particularly Methanobrevibacter smithii (M. smithii). Stratifying individuals by median-centered log ratios (CLR) of M. smithii (low and high M. smithii groups: LMs and HMs) revealed that HMs exhibited better cognition and distinct gut bacterial profiles (PERMANOVA p = 0.001), characterized by increased levels of Verrucomicrobia, Synergistetes and Lentisphaerae species and reduced levels of Bacteroidetes and Proteobacteria. Several of these species were linked to the cognitive test scores. These findings were replicated in a large-scale validation cohort (Aging Imageomics, n = 942). Functional analyses revealed an enrichment of energy, butyrate, and bile acid metabolism in HMs in both cohorts. Global plasma metabolomics by CIL LC-MS in IRONMET identified an enrichment of methylhistidine, phenylacetate, alpha-linolenic and linoleic acid, and secondary bile acid metabolism associated with increased levels of 3-methylhistidine, phenylacetylgluamine, adrenic acid, and isolithocholic acid in the HMs group. Phenylacetate and linoleic acid metabolism also emerged in the Aging Imageomics cohort performing untargeted HPLC-ESI-MS/MS metabolic profiling, while a targeted bile acid profiling identified again isolithocholic acid as one of the most significant bile acid increased in the HMs. 3-Methylhistidine levels were also associated with intense physical activity in a second validation cohort (IRONMET-CGM, n = 116). Finally, FMT from HMs donors improved cognitive flexibility, reduced weight, and altered SCFAs, histidine-, linoleic acid- and phenylalanine-related metabolites in the dorsal striatum of recipient mice. M. smithii seems to interact with the bacterial ecosystem affecting butyrate, histidine, phenylalanine, and linoleic acid metabolism with a positive impact on cognition, constituting a promising therapeutic target to enhance cognitive performance, especially in subjects with obesity.
Collapse
Affiliation(s)
- Andrea Fumagalli
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain
- Integrative Systems Medicine and Biology Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Salt, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III; Madrid, Spain
| | - Anna Castells-Nobau
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain
- Integrative Systems Medicine and Biology Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Salt, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III; Madrid, Spain
| | - Dakshat Trivedi
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Josep Garre-Olmo
- serra-hunter program Department of Nursing, University of Girona, Girona, Spain
| | - Josep Puig
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
- Institute of Diagnostic Imaging (IDI)-Research Unit (IDIR), Parc Sanitari Pere Virgili, Barcelona, Spain
- Medical Imaging, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- Department of Radiology (IDI), Dr. Josep Trueta University Hospital, Girona, Spain
| | - Rafel Ramos
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
- Vascular Health Research Group of Girona (ISV-Girona), Jordi Gol Institute for Primary Care Research (Institut Universitari per a la Recerca en Atenció Primària Jordi Gol I Gorina -IDIAPJGol), Red de Investigación en Cronicidad, Atención Primaria y Promoción de la Salud-RICAPPS- ISCIII Girona Biomedical Research Institute (IDIBGI), Dr. Josep Trueta University Hospital, Girona, Catalonia, Spain
- Research in Vascular Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Dr. Josep Trueta University Hospital, Girona, Spain
| | - Lluís Ramió-Torrentà
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
- Neuroimmunology and Multiple Sclerosis Unit, Department of Neurology, Dr. Josep Trueta University Hospital, Girona, Spain
- Neurodegeneration and Neuroinflammation Research Group, IDIBGI-CERCA, Girona, Spain
| | - Vicente Pérez-Brocal
- Area of Genomics and Health, Foundation for the Promotion of Sanitary and Biomedical Research of Valencia Region (FISABIO-Public Health), Valencia, Spain
- Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Andrés Moya
- Area of Genomics and Health, Foundation for the Promotion of Sanitary and Biomedical Research of Valencia Region (FISABIO-Public Health), Valencia, Spain
- Biomedical Research Networking Center for Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Institute for Integrative Systems Biology (I2SysBio), University of Valencia and Spanish National Research Council (CSIC), Valencia, Spain
| | - Jonathan Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Elena Martin-Garcia
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Rafael Maldonado
- Laboratory of Neuropharmacology, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, Spain
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III; Madrid, Spain
| | - Jordi Mayneris-Perxachs
- Department of Diabetes, Endocrinology and Nutrition, Dr. Josep Trueta University Hospital, Girona, Spain
- Integrative Systems Medicine and Biology Group, Girona Biomedical Research Institute (IDIBGI-CERCA), Salt, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III; Madrid, Spain
| |
Collapse
|
2
|
He Z, Xiong H, Cai Y, Chen W, Shi M, Liu L, Wu K, Deng X, Deng X, Chen T. Clostridium butyricum ameliorates post-gastrectomy insulin resistance by regulating the mTORC1 signaling pathway through the gut-liver axis. Microbiol Res 2025; 297:128154. [PMID: 40188705 DOI: 10.1016/j.micres.2025.128154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 05/04/2025]
Abstract
Postoperative insulin resistance (IR) is a metabolic disorder characterized by decreased insulin sensitivity and elevated blood glucose levels following major surgery. Our previous clinical study identified a notable correlation between postoperative IR and gut microbiota, particularly butyrate-producing bacteria, yet the mechanisms remain unclear. In this study, we established gastric resection SD rat models to evaluate the impact of Clostridium butyricum NCU-27 (butyrate-producing bacteria) on postoperative IR. The results demonstrated significant reductions in fasting blood glucose (FBG), fasting insulin (FIns) levels, and HOMA-IR (6.64 ± 0.76 vs. 11.47 ± 1.32; 4.27 ± 0.59 vs. 7.40 ± 0.54) in the postoperative period compared to the control group (P < 0.05). Additionally, glucose tolerance and hepatic glycogen content were markedly improved (P < 0.001). Further exploration of butyrate demonstrated effects similar to C. butyricum NCU-27, potentially mediated through the gut-liver axis by inhibiting mTORC1 expression in liver cells, activating the IRS1/AKT pathway, enhancing glucose uptake and glycogen synthesis, suppressing gluconeogenesis, increasing insulin sensitivity, and improving IR. Finally, the use of mTORC1 agonists and inhibitors further confirmed the critical role of the mTORC1 pathway in mediating the beneficial effects of C. butyricum NCU-27 and butyrate on postoperative IR. In conclusion, this study elucidated that C. butyricum NCU-27 improves postoperative IR by regulating butyrate metabolism and inhibiting the mTORC1 pathway, offering new insights for preventing and treating post-gastrectomy IR.
Collapse
Affiliation(s)
- Zhipeng He
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China; Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Huan Xiong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yujie Cai
- Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Wenjing Chen
- Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Meng Shi
- Department of Gastrointestinal Surgery, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, Hubei 442008, China
| | - Lulin Liu
- Department of Vascular Surgery, Heyuan Hospital of Guangdong Provincial People's Hospital, Heyuan, Guangdong 51700, China
| | - Kai Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Xi Deng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Xiaorong Deng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China.
| | - Tingtao Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China; Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang 330031, China; National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China.
| |
Collapse
|
3
|
Salarini JS, Barroso LN, Freitas JV, Leite NC, de Paula TP, Padilha PDC, Peres WAF. The Brazilian traditional dietary pattern was associated with a lower risk of advanced steatosis in patients with metabolic dysfunction-associated steatotic liver disease. Nutr Res 2025; 139:66-77. [PMID: 40435907 DOI: 10.1016/j.nutres.2025.04.012] [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: 12/30/2024] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 06/29/2025]
Abstract
Identifying dietary patterns in patients with metabolic dysfunction-associated steatotic liver disease (MASLD) is critical, since nutrition is a modifiable risk factor that contributes to the etiology and progression of the disease. This study evaluated the association between dietary patterns and lipid and glycemic profiles, liver biomarkers, and degrees of steatosis and fibrosis in patients with MASLD. In this cross-sectional study, 70 patients who completed the entire dietary assessment were included in the analyses. Dietary intake was assessed through 24-hour dietary recalls, and 4 dietary patterns were identified by exploratory factor analysis and principal component analysis. Mean scores were analyzed by sex, age group, and nutritional status, and associations with main outcomes were tested by binomial logistic regression, with significance set at P > .05. There was greater adherence to the prudent pattern among men (95% CI 2.1-2.84), which consisted of fresh fruits and fruit juices, whole grains and breads, eggs and egg-based preparations, and coffee and tea, and a lower chance of advanced steatosis in individuals with greater adherence to the traditional Brazilian pattern (OR 0.46, 95% CI 0.20-0.97). This pattern, based on minimally processed foods rich in fiber, bioactive compounds, and antioxidants, was shown to be protective against advanced steatosis.
Collapse
Affiliation(s)
- Jéssica Silva Salarini
- Nutrition sector, Clementino Fraga Filho University Hospital - HUCFF, Rio de Janeiro Federal University - UFRJ, Rio de Janeiro, RJ, Brazil; Department of Nutrition and Dietetics, Josué de Castro Nutrition Institute - INJC, Rio de Janeiro Federal University - UFRJ, Rio de Janeiro, RJ, Brazil
| | - Lygia Nestal Barroso
- Nutrition sector, Clementino Fraga Filho University Hospital - HUCFF, Rio de Janeiro Federal University - UFRJ, Rio de Janeiro, RJ, Brazil; Department of Nutrition and Dietetics, Josué de Castro Nutrition Institute - INJC, Rio de Janeiro Federal University - UFRJ, Rio de Janeiro, RJ, Brazil
| | - Jade Veloso Freitas
- Department of Epidemiology, Institute of Social Medicine, Rio de Janeiro State University, Rio de Janeiro, RJ, Brazil
| | - Nathalie Carvalho Leite
- School of Medicine, Clementino Fraga Filho University Hospital - HUCFF, Rio de Janeiro Federal University - UFRJ, Rio de Janeiro, RJ, Brazil
| | - Tatiana Pereira de Paula
- Nutrition sector, Clementino Fraga Filho University Hospital - HUCFF, Rio de Janeiro Federal University - UFRJ, Rio de Janeiro, RJ, Brazil
| | - Patrícia de Carvalho Padilha
- Department of Nutrition and Dietetics, Josué de Castro Nutrition Institute - INJC, Rio de Janeiro Federal University - UFRJ, Rio de Janeiro, RJ, Brazil
| | - Wilza Arantes Ferreira Peres
- Department of Nutrition and Dietetics, Josué de Castro Nutrition Institute - INJC, Rio de Janeiro Federal University - UFRJ, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
4
|
Qiu X, Gao Q, Wang J, Zhang Z, Tao L. The microbiota-m 6A-metabolism axis: Implications for therapeutic strategies in gastrointestinal cancers. Biochim Biophys Acta Rev Cancer 2025; 1880:189317. [PMID: 40222422 DOI: 10.1016/j.bbcan.2025.189317] [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: 09/11/2024] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
Gastrointestinal (GI) cancers remain a leading cause of cancer-related mortality worldwide, with metabolic reprogramming recognized as a central driver of tumor progression and therapeutic resistance. Among the key regulatory layers, N6-methyladenosine (m6A) RNA modification-mediated by methyltransferases (writers such as METTL3/14), RNA-binding proteins (readers like YTHDFs and IGF2BPs), and demethylases (erasers including FTO and ALKBH5), plays a pivotal role in controlling gene expression and metabolic flux in the tumor context. Concurrently, the gut microbiota profoundly influences GI tumorigenesis and immune evasion by modulating metabolite availability and remodeling the tumor microenvironment. Recent evidence has uncovered a bidirectional crosstalk between microbial metabolites and m6A methylation: microbiota-derived signals dynamically regulate m6A deposition on metabolic and immune transcripts, while m6A modifications, in turn, regulate the stability and translation of key mRNAs such as PD-L1 and FOXP3. This reciprocal interaction forms self-reinforcing epigenetic circuits that drive tumor plasticity, immune escape, and metabolic adaptation. In this review, we dissect the molecular underpinnings of the microbiota-m6A-metabolism axis in GI cancers and explore its potential to inform novel strategies in immunotherapy, metabolic intervention, and microbiome-guided precision oncology.
Collapse
Affiliation(s)
- Xiuxiu Qiu
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qi Gao
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jiahui Wang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Zhanxia Zhang
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Li Tao
- Department of Oncology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
5
|
Ye Q, Hu Y, Jiang H, Luo T, Han L, Chen Y, Chen J, Ma L, He Z, Yan X. Maternal intestinal L. vaginalis facilitates embryo implantation and survival through enhancing uterine receptivity in sows. MICROBIOME 2025; 13:145. [PMID: 40533850 PMCID: PMC12175382 DOI: 10.1186/s40168-025-02141-7] [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] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 05/12/2025] [Indexed: 06/22/2025]
Abstract
BACKGROUND The embryo implantation quality during early pregnancy is the predominant factor for embryo survival and litter performance in sows. Gut microbiota is demonstrated to show a correlation to pregnancy outcomes by participating in regulating maternal metabolism. However, the specific functional microbiota and its mechanical effects on regulating embryo implantation and survival remain unclear. The objective of this study was to clarify whether embryo implantation and litter performance were affected by maternal intestinal microbiota, and to identify specific microbial communities and its mechanism in regulating embryo implantation. RESULTS In this study, we first conducted 16S rRNA sequencing and metabolomic analysis revealing the intestinal microbiota and metabolism of 42 sows with different litter size to select the potential functional microbiota that may contribute to embryo survival. Then, we explored the effects of that microbiota on embryo implantation and litter performance through microbiota transplantation in mice and sows. We found that maternal intestinal L. vaginalis exhibits enrichment in sows with higher litter size, which could facilitate embryo implantation and survival and ultimately increases litter size in mice. We further employed transcriptomic analysis to determine the characteristics of uterus, which found an enhanced uterine receptivity after L. vaginalis gavage. The plasma untargeted metabolomic analysis after L. vaginalis gavage in mice and targeted metabolomics analysis of in vitro cultured medium of L. vaginalis were used to evaluate the metabolic regulation of L. vaginalis and to reveal the underlying functional metabolites. Next, an increasing adhesion rate of endometrial-embryonic cells and an obvious increasing formation of pinopodes in cell surface of porcine endometrial epithelial cells were observed after treatments of L. vaginalis metabolites, especially galangin and daidzein. Also, the gene expression levels related to uterine receptivity were increased after treatments of L. vaginalis metabolites in porcine endometrial epithelial cells. Finally, we found that L. vaginalis or its metabolites supplementation during early gestation significantly increased the litter performance in sows. CONCLUSIONS Overall, intestinal microbial-host interactions can occur during early pregnancy and may be contribute to maternal metabolic changes and influence pregnancy outcomes in mammals. Our study provides insights of maternal intestinal L. vaginalis to enhance uterine receptivity and to benefit embryo/fetal survival through a gut-uterus axis, contributing to advanced concept and novel strategy to manipulate gut microbiota during early pregnancy, and in turn to improve embryo implantation and reduce embryo loss in sows. Video Abstract.
Collapse
Affiliation(s)
- Qianhong Ye
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Yifan Hu
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Haoyi Jiang
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Tingting Luo
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Longshan Han
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Yuwen Chen
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Jiaying Chen
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Libao Ma
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Ziyi He
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China
| | - Xianghua Yan
- National Key Laboratory of Agricultural Microbiology, Frontiers Science Center for Animal Breeding and Sustainable Production, Hubei Hongshan Laboratory, College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
- The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, 430070, Hubei, China.
- Hubei Provincial Engineering Laboratory for Pig Precision Feeding and Feed Safety Technology, Wuhan, 430070, Hubei, China.
- National Engineering Research Center for Green Feed and Healthy Breeding, Key Laboratory of Animal Molecular Nutrition, Ministry of Education, Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory of Nutrition and Breeding for High-quality Animal Products Institute of Feed Science, College of Animal Science, Zhejiang University, Zhejiang, 310058, Hangzhou, China.
| |
Collapse
|
6
|
Liu C, Chao S, Jia L, Yang Q, Chen Q, Niu Y. Integrative analyses of 16S rDNA sequencing and serum metabolomics demonstrate significant roles for the oral microbiota and serum metabolites in post-kidney transplant diabetes mellitus. Microbiol Spectr 2025:e0089225. [PMID: 40492760 DOI: 10.1128/spectrum.00892-25] [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: 04/04/2025] [Accepted: 05/09/2025] [Indexed: 06/12/2025] Open
Abstract
Oral microbiota and serum metabolites play crucial roles in diabetes, but their relationship with post-transplant diabetes mellitus (PTDM), a common complication post-kidney transplantation, is not well characterized. This study investigated the relationship of oral microbiota and serum metabolites with PTDM using integrative analysis of 16S rDNA sequencing and serum metabolomics. We recruited 61 kidney transplant recipients, including 30 in the PTDM group and 31 in normal glucose tolerance controls. Oral samples and serum samples were collected from all the kidney transplant patients to perform 16S rDNA sequencing and serum metabolomics analysis. We annotated 689 oral microbial species, including 134 species unique to the PTDM group and 157 species unique to the control group. PTDM group showed upregulation of 36 metabolites and downregulation of 19 metabolites. Based on the random forest machine learning algorithm, genera such as UCG-005 (AUC = 0.9355), Succinivibrio (AUC = 0.8108); Akkermansia (AUC = 0.7742), Anaerovibrio (AUC = 0.2667), and Schwartzia (AUC = 0.2667), and serum metabolites such as LPI 18:0 (AUC: 0.8086), methylglyoxal (AUC: 0.7946), Vulgarin (AUC: 0.7828), 2-mercaptobenzothiazole (AUC: 0.7591), and PI(18:0/20:3(5Z,8Z,11Z)) (AUC: 0.7419) showed high diagnostic potential and may serve as clinical biomarkers. Furthermore, clinical indicators in PTDM patients, such as creatinine, cystatin C, and urea, showed a significant association with the differential oral microbiota and serum metabolites. Dysbiosis in the oral microbiota of the PTDM patients was associated with changes in the serum metabolites and alterations in their functions. These findings provide new insights toward identifying mechanisms by which oral microbiota and serum metabolites contribute to the development of PTDM.IMPORTANCEThis study reveals an imbalance in oral microbiota in patients with post-transplant diabetes and uncovers the potential relationship between oral microbiota and serum metabolites. These findings provide new insights into the role of oral microbiota and serum metabolites in the treatment of post-transplant diabetes, offering relevant biomarkers for clinicians and future research.
Collapse
Affiliation(s)
- Chao Liu
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Urinary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Sheng Chao
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Lei Jia
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Qizhen Yang
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Qian Chen
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Yulin Niu
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
7
|
Hamari N, Blaak EE, Canfora EE. The impact of butyrate on glycemic control in animals and humans: a comprehensive semi-systemic review. Front Nutr 2025; 12:1603490. [PMID: 40557239 PMCID: PMC12185432 DOI: 10.3389/fnut.2025.1603490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 05/17/2025] [Accepted: 05/27/2025] [Indexed: 06/28/2025] Open
Abstract
The gut microbiome has been identified as a significant factor in host metabolism, playing a key role in the etiology of obesity, type 2 diabetes and cardiometabolic risk. Butyrate, produced by the gut microbiome from indigestible carbohydrates, has been shown to have beneficial effects on body weight control, inflammation, and insulin resistance, primarily evidenced by animal studies and in vitro experiments. However, translating these benefits to humans remains challenging due to variability in mode of butyrate administration or production upon fermentation of dietary fibers, as well as in butyrate absorption, and its metabolism. For instance, oral butyrate supplementation can directly increase circulating butyrate levels, thereby targeting peripheral tissues. In contrast, butyrate produced by the gut microbiome may also influence metabolism through local signaling mechanisms affecting peripheral tissues. Additionally, there may be large heterogeneity in the response of the individuals to butyrate interventions. Future research should aim to better understand butyrate kinetics and dynamics and its mechanisms in regulating intestinal and metabolic health. In human studies, longer-term, placebo-controlled trials are needed to establish the efficacy of either targeting butyrate production or supplementation in individuals with obesity and/or metabolic disturbances. Personalized dietary interventions based on individual microbiota composition and/or function and metabolic profiles may optimize butyrate production and its metabolic benefits. This could pave the way for effective butyrate-based interventions to improve metabolic health and prevent obesity-related complications.
Collapse
Affiliation(s)
| | | | - Emanuel E. Canfora
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center +, Maastricht, Netherlands
| |
Collapse
|
8
|
Tang W, Long Z, Xiao Y, Du J, Tang C, Chen J, Hou C. Dietary butyric acid intake, kidney function, and survival: The National Health and Nutrition Examination Surveys, 2005-2018. Clin Nutr ESPEN 2025; 67:453-462. [PMID: 40147762 DOI: 10.1016/j.clnesp.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 02/23/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Although preclinical data support the hypothesis that butyric acid supplementation improves kidney health, the clinical significance of dietary butyric acid intake in patients with chronic kidney disease (CKD) remains unconfirmed in large-sample studies. This study aimed to investigate the association between dietary butyric acid intake and all-cause mortality in the United States population, stratified by kidney function. METHODS We examined the relationship between dietary butyric acid intake, assessed through a 24-h dietary recall, and all-cause mortality among 23,008 consecutive adult participants from the National Health and Nutrition Examination Surveys (NHANES, 2005-2018), categorized by impaired versus normal kidney function (estimated glomerular filtration rate <60 vs ≥ 60 mL/min/1.72 m2), using multivariable Cox models. We also employed a restricted cubic spline based on Cox regression models to elucidate the nonlinear relationship between dietary butyric acid intake and mortality in patients. RESULT In participants with impaired kidney function, high dietary butyric acid intake was associated with lower mortality, while lower intake levels (reference) showed no such association: adjusted HRs (aHRs) were 0.67 (95 % CI: 0.45, 1.00), 0.65 (95 % CI: 0.45, 0.94), and 0.58 (95 % CI: 0.38, 0.89) for intake levels of the square root of butyric acid 0.25-0.45, 0.45-0.75, and >0.75 g/day, respectively. However, in participants with normal kidney function, no association between butyric acid levels and mortality was observed. Additionally, we identified an L-shaped association between the levels of the square root of dietary butyric acid intake and all-cause mortality in the CKD population, reaching a plateau at 0.52 g/day (butyric acid intake of approximately 0.27 g/day). CONCLUSION This study revealed a nonlinear association between high dietary butyric acid intake and reduced all-cause mortality in patients with chronic kidney disease. A plateau occurs after 0.27 g/day, and for individuals with CKD whose butyric acid intake is below approximately 0.27 g/day, increasing a butyrate-rich diet or supplementing with butyric acid preparations may help prevent progression to renal failure and associated adverse outcomes in CKD patients, thereby reducing mortality. Therefore, it can be considered a new therapeutic strategy for the treatment of chronic kidney disease.
Collapse
Affiliation(s)
- Wei Tang
- Hunan Key Laboratory of Kidney Disease and Blood Purification, and Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhengyi Long
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yang Xiao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jingyun Du
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Chenyuan Tang
- Hunan Key Laboratory of Kidney Disease and Blood Purification, and Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - JunXiang Chen
- Hunan Key Laboratory of Kidney Disease and Blood Purification, and Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Can Hou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Fan G, Liu Y, Tao L, Wang D, Huang Y, Yang X. Sodium butyrate alleviates colitis by inhibiting mitochondrial ROS mediated macrophage pyroptosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167756. [PMID: 40044062 DOI: 10.1016/j.bbadis.2025.167756] [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: 11/20/2024] [Revised: 02/06/2025] [Accepted: 02/26/2025] [Indexed: 04/15/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory bowel disease with unclear causes and limited treatment options. Sodium butyrate (NaB), a byproduct of dietary fiber in the intestine, has demonstrated efficacy in treating inflammation. However, the precise anti-inflammatory mechanisms of NaB in colon inflammation remain largely unexplored. This study aims to investigate the effects of NaB on dextran sulfate sodium (DSS)-induced colitis in rats. The findings indicate that oral administration of NaB effectively prevent colitis and reduce levels of serum or colon inflammatory factors. Additionally, NaB demonstrated in vitro inhibition of RAW264.7 inflammation cytokines induced by LPS, along with suppression of the ERK and NF-κB signaling pathway activation. Moreover, NaB mitigated LPS and Nigericin-induced RAW264.7 pyroptosis by reducing indicators of mitochondrial damage, including increased mitochondrial membrane potential (JC-1) levels and decreased Mito-ROS production. NaB increases ZO-1 and Occludin expression in CaCo2 cells by inhibiting RAW264.7 pyroptosis. These results suggest that NaB could be utilized as a therapeutic agent or dietary supplement to alleviate colitis.
Collapse
Affiliation(s)
- Guoqiang Fan
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yaxin Liu
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Limei Tao
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Danping Wang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yizhu Huang
- Singao Xiamen Company, Xiamen 361006, PR China
| | - Xiaojing Yang
- Key Laboratory of Animal Physiology & Biochemistry, Nanjing Agricultural University, Nanjing 210095, PR China; MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
10
|
Kundu S, Das S, Maitra P, Halder P, Koley H, Mukhopadhyay AK, Miyoshi SI, Dutta S, Chatterjee NS, Bhattacharya S. Sodium butyrate inhibits the expression of virulence factors in Vibrio cholerae by targeting ToxT protein. mSphere 2025; 10:e0082424. [PMID: 40261078 DOI: 10.1128/msphere.00824-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 03/21/2025] [Indexed: 04/24/2025] Open
Abstract
Cholera, a diarrheal disease caused by the gram-negative bacterium Vibrio cholerae, remains a global health threat in developing countries due to its high transmissibility and increased antibiotic resistance. There is a pressing need for alternative strategies, with an emphasis on anti-virulent approaches to alter the outcome of bacterial infections, given the increase in antimicrobial-resistant strains. V. cholerae causes cholera by secreting virulence factors in the intestinal epithelial cells. These virulence factors facilitate bacterial colonization and cholera toxin production during infection. Here, we demonstrate that sodium butyrate (SB), a small molecule, had no effect on bacterial viability but was effective in suppressing the virulence attributes of V. cholerae. The production of cholera toxin (CT) was significantly reduced in a standard V. cholerae El Tor strain and two clinical isolates when grown in the presence of SB. Analysis of mRNA and protein levels further revealed that SB reduced the expression of the ToxT-dependent virulence genes like tcpA and ctxAB. DNA-protein interaction assays, conducted at cellular (ChIP) and in vitro conditions (EMSA), indicated that SB weakens the binding between ToxT and its downstream promoter DNA, likely by blocking DNA binding. Furthermore, the anti-virulence efficacy of SB was confirmed in animal models. These findings suggest that SB could be developed as an anti-virulence agent against V. cholerae, serving as a potential alternative to conventional antibiotics or as an adjunctive therapy to combat cholera. IMPORTANCE The world has been facing an upsurge in cholera cases since 2021, a similar trend continuing into 2022, with over 29 countries reporting cholera outbreaks (World Health Organization, 16 December 2022, Disease Outbreak News, Cholera-global situation). Treatment of cholera involves oral rehydration therapy coupled with antibiotics to reduce the duration of the illness. However, in recent years, indiscriminate use of antibiotics has contributed to the emergence of antibiotic-resistant strains. In this study, we have addressed the problem of antibiotic resistance by targeting virulence factors. Screening various compounds using in silico methods led to the identification of a small molecule, SB, that inhibits the virulence cascade in V. cholerae. We demonstrated that (i) SB intervened in ToxT protein-DNA binding and subsequently affected the expression of ToxT-regulated virulence genes (ctxAB and tcpA) and (ii) SB is a potential therapeutic candidate for the development of a novel antimicrobial agent.
Collapse
Affiliation(s)
- Sushmita Kundu
- Division of Biochemistry, ICMR-National Institute for Research in Bacterial Infections (Formerly ICMR-National Institute of Cholera and Enteric Diseases), Kolkata, India
| | - Suman Das
- Division of Biochemistry, ICMR-National Institute for Research in Bacterial Infections (Formerly ICMR-National Institute of Cholera and Enteric Diseases), Kolkata, India
| | - Priyanka Maitra
- Division of Biochemistry, ICMR-National Institute for Research in Bacterial Infections (Formerly ICMR-National Institute of Cholera and Enteric Diseases), Kolkata, India
| | - Prolay Halder
- Division of Bacteriology, ICMR-National Institute for Research in Bacterial Infections (Formerly ICMR-National Institute of Cholera and Enteric Diseases), Kolkata, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute for Research in Bacterial Infections (Formerly ICMR-National Institute of Cholera and Enteric Diseases), Kolkata, India
| | - Asish K Mukhopadhyay
- Division of Bacteriology, ICMR-National Institute for Research in Bacterial Infections (Formerly ICMR-National Institute of Cholera and Enteric Diseases), Kolkata, India
| | - Shin-Ichi Miyoshi
- Division of Pharmaceutical Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Shanta Dutta
- Division of Bacteriology, ICMR-National Institute for Research in Bacterial Infections (Formerly ICMR-National Institute of Cholera and Enteric Diseases), Kolkata, India
| | - Nabendu Sekhar Chatterjee
- Division of Biochemistry, ICMR-National Institute for Research in Bacterial Infections (Formerly ICMR-National Institute of Cholera and Enteric Diseases), Kolkata, India
| | - Sushmita Bhattacharya
- Division of Biochemistry, ICMR-National Institute for Research in Bacterial Infections (Formerly ICMR-National Institute of Cholera and Enteric Diseases), Kolkata, India
| |
Collapse
|
11
|
Ding Y, Fernández-Montero A, Mani A, Casadei E, Miyazawa R, Zhou C, Chaumont L, Posavi M, Cole SD, Shibasaki Y, Takizawa F, Salinas I, Sunyer JO. Secretory IgM regulates gut microbiota homeostasis and metabolism. Nat Microbiol 2025:10.1038/s41564-025-02013-8. [PMID: 40410336 DOI: 10.1038/s41564-025-02013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 04/11/2025] [Indexed: 05/25/2025]
Abstract
The coating of microbiota by secretory immunoglobulins (sIgs) determines which bacteria colonize the gut and influences bacterial metabolism. Previous work has identified sIgA and sIgT as mediators of gut homeostasis. However, sIgM coats a large proportion of the gut microbiota in humans and teleost fish, thus suggesting a conserved role of sIgM in microbiota homeostasis. Here, to investigate this hypothesis, we used the teleost rainbow trout as a model system. Depletion of IgM from trout resulted in severe microbiota-dependent gut tissue damage, body weight loss, bacterial translocation and gut dysbiosis. IgM depletion led also to alterations in microbiota-derived metabolites, including short-chain fatty acids and essential amino acids. Supporting a protective role for sIgM in the gut, high mortality of IgM-depleted fish occurred in an experimental colitis model as a result of severe systemic bacteraemia and septic shock. Our findings uncover sIgM as a previously unrecognized regulator of microbiota homeostasis and metabolism.
Collapse
Affiliation(s)
- Yang Ding
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alvaro Fernández-Montero
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amir Mani
- Center for Evolutionary and Theoretical Immunology (CETI), Department of Biology, University of New Mexico, Albuquerque, NM, USA
| | - Elisa Casadei
- Center for Evolutionary and Theoretical Immunology (CETI), Department of Biology, University of New Mexico, Albuquerque, NM, USA
- Integrative Biology Department, Oklahoma State University (OSU-Stillwater), Stillwater, OK, USA
| | - Ryuichiro Miyazawa
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Congjin Zhou
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lise Chaumont
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marijan Posavi
- Center for Evolutionary and Theoretical Immunology (CETI), Department of Biology, University of New Mexico, Albuquerque, NM, USA
| | - Stephen D Cole
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yasuhiro Shibasaki
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Marine Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Fumio Takizawa
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Faculty of Marine Science and Technology, Fukui Prefectural University, Obama, Japan
| | - Irene Salinas
- Center for Evolutionary and Theoretical Immunology (CETI), Department of Biology, University of New Mexico, Albuquerque, NM, USA.
| | - J Oriol Sunyer
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Xu W, Wang L, Chen R, Liu Y, Chen W. Pyroptosis and its role in intestinal ischemia-reperfusion injury: a potential therapeutic target. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04261-1. [PMID: 40372474 DOI: 10.1007/s00210-025-04261-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025]
Abstract
Intestinal ischemia-reperfusion injury (II/RI) is a critical acute condition characterized by complex pathological mechanisms, including various modes of cell death. Among these, pyroptosis has garnered significant attention in recent years. This review explores the characteristics, molecular mechanisms, and implications of pyroptosis in II/RI, with a focus on therapeutic strategies targeting the pyroptosis pathway. Key processes such as NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation, caspase-1 activation, and gasdermin D (GSDMD)-mediated membrane pore formation are identified as central to pyroptosis. Compounds like MCC950, CY-09, metformin, and curcumin have shown promise in attenuating II/RI in preclinical studies by modulating these pathways. However, challenges remain in understanding non-canonical pyroptosis pathways, unraveling the exact mechanisms of GSDMD-induced pore formation, and translating these findings into clinical applications. Addressing these gaps will be crucial for developing innovative and effective treatments for II/RI.
Collapse
Affiliation(s)
- Wenping Xu
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Lang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Ruili Chen
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Yi Liu
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Wendong Chen
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China.
| |
Collapse
|
13
|
Zeng X, Yu P, Li D, Li Y, Wang X, Yang X, Ren D. Structural characterization and alleviative effects of novel polysaccharides from Artemisia sphaerocephala Krasch seed on obese mice by regulating gut microbiota. Int J Biol Macromol 2025; 310:143407. [PMID: 40274139 DOI: 10.1016/j.ijbiomac.2025.143407] [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: 04/07/2025] [Accepted: 04/20/2025] [Indexed: 04/26/2025]
Abstract
This study aimed to investigate the efficacy of polysaccharides from Artemisia sphaerocephala Krasch (ASK) seed in alleviating high fat diet (HFD) caused obesity. Here, three polysaccharide fractions (ASKP1, ASKP2 and ASKP3) were purified from ASK seed. Chemical characteristic analysis revealed that ASKP1 is a neutral heteropolysaccharide with the average molecular weight of 9.08 × 105 Da, while ASKP2 and ASKP3 are acidic heteropolysaccharides with the molecular weight of 9.39 × 105 and 8.41 × 105 Da, respectively. Animal experiment found that three ASKP fractions obviously relieved obesity and related metabolic disorders induced by HFD, while ASKP1 was more effective in reducing the blood glucose and serum LDL levels. 16S rDNA sequencing showed that ASKP fractions improved the gut microbiota imbalance of obese mice, and ASKP1 promoted the proliferation of beneficial bacterium Akkermansia more effectively than ASKP2 and ASKP3. Furthermore, ASKP fractions facilitated thermogenesis of brown adipose tissue (BAT) of obese mice, as evidenced by increased expression of thermogenic marker genes UCP1 in BAT, and the thermogenesis effect of ASKP1 was the most obvious. Taken together, our results show that ASKP1 is a novel prebiotic that may be used to treat obesity and its related abnormal metabolism.
Collapse
Affiliation(s)
- Xiaoqian Zeng
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Pinglian Yu
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China; Key Laboratory of YunNan University for Plateau Characteristic Functional Food, School of Chemistry and Chemical Engineering, Zhaotong University, 657000, China.
| | - Donglu Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yixiao Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xuejie Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Daoyuan Ren
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
14
|
Ma J, Li T, Lin L, Du C, Wei C, Yin F, Sun X, Lyu G, Gan S. The expression pattern of butyric acid transporter in the large intestine with growth and development of suckling lambs. Anim Biosci 2025; 38:968-980. [PMID: 39901713 PMCID: PMC12062808 DOI: 10.5713/ab.24.0490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/27/2024] [Accepted: 11/12/2024] [Indexed: 02/05/2025] Open
Abstract
OBJECTIVE The objective of this research was to explore the changes of morphological parameters, short chain fatty acid contents and butyric acid transporter expression levels in the large intestine with growth and development of sucking lambs. METHODS A total of 48 newborn male Hu sheep lambs (body weight = 2.94±0.22 kg) were selected in this experiment. At 0, 7, 14, 28 and 42 days of ages, 6 lambs were slaughtered to collect cecal and colonic samples to analyze morphological parameters, short chain fatty acid contents and butyric acid transporter mRNA expression. RESULTS The organ index of the cecum in d 0, 28 and 42 groups was higher (p<0.05) than that in d 7 and 14 groups. Compared with other age groups, the organ index of the colon was significantly increased (p<0.05) in d 42 group. After 7 days of age, the contents of acetic, propionic and butyric acids in the cecum and colon were significantly increased (p<0.05) as age increased. A similar trend of mRNA expressions of butyric acid transporters, including monocarboxylate transporter, sodium-coupled monocarboxylate transporter, sodium-hydrogen exchanger, down regulated in adenoma, putative anion transporter-1 and anion exchanger-2, was found. In the cecum, the Escherichia coli count in d 14 group was higher (p<0.05) than that in other age groups, whereas an opposite tendency was found in Lactobacillus count between d 14 and other groups. Additionally, in the cecum and colon, the relative expression of claudin-1 in d 14 group was lower (p<0.05) than in d 42 group. CONCLUSION Overall, the current results indicate that the expression levels of butyric acid transporters in the cecum and colon of lambs are significantly influenced by days of age.
Collapse
Affiliation(s)
- Jian Ma
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang,
China
| | - Tao Li
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang,
China
| | - Lu Lin
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang,
China
| | - Chunmei Du
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang,
China
| | - Chen Wei
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang,
China
| | - Fuquan Yin
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang,
China
| | - Xuemei Sun
- Xinjiang Taikun Group Co. Ltd., Changji,
China
| | - Gang Lyu
- Xinjiang Taikun Group Co. Ltd., Changji,
China
| | - Shangquan Gan
- College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang,
China
| |
Collapse
|
15
|
Arioglu‐Tuncil S. A Comparative Assessment of Flaxseed ( Linum usitatissimum L.) and Chia Seed ( Salvia hispanica L.) in Modulating Fecal Microbiota Composition and Function In Vitro. Food Sci Nutr 2025; 13:e70243. [PMID: 40321609 PMCID: PMC12048700 DOI: 10.1002/fsn3.70243] [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: 03/01/2025] [Revised: 04/13/2025] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
Flaxseed (Linum usitatissimum L.) and chia seed (Salvia hispanica L.) have become increasingly popular in the design of various functional food products. However, information on their functional properties is scarce. The aim of this study is to comparatively evaluate the effects of the dietary fibers (DFs) of flaxseed and chia seed on colonic microbiota composition and metabolic outputs. The neutral and acidic monosaccharide compositions of DFs of flaxseed and chia seed were determined using gas chromatography/mass spectrometry (GC/MS) and spectrophotometer, respectively. Next, in vitro fecal fermentation assays were applied, and samples were collected at different time points for short-chain fatty acids (SCFA) measurements using GC, and fecal microbiota changes before and after fermentation were evaluated through 16S rRNA sequencing. The results revealed that DFs of flaxseed were dominated by xylose and uronic acid moieties, while that of chia seed was dominated by glucose units, indicating that their DFs are structurally different. Higher SCFA generations were observed in the case of flaxseed, suggesting that flaxseed DFs are more readily fermentable by gut microbiota. Flaxseed and chia seed DFs differentially impacted the microbiota compositions at the OTU level; for example, significant increases in the relative abundances of Acidaminococcaceae and Bacteroides stercoris related OTUs, which are known to be propionate producers, were observed in the case of flaxseed, but not chia seed. Interestingly, flaxseed, but not chia seed, DFs suppressed the growth of some pathogenic bacteria. Overall, this study suggests that the functionality of flaxseed and chia seed DFs in relation to colonic microbiota may differ, with flaxseed being more readily fermented and potentially promoting beneficial microbes to a greater extent. Thus, flaxseed could hold promise for developing functional food recipes aimed at supporting colonic health.
Collapse
Affiliation(s)
- Seda Arioglu‐Tuncil
- Department of Nutrition and DieteticsNecmettin Erbakan UniversityKonyaTürkiye
| |
Collapse
|
16
|
Vella VR, Ainsworth-Cruickshank G, Luft C, Wong KE, Parfrey LW, Vogl AW, Holman PJ, Bodnar TS, Raineki C. Dysregulation of immune system markers, gut microbiota and short-chain fatty acid production following prenatal alcohol exposure: A developmental perspective. Neurochem Int 2025; 185:105952. [PMID: 39988283 DOI: 10.1016/j.neuint.2025.105952] [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: 11/04/2024] [Revised: 01/25/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Prenatal alcohol exposure (PAE) can severely impact fetal development, including alterations to the developing immune system. Immune perturbations, in tandem with gut dysbiosis, have been linked to brain and behavioral dysfunction, but this relationship is poorly understood in the context of PAE. This study takes an ontogenetic approach to evaluate PAE-induced alterations to brain and serum cytokine levels and both the composition and metabolic output of the gut microbiota. Using a well-established rat model of PAE, cytokine levels in the serum, prefrontal cortex, amygdala, and hypothalamus as well as gut microbiota composition and short-chain fatty acid (SCFA) levels were assessed at three postnatal (P) timepoints: P8 (infancy), P22 (weaning), and P38 (adolescence). Male PAE rats had increased cytokine levels in the amygdala and hypothalamus, but not prefrontal cortex, at P8. This altered neuroimmune function was not seen in the PAE females. The effect of PAE on central cytokine levels was reduced at P22/38, the same age at which PAE-induced alterations in serum cytokine levels emerge in both sexes. PAE reduced bacterial diversity in both sexes at P8, but only in females at P38, where a PAE-induced unique community composition emerged. Both sexes had alterations to specific bacterial taxa (e.g., Firmicutes), some of which are important in producing the SCFA butyric acid, which was decreased in PAE animals at P22. These results demonstrate that PAE leads to sex- and age-specific alterations in immune function, gut microbiota and SCFA production, highlighting the need to consider both age and sex in future work.
Collapse
Affiliation(s)
- Victoria R Vella
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | | | - Carolina Luft
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | - Kingston E Wong
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | - Laura W Parfrey
- Department of Botany, University of British Columbia, British Columbia, Canada
| | - A Wayne Vogl
- Life Sciences Centre, Department of Cellular and Physiological Sciences, University of British Columbia, British Columbia, Canada
| | - Parker J Holman
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada
| | - Tamara S Bodnar
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Charlis Raineki
- Department of Psychology, Brock University, St. Catharines, Ontario, Canada.
| |
Collapse
|
17
|
Onuma M, Ataka K, Murakami A. Evaluating the safety and functionality of a novel compound containing prebiotics, probiotics, and postbiotics in healthy cats and dogs. Open Vet J 2025; 15:1969-1981. [PMID: 40557076 PMCID: PMC12184448 DOI: 10.5455/ovj.2025.v15.i5.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/06/2025] [Accepted: 04/19/2025] [Indexed: 06/28/2025] Open
Abstract
Background Although various prebiotics, probiotics, and postbiotics are available, their safety and efficacy in combination are unknown. Aim We investigated the safety and functionality of a newly developed supplement, previously unreported in pet animals, containing 26 types of biotic material bacteria (2 prebiotics, 1 probiotic, and 23 postbiotics) in cats and dogs. The biotic materials included were selected based on current evidence from cats and dogs. Methods A new supplement developed using species tested in cats and dogs was administered. One-way analysis was used for data obtained from 3 cats (7 days of treatment and 7 days of nontreatment), and a parallel, controlled study was performed in 20 dogs (n = 10 each in control and test groups, for 27 days). Results In cats, no abnormal values were observed in complete blood count or blood chemistry tests, whereas significant decreases in blood glucose and total cholesterol were confirmed (p < 0.05 each). In the feline lymphocyte subset test, significant increases were observed in T and B cells (p < 0.05). A significant difference in fecal pH was observed in the test group (p < 0.01). In addition, 60% (9/15) of the test group had an increase in total organic acids. In dogs, only indole showed a consistent decrease among putrefactive products (p = 0.055). Regarding analyses of intestinal flora from feces using a gene sequencer at the genus level, no changes were observed in cats. Conversely, Lachnospira and Anaeroplasma genera tended to be decreased in the control group but increased by 23.1% and 45%, respectively, in the test group. In addition, Escherichia-Shigella and Tyzzerella genera showed slight increases or changes in the control group but significant decreases in the test group. Regarding the Firmicutes/Bacteroidetes ratio, an increase in the control group and a decrease in the test group were observed in all cats, whereas no differences were observed in dogs. Conclusion The supplement is safe for both cats and dogs. Results of comprehensive analyses suggested that the supplement improved the intestinal environment by regulating the gastrointestinal microbiota.
Collapse
Affiliation(s)
- Mamoru Onuma
- Oosagami Animal Clinic, Koshigaya-shi, Japan
- Department of Animal Risk Management, Faculty of Risk and Crisis Management, Chiba Institute of Science, Choshi, Japan
| | | | | |
Collapse
|
18
|
Han R, Wang Z, Li Y, Ke L, Li X, Li C, Tian Z, Liu X. Gut microbiota Lactobacillus johnsonii alleviates hyperuricemia by modulating intestinal urate and gut microbiota-derived butyrate. Chin Med J (Engl) 2025:00029330-990000000-01534. [PMID: 40304365 DOI: 10.1097/cm9.0000000000003603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Gut microbiota are important for uric acid (UA) metabolism within hyperuricemia (HUA); however, the underlying mechanisms of how the gut microbiota regulate intestinal UA metabolism remain unclear. This study aimed to explore the function of the intestine in HUA and to further reveal the possible mechanism. METHODS We conducted gut microbiota depletion to validate the role of gut microbiota in UA metabolism. A mouse model of HUA was established, and the gut microbiota and microbiome-derived metabolites were analyzed via 16S RNA gene sequencing and metabolomics analysis. The mechanism of the gut microbiota in HUA was elucidated by in vivo and in vitro experiments. RESULTS Antibiotic treatment elevated serum UA, disturbed purine metabolism, and decreased the relative abundance of Lactobacillus. HUA mice had a lower relative abundance of Lactobacillus johnsonii (L. johnsonii) and decreased gut butyrate concentration. Supplementation of L. johnsonii significantly reduces serum UA in hyperuricemia mice by preventing UA synthesis and promoting the excretion of gut purine metabolites. In addition, L. johnsonii enhanced intestinal UA excretion by heightening the urate transporter ABCG2 (adenosine triphosphate-binding cassette transporter, subfamily G, member 2) expression, and increasing the levels of butyrate, which upregulated ABCG2 expression via the Wnt5a/b/β-catenin signaling pathway. CONCLUSION Our results suggest that gut microbiota and microbiota-derived metabolites directly regulate gut UA metabolism, highlighting potential applications in the treatment of diet-induced HUA by targeting gut microbiota and its metabolites.
Collapse
Affiliation(s)
- Rongshuang Han
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Zan Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Yukun Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Leyong Ke
- Department of Gastroenterology, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, China
| | - Xiang Li
- Department of Gastroenterology, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, China
| | - Changgui Li
- Institute of Metabolic Diseases, Qingdao University, Qingdao, Shandong 266003, China
| | - Zibin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Xin Liu
- Department of Gastroenterology, Zigong Fourth People's Hospital, Zigong, Sichuan 643000, China
| |
Collapse
|
19
|
Cheng X, Liang Y, Ji K, Feng M, Du X, Jiao D, Wu X, Zhong C, Cong H, Yang G. Enhanced propionate and butyrate metabolism in cecal microbiota contributes to cold-stress adaptation in sheep. MICROBIOME 2025; 13:103. [PMID: 40275300 PMCID: PMC12023611 DOI: 10.1186/s40168-025-02096-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 03/17/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND During cold stress, gut microbes play crucial roles in orchestrating energy metabolism to enhance environmental adaptation. In sheep, hindgut microbes ferment carbohydrates to generate short-chain fatty acids (SCFAs) as an energy source. However, the mechanisms by which hindgut microbes and their metabolites interact with the host to facilitate adaptation to cold environments remain ambiguous. Herein, we simulated a winter environment (- 20 °C) and provided a rationed diet to compare the cold adaptation mechanisms between Hulunbuir and Hu sheep. RESULTS Our findings show that cold exposure enhances SCFA metabolism in the sheep cecum. In Hu sheep, acetate, butyrate, and total SCFA concentrations increased, whereas in Hulunbuir sheep, propionate and butyrate concentrations increased, with a notable increase in total SCFAs. Notably, butyrate concentration was higher in Hulunbuir sheep than in Hu sheep under cold stress. Following cold exposure, the proinflammatory cytokine IL-1β levels increased in both breeds. In addition, Hu sheep showed increased IL-10, whereas Hulunbuir sheep exhibited elevated secretory IgA levels. The cecal microbiota responded differently, Hu sheep showed no notable changes in alpha and beta diversity, whereas Hulunbuir sheep exhibited considerable alterations. In Hu sheep, the abundance of fungi, specifically Blastocystis sp. subtype 4, decreased, and that of several Lachnospiraceae species (Roseburia hominis, Faecalicatena contorta, and Ruminococcus gnavus) involved in SCFA metabolism increased. Pathways related to carbohydrate metabolism, such as starch and sucrose metabolism, galactose metabolism, and pentose and glucuronate interconversions, were upregulated. In Hulunbuir sheep, the abundance of Treponema bryantii, Roseburia sp. 499, and Prevotella copri increased, with upregulation in pathways related to amino acid metabolism and energy metabolism. Cold exposure increased node connectivity within the symbiotic networks of both breeds, with increased network vulnerability in Hu sheep. Following cold exposure, the microbial community of Hulunbuir sheep showed a decrease in the influence of stochastic processes on community assembly, with a corresponding increase in the role of environmental selection. Conversely, no such shift was evident in Hu sheep. Further transcriptomic analysis revealed distinct regulatory mechanisms between breeds. In Hu sheep, protein synthesis, energy metabolism, and thermogenesis pathways were substantially upregulated. By contrast, Hulunbuir sheep showed considerable upregulation of immune pathways and energy conservation through reduced ribosome synthesis. Correlation analysis indicated that butyrate holds a central position in both networks, with Hulunbuir sheep exhibiting a more complex and tightly regulated network involving SCFAs, microbiota, microbial functions, and transcriptomes. Partial least squares path modeling revealed that cold exposure substantially altered the cecal microbiota and transcriptomes of Hulunbuir sheep, affecting SCFAs and cytokines. CONCLUSIONS The findings of this study suggest that under cold exposure, Hu sheep enhance acetate fermentation and rely on tissue thermogenesis for adaptation. By contrast, Hulunbuir sheep exhibit changes in microbial diversity and function, leading to increased propionate and butyrate metabolism. This may promote physiological energy conservation and innate immune defense, balancing heat loss and enhancing cold adaptation.
Collapse
Affiliation(s)
- Xindong Cheng
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanping Liang
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kaixi Ji
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute of Animal Husbandry and Veterinary Medicine, Shandong Academy of Agricultural Sciences/Shandong Key Laboratory of Animal Microecologics and Efficient Breeding of Livestock and Poultry, Jinan, 250100, China
| | - Mengyu Feng
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xia Du
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dan Jiao
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiukun Wu
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China
- Key Laboratory of Extreme Environmental Microbial Resources and Engineering, Lanzhou, 730000, China
| | - Chongyue Zhong
- Dongying Animal Husbandry and Veterinary Station, Dongying, 257000, China
| | - Haitao Cong
- Shandong Huakun Rural Revitalization Institute Co., Ltd, Jinan, 250014, China
| | - Guo Yang
- Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Lanzhou Ecological Agriculture Experimental Research Station for Agricultural Ecosystem, Northwest Institute of Eco-Environment and Resources, Chinese Academy of Sciences, Lanzhou, 730000, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
20
|
Miao M, Cheng J, Yan Q, Jiang Z, Yang S. Prebiotic activity comparison of eight oligosaccharides: selection of a potential synbiotic containing konjac manna-oligosaccharides and Bifidobacterium animalis BB-12. Int J Food Sci Nutr 2025:1-11. [PMID: 40264375 DOI: 10.1080/09637486.2025.2494148] [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: 12/02/2024] [Revised: 03/26/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025]
Abstract
Various types of non-digestible oligosaccharides (NDOs) have attracted tremendous interest due to their healthy functions in regulating intestinal microbiota. Whereas the specificity of different NDOs towards certain intestinal bacterial species remains unclear. In this study, konjac manna-oligosaccharides (KMOS) were selected from eight NDOs through in vitro faecal batch fermentation. KMOS accelerated increase of recognised probiotics (Bifidobacterium spp. and Akkermansia spp.) and achieved the highest productions of lactic acid and total short-chain fatty acids (42.0 mM). β-Mannosidase and β-glucosidase played important role in the utilisation of KMOS, and mannobiose and glucosyl-mannobiose were preferentially consumed by faecal microbiota. In pure culture, the utilisation of KMOS was tested with nine Bifidobacterium strains. Amongst, KMOS increased the cell density of B. animalis BB-12 by 3.5 folds and improved its adhesion ability to Caco-2 cell by 3.1 folds, suggesting that KMOS and B. animalis BB-12 may be developed as a potential synbiotic combination.
Collapse
Affiliation(s)
- Miao Miao
- College of Food Science and Nutritional Engineering, Key Laboratory of Food Bioengineering (China National Light Industry), China Agricultural University, Beijing, China
| | - Jiaobo Cheng
- College of Food Science and Nutritional Engineering, Key Laboratory of Food Bioengineering (China National Light Industry), China Agricultural University, Beijing, China
| | - Qiaojuan Yan
- College of Engineering, Bioresource Utilization Laboratory, China Agricultural University, Beijing, China
| | - Zhengqiang Jiang
- College of Food Science and Nutritional Engineering, Key Laboratory of Food Bioengineering (China National Light Industry), China Agricultural University, Beijing, China
| | - Shaoqing Yang
- College of Food Science and Nutritional Engineering, Key Laboratory of Food Bioengineering (China National Light Industry), China Agricultural University, Beijing, China
| |
Collapse
|
21
|
Ma X, Duan C, Wang X, Tao Y, Yang L, Teng Y, Pan Y, Zhang M, Xu J, Sheng J, Wang X, Jin P. Human gut microbiota adaptation to high-altitude exposure: longitudinal analysis over acute and prolonged periods. Microbiol Spectr 2025:e0291624. [PMID: 40257273 DOI: 10.1128/spectrum.02916-24] [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: 12/04/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025] Open
Abstract
This study investigated the longitudinal effects of acute (7-day) and prolonged (3-month) high-altitude exposure on gut microbiota in healthy adult males, addressing the limited data available in human populations. A cohort of 406 healthy adult males was followed, and fecal samples were collected at three time points: baseline at 800 m (406 samples), 7 days after ascending to 4,500 m (406 samples), and 2 weeks post-return to 800 m following 3 months at high altitude (186 samples). High-throughput 16S ribosomal DNA sequencing was employed to analyze microbiota composition and diversity. Results revealed significant changes in alpha- and beta-diversity, with acute high-altitude exposure inducing more pronounced effects compared to prolonged exposure. Specifically, acute exposure increased opportunistic pathogens (Ruminococcus and Oscillibacter) but decreased beneficial short-chain fatty acid producers (Faecalibacterium and Bifidobacterium). Notably, these changes in microbiota persisted even after returning to low altitude, indicating long-term remodeling. Functional analyses revealed substantial changes in metabolic pathways, suggesting microbiota-driven adaptations to energy utilization under high-altitude hypoxic conditions. In summary, acute high-altitude exposure caused dramatic changes in gut microbiota, while prolonged exposure led to structural and functional reshaping. These findings enhance our understanding of how high-altitude environments reshape gut microbiota. IMPORTANCE This study is the first to investigate the impact of high-altitude exposure on gut microbiota adaptation in a large-scale longitudinal cohort. It seeks to enhance understanding of how high-altitude environments reshape gut microbiota. Acute exposure to high altitude significantly affected both α-diversity and β-diversity of gut microbiota, with acute exposure causing more pronounced changes than prolonged adaptation, indicating temporary disruptions in microbial communities. Notable shifts in microbial abundance were observed, including increased levels of genera linked to hypoxic stress (e.g., Gemmiger, Ruminococcus, and Parabacteroides) and decreased levels of beneficial bacteria (e.g., Faecalibacterium, Roseburia, and Bifidobacterium), suggesting possible adverse health effects. Functional analysis indicated changes in metabolism-related pathways post-exposure, supporting the idea that high-altitude adaptations involve metabolic adjustments for energy management. These findings enhance understanding of high-altitude physiology, illustrating the role of gut microbiota in hypoxic health.
Collapse
Affiliation(s)
- Xianzong Ma
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | | | - Xiaoying Wang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yurong Tao
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lang Yang
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yongsheng Teng
- Department of Gastroenterology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Mingjie Zhang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Junfeng Xu
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jianqiu Sheng
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Xin Wang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Peng Jin
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| |
Collapse
|
22
|
Xiao M, Zhou N, Tian Z, Sun C. Endogenous Metabolites in Metabolic Diseases: Pathophysiologic Roles and Therapeutic Implications. J Nutr 2025:S0022-3166(25)00227-5. [PMID: 40250565 DOI: 10.1016/j.tjnut.2025.04.017] [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: 04/05/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
Breakthroughs in metabolomics technology have revealed the direct regulatory role of metabolites in physiology and disease. Recent data have highlighted the bioactive metabolites involved in the etiology and prevention and treatment of metabolic diseases such as obesity, nonalcoholic fatty liver disease, type 2 diabetes mellitus, and atherosclerosis. Numerous studies reveal that endogenous metabolites biosynthesized by host organisms or gut microflora regulate metabolic responses and disorders. Lipids, amino acids, and bile acids, as endogenous metabolic modulators, regulate energy metabolism, insulin sensitivity, and immune response through multiple pathways, such as insulin signaling cascade, chemical modifications, and metabolite-macromolecule interactions. Furthermore, the gut microbial metabolites short-chain fatty acids, as signaling regulators have a variety of beneficial impacts in regulating energy metabolic homeostasis. In this review, we will summarize information about the roles of bioactive metabolites in the pathogenesis of many metabolic diseases. Furthermore, we discuss the potential value of metabolites in the promising preventive and therapeutic perspectives of human metabolic diseases.
Collapse
Affiliation(s)
- Mengjie Xiao
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China
| | - Ning Zhou
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China
| | - Zhen Tian
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China.
| | - Changhao Sun
- National Key Discipline, Department of Nutrition and Food Hygiene, School of Public Health, Harbin Medical University, Harbin, P. R. China; Department of Nutrition and Food Hygiene, School of Public Health, Key Laboratory of Precision Nutrition and Health, Ministry of Education, Harbin Medical University, Heilongjiang, Harbin, P. R. China.
| |
Collapse
|
23
|
Yang K, Li G, Li Q, Wang W, Zhao X, Shao N, Qiu H, Liu J, Xu L, Zhao J. Distribution of gut microbiota across intestinal segments and their impact on human physiological and pathological processes. Cell Biosci 2025; 15:47. [PMID: 40241220 PMCID: PMC12001467 DOI: 10.1186/s13578-025-01385-y] [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: 01/13/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
In recent years, advancements in metagenomics, metabolomics, and single-cell sequencing have enhanced our understanding of the intricate relationships between gut microbiota and their hosts. Gut microbiota colonize humans from birth, with their initial composition significantly influenced by the mode of delivery and feeding method. During the transition from infancy to early childhood, exposure to a diverse diet and the maturation of the immune system lead to the gradual stabilization of gut microbiota's composition and distribution. Numerous studies have demonstrated that gut microbiota can influence a wide range of physiological functions and pathological processes by interacting with various tissues and organs through the gut-organ axis. Different intestinal segments exhibit unique physical and chemical conditions, which leads to the formation of vertical gradients along the intestinal tract: aerobes and facultative aerobes mainly live in the small intestine and anaerobic bacteria mainly live in the large intestine, and horizontal gradients: mucosa-associated microbiota and lumen-associated microbiota. In this review, we systematically summarize the distribution characteristics of gut microbiota across six intestinal segments: duodenum, jejunum, ileum, cecum, colon, and rectum. We also draw a conclusion that gut microbiota distributed in different intestinal segments affect the progression of different diseases. We hope to elucidate the role of microbiota at specific anatomic sites within the gut in precisely regulating the processes of particular diseases, thereby providing a solid foundation for developing novel diagnostic and therapeutic strategies for related diseases.
Collapse
Affiliation(s)
- Ke Yang
- The First Clinical Institute, Zunyi Medical University, Zunyi, 563000, China
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Guangqin Li
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Qihong Li
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Wei Wang
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Xu Zhao
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
- Guizhou University Medical College, Guiyang, 550025, Guizhou, China
| | - Nan Shao
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Hui Qiu
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Jing Liu
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Lin Xu
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
- Department of Immunology, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| | - Juanjuan Zhao
- Key Laboratory for Cancer Prevention and treatment of Guizhou Province, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
- Department of Immunology, Zunyi Medical University, Zunyi, 563000, Guizhou, China.
| |
Collapse
|
24
|
Zhou P, Wu Y, Shen J, Duan T, Che L, Zhang Y, Zhao Y, Yan H. Gestational Inulin Supplementation in Low-/High-Fat Sow Diets: Effects on Growth Performance, Lipid Metabolism, and Meat Quality of Offspring Pigs. Foods 2025; 14:1314. [PMID: 40282717 PMCID: PMC12027208 DOI: 10.3390/foods14081314] [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: 02/22/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
This study investigated whether the supplementation of prebiotic inulin to gestating sows programmatically affects offspring growth performance and meat quality while exploring its epigenetic effects through histone acetylation modulation. After mating, sixty multiparous sows (Landrace × Yorkshire; parity 2-3) were assigned to a 2 × 2 factorial arrangement with inulin (0% vs. 1.5%) and fat (0% or 5%) supplementation until farrowing. Post-weaning, five litters (10 piglets per litter) per treatment were selected and maintained in their original litter for fattening under standardized feeding. The results demonstrated that maternal inulin supplementation during gestation accomplished the following: (1) Increased offspring liver index by 13.4% at weaning and 6.8% at finishing (p < 0.05) while reducing the finishing-phase backfat thickness by 11.6% (p < 0.01), with a significant inulin × fat interaction attenuating fat-induced abdominal lipid accumulation at weaning (p = 0.05). (2) Decreased longissimus dorsi muscle lightness (L*) by 4.5% in finishing pigs (p = 0.02) without altering the other meat quality parameters. (3) Suppressed offspring liver lipid deposition at birth and finishing (p < 0.05), concomitant with upregulated hepatic PGC-1α and CPT1A expression (p < 0.05). (4) Elevated neonatal serum butyrate by 15.6% (p = 0.06) while inhibiting hepatic histone deacetylase (HDAC) activity and enhancing histone H3/H4 acetylation (p < 0.01). These findings suggest that maternal inulin supplementation during gestation mitigates offspring hepatic lipid deposition through butyrate-mediated epigenetic regulation, where microbial-derived butyrate from inulin fermentation inhibits HDAC activity, enhances histone acetylation levels, and upregulates fatty acid β-oxidation gene expression. This study provides novel mechanistic insights into how maternal dietary fiber nutrition programs offspring development through epigenetic reprogramming.
Collapse
Affiliation(s)
- Pan Zhou
- School of Life Science and Agro-Forestry, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang 621010, China; (P.Z.); (Y.W.); (J.S.); (T.D.); (Y.Z.)
| | - Yachao Wu
- School of Life Science and Agro-Forestry, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang 621010, China; (P.Z.); (Y.W.); (J.S.); (T.D.); (Y.Z.)
| | - Jianbo Shen
- School of Life Science and Agro-Forestry, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang 621010, China; (P.Z.); (Y.W.); (J.S.); (T.D.); (Y.Z.)
| | - Tao Duan
- School of Life Science and Agro-Forestry, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang 621010, China; (P.Z.); (Y.W.); (J.S.); (T.D.); (Y.Z.)
| | - Long Che
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, No. 6 North Longzihu Road, Zhengdong New District, Zhengzhou 450046, China;
| | - Yong Zhang
- School of Life Science and Agro-Forestry, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang 621010, China; (P.Z.); (Y.W.); (J.S.); (T.D.); (Y.Z.)
| | - Yang Zhao
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Animal Science Academy, Chengdu 610066, China
| | - Honglin Yan
- School of Life Science and Agro-Forestry, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang 621010, China; (P.Z.); (Y.W.); (J.S.); (T.D.); (Y.Z.)
| |
Collapse
|
25
|
Sun H, Du Z, Fan L, Xu Z, Yang W, Zhang G, Liu X. Structural alterations in butyrylated and recrystallized high-amylose maize starch and their effect on gut microbiota during in vitro fermentation. Int J Biol Macromol 2025; 302:139970. [PMID: 39826737 DOI: 10.1016/j.ijbiomac.2025.139970] [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: 09/09/2024] [Revised: 01/05/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
In this study, type-3 resistant starch (RS) with enhanced thermal stability and excellent short-chain fatty acid (SCFA) production was obtained through the butyrylation and subsequent recrystallization at 4 °C of high-amylose maize starch (HAMS). We comprehensively examined and contrasted the structural attributes and in vitro human fecal fermentation behavior of butyrylated RS (BRS) with varying degrees of substitution. Fourier-transform infrared analysis validated the successful integration of carbonyl groups into the starch matrix. This phenomenon was evident through the characteristic peaks at 1727 cm-1. X-ray diffraction and thermal stability analyses delineated the distinct B-type crystalline structure and high relative crystallinity of 25.31 % and 22.23 % of the 10 % and 15 % butyric anhydride BRS-10 and BRS-15, respectively, which differed from that of HAMS. Their second peak gelatinization temperature values reached 95.6 °C and 92.6 °C. The in vitro fermentation of BRS fostered SCFA production, boosting the relative abundance of beneficial bacteria (e.g., Ligilactobacillus and Roseburia). Meanwhile, the extensively modified BRS favored the propagation of Faecalibacterium and Bifidobacterium, maintaining intestinal microbiota advantage. These findings underscore the significant effects of butyrylation modification on fermentation kinetics, metabolite profiles, and gut microbiota composition, providing invaluable insights into the development of functional food products that aim to bolster colonic health.
Collapse
Affiliation(s)
- Hui Sun
- College of Tea and Food Science, Wuyi University, Fujian 354300, PR China
| | - Zhongda Du
- College of Tea and Food Science, Wuyi University, Fujian 354300, PR China
| | - Li Fan
- College of Tea and Food Science, Wuyi University, Fujian 354300, PR China
| | - Zhenyi Xu
- College of Tea and Food Science, Wuyi University, Fujian 354300, PR China
| | - Weisen Yang
- College of Tea and Food Science, Wuyi University, Fujian 354300, PR China
| | - Guoshou Zhang
- College of Tea and Food Science, Wuyi University, Fujian 354300, PR China
| | - Xiong Liu
- College of Food Science, Southwest University, Tiansheng Road 2, Chongqing, 400715, PR China.
| |
Collapse
|
26
|
Ogory RO, Cumberford G, Adewole D. Ahiflower seed and its press cake as sources of nutrients for laying hens and omega-3 fatty acids in their eggs. Poult Sci 2025; 104:104936. [PMID: 40058003 PMCID: PMC11930598 DOI: 10.1016/j.psj.2025.104936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/15/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
240 64-week-old Lohman LSL-Lite laying hens were used to evaluate the effect of ahiflower seed (AS) and its press cake (APC) on egg yolk fatty acid profile, production performance, apparent total tract nutrient digestibility (ATTD), egg quality, eggshell mineral content, and fecal microbiota composition for 12 weeks in a completely randomized design, with 6 replicates of 5 birds in a cage. The diets included a control (CD), CD supplemented with 10 % flaxseed (FS), and CD supplemented with AS at 1, 5, and 10 % inclusion levels and APC at 5, 10, and 15 % inclusion levels. Diet did not affect eggshell Ca (P=0.1168) and P (P=0.8212) levels, and feed conversion ratio (P=0.136), but the 10 % FS reduced body weight gain (P=0.044), hen day egg production (P= 0.000) and feed intake (P<.0001) compared to other treatments. The yolk lightness L* was reduced (P=0.030) by 5 % APC compared to 10 % APC, redness a* was reduced (P= 0.002) by 10 % FS and 15 %APC compared to 10 %APC, CD, and 1 % AS. The 10 % FS and 15 %APC also reduced (P<0.001) yellowness *b compared to 1 %AS and 5 %APC. Apparent metabolizable energy (AME) and nitrogen-corrected apparent metabolizable energy (AMEn) increased (P<0.001) in 10 %FS and all AS and APC levels compared to CD. Compared to CD (87 %), ATTD of energy was increased (P<0.001) in hens fed 10 %FS (93 %), 1 %AS (93 %), and 15 %APC (92 %). However, 10 %FS (78.7 %) and 1 %AS (81.7 %) had higher (P=0.011) ATTD of P than 10 %APC (64.6 %). Similarly, ATTD of Ca was reduced (P<0.001) in hens fed 10 %APC compared to CD and 10 %AS. Compared to other treatments, total n-3 and stearidonic acids were increased (P<0.001) by 10 %FS and 10 %AS, respectively, and the total n-6 FAs and linoleic acid were highest (P=0.001) in 15 %APC. Both 10 %AS and 10 %FS increased (P<0.001) eicosapentaenoic, docosahexaenoic, and alpha-linolenic acid, compared to CD. The n-6/n-3 ratio was reduced (P<0.001) by 10 %FS and 10 %AS compared to APC and CD. Dietary treatments modulated fecal microbiota differently, but notably, Lactobacillus was more abundant when hens were fed 5 %AS compared to other treatments. In conclusion, the dietary supplementation of 10 %AS increased n3-FAs deposition in eggs similar to 10 %FS. However, 10 %FS reduced production performance. All levels of AS and APC increased diet metabolizable energy with no negative effect on production performance.
Collapse
Affiliation(s)
- Roseline O Ogory
- Department of Animal and Poultry Science, College of Agriculture and Bioresources, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada
| | - Greg Cumberford
- Natures Crops International, 12682 Route 6, PO Box 248, Kensington, PE C0B 1M0, Canada
| | - Deborah Adewole
- Department of Animal and Poultry Science, College of Agriculture and Bioresources, University of Saskatchewan, Saskatoon, SK S7N 5A8, Canada.
| |
Collapse
|
27
|
Luo Z, Ou H, Tan Z, Jiao J. Rumen-protected methionine and lysine supplementation to the low protein diet improves animal growth through modulating colonic microbiome in lambs. J Anim Sci Biotechnol 2025; 16:46. [PMID: 40102971 PMCID: PMC11917156 DOI: 10.1186/s40104-025-01183-z] [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: 11/14/2024] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Dietary protein level and amino acid (AA) balance are crucial determinants of animal health and productivity. Supplementing rumen-protected AAs in low-protein diets was considered as an efficient strategy to improve the growth performance of ruminants. The colon serves as a crucial conduit for nutrient metabolism during rumen-protected methionine (RPMet) and rumen-protected lysine (RPLys) supplementation, however, it has been challenging to clarify which specific microbiota and their metabolites play a pivotal role in this process. Here, we applied metagenomic and metabolomic approaches to compare the characteristic microbiome and metabolic strategies in the colon of lambs fed a control diet (CON), a low-protein diet (LP) or a LP diet supplemented with RPMet and RPLys (LR). RESULTS The LP treatment decreased the average daily weight gain (ADG) in lambs, while the LR treatment tended to elicit a remission in ADG. The butyrate molar concentration was greater (P < 0.05), while acetate molar concentration (P < 0.05) was lower for lambs fed the LP and LR diets compared to those fed the CON diet. Moreover, the LP treatment remarkably decreased total AA concentration (P < 0.05), while LR treatment showed an improvement in the concentrations of methionine, lysine, leucine, glutamate, and tryptophan. Metagenomic insights proved that the microbial metabolic potentials referring to biosynthesis of volatile fatty acids (VFAs) and AAs in the colon were remarkably altered by three dietary treatments. Metagenomic binning identified distinct microbial markers for the CON group (Alistipes spp., Phocaeicola spp., and Ruminococcus spp.), LP group (Fibrobacter spp., Prevotella spp., Ruminococcus spp., and Escherichia coli), and LR group (Akkermansia muciniphila and RUG099 spp.). CONCLUSIONS Our findings suggest that RPMet and RPLys supplementation to the low-protein diet could enhance the microbial biosynthesis of butyrate and amino acids, enriche the beneficial bacteria in the colon, and thereby improve the growth performance of lambs.
Collapse
Affiliation(s)
- Zhibin Luo
- State Key Laboratory of Forage Breeding-by-Design and Utilization, CAS Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China
| | - Huimin Ou
- State Key Laboratory of Forage Breeding-by-Design and Utilization, CAS Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China
| | - Zhiliang Tan
- State Key Laboratory of Forage Breeding-by-Design and Utilization, CAS Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China
| | - Jinzhen Jiao
- State Key Laboratory of Forage Breeding-by-Design and Utilization, CAS Key Laboratory of Agroecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, 410125, China.
| |
Collapse
|
28
|
Gao H, Wang Y, Zhao X, Yu Y, Guo Y, Li Z, Zhou Z. Growth Performance and Gut Health of Cold-Stressed Broilers in Response to Supplementation with a Combination of Sodium Butyrate and Vitamin D3. Animals (Basel) 2025; 15:861. [PMID: 40150390 PMCID: PMC11939318 DOI: 10.3390/ani15060861] [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: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
The current experiment aimed to investigate the effects of sodium butyrate (SB) and vitamin D3 (VD3) supplementation on the growth performance, immune status, antioxidant capacity, and gut health of young broilers under cold stress. A total of 144 1-day-old Arbor Acres chicks were randomly allotted to three treatments with 6 replicates of 8 birds: (1) basal diet; (2) basal diet + cold stress; and (3) basal diet with 1 g/kg SB and 2000 IU/kg VD3 + cold stress. Birds were exposed to cold stress at 16 ± 1 °C for 72 h (d 18-21) and 26 ± 1 °C for the control. The results indicated that the SB/VD3 diet could alleviate the reduction in average daily gain (ADG) caused by cold stress (p < 0.05). The SB/VD3 diet decreased the serum endotoxin level and ileal interleukin-1β gene expression and upregulated interleukin-10 and nuclear factor erythroid 2-related factor 2 (Nrf2) gene expression compared with cold-stressed birds (p < 0.05). Furthermore, cold stress altered the composition of gut microbiota, including a decrease in Clostridium_sensu_stricto_1, whereas the SB/VD3 diet prevented the reduction. In conclusion, the SB/VD3 diet mitigated the negative effects of cold stress on growth performance and the intestines by strengthening intestinal barrier function and stabilizing gut microbiota balance in broiler chicks, and these results can help to manage cold stress.
Collapse
Affiliation(s)
- Hang Gao
- College of Veterinary Medicine, Southwest University, Chongqing 400715, China; (H.G.)
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yi Wang
- College of Veterinary Medicine, Southwest University, Chongqing 400715, China; (H.G.)
| | - Xingkai Zhao
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yaling Yu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Yizhe Guo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhendong Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhenlei Zhou
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
29
|
Wang Y, Li S, Li T, Wu J, Huang Y, Liu W, Ding C, Huang L, Xu X, Wang Y, Gu S, Liu K, Qian K, Sun X. Metabolic Fingerprint of Dual Body Fluids Deciphers Diabetic Retinopathy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2412195. [PMID: 39871789 DOI: 10.1002/smll.202412195] [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: 12/15/2024] [Revised: 01/10/2025] [Indexed: 01/29/2025]
Abstract
Diabetic retinopathy (DR) is a microvascular complication of diabetes, affecting 34.6% of diabetes patients worldwide. Early detection and timely treatment can effectively improve the prognosis of DR. Metabolomic analysis provides a powerful tool for studying pathophysiological processes. Conducting metabolomic analyses on DR-related biofluids helps identify differential metabolic expressions during disease progression, thereby discovering potential biomarkers to support clinical diagnosis and treatment. Here, an innovative workflow for vitreous liquid analysis is established, and a machine learning-based DR analysis platform integrating vitreous liquid metabolic fingerprint (VL-MF) and plasma metabolic fingerprint (P-MF) derived via nanoparticle enhanced laser desorption/ionization mass spectrometry is developed. Direct VL-MF and P-MF are obtained with desirable reproducibility (coefficient of variation, CV <5%) and remarkable speed (3 s per sample), and DR patients are distinguished from healthy controls applying dual biofluid-MF with an area under the curve (AUC) of 0.957. Moreover, a biomarker candidate panel from vitreous liquid and plasma with an AUC of 0.945 is constructed and the related metabolic pathways are identified by metabolomics pathway analysis (MetPA). This work offers a powerful multi-biofluid platform that can not only contribute to DR but also provide solid references for other clinical applications.
Collapse
Affiliation(s)
- Yihan Wang
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Shunxiang Li
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Tong Li
- Department of Ophthalmology, National Clinical Research Center for Eye Diseases, Shanghai Gene Therapy Center, Shanghai Key Laboratory of Ocular Fundus Disease, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, P. R. China
| | - Jiao Wu
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Yida Huang
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Wanshan Liu
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Chunmeng Ding
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Lin Huang
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Xiaoyu Xu
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Yuning Wang
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Sai Gu
- Department of Chemical Engineering, The University of Warwick, Coventry, CV4 8UW, UK
| | - Kun Liu
- Department of Ophthalmology, National Clinical Research Center for Eye Diseases, Shanghai Gene Therapy Center, Shanghai Key Laboratory of Ocular Fundus Disease, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200040, P. R. China
| | - Kun Qian
- State Key Laboratory for Oncogenes and Related Genes, School of Biomedical Engineering, Institute of Medical Robotics and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, 200030, P. R. China
| | - Xiaodong Sun
- Department of Ophthalmology, National Clinical Research Center for Eye Diseases, Shanghai Gene Therapy Center, Shanghai Key Laboratory of Ocular Fundus Disease, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200040, P. R. China
| |
Collapse
|
30
|
Song H, Zhang Y, Wang F, Wang L, Xiong L, Shen X. Pectin: Structural Characteristics, ADME Profiles, and Their Interrelationship. Chem Biodivers 2025:e202402532. [PMID: 39920038 DOI: 10.1002/cbdv.202402532] [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: 10/05/2024] [Revised: 02/06/2025] [Accepted: 02/07/2025] [Indexed: 02/09/2025]
Abstract
Pectin, a plant-derived polysaccharide, is highly valued for its gelling, thickening, and stabilizing properties, with extensive applications in the food and pharmaceutical industries. This review provides a comprehensive analysis of pectin's structure, categorized by its degree of methyl esterification (DM) and key components, including homogalacturonan (HG) and rhamnogalacturonans (RG-I and RG-II). The influence of diverse extraction methods, such as subcritical water and microwave-assisted techniques, on its structure and functionality is critically examined. Furthermore, the review investigates the absorption, distribution, metabolism, and excretion (ADME) profiles of pectin, emphasizing how structural factors like molecular weight, DM, and neutral sugars impact bioavailability and interactions with gut microbiota. Notably, this review highlights emerging research methodologies, offering novel insights into pectin's pharmacokinetics. By addressing these interrelationships, the review underscores pectin's potential applications in functional foods, personalized nutrition, and targeted therapeutics and identifies key knowledge gaps for future research.
Collapse
Affiliation(s)
- Haizhao Song
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, China
| | - Yanhui Zhang
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, China
| | - Fang Wang
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, China
| | - Luanfeng Wang
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, China
| | - Ling Xiong
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, China
| | - Xinchun Shen
- College of Food Science and Engineering/Collaborative Innovation Center for Modern Grain Circulation and Safety/Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, China
| |
Collapse
|
31
|
Song N, Gao H, Li J, Liu Y, Wang M, Ma Z, Zhang N, Zhang W. Microbiota from young mice counteracts susceptibility to age-related gout through modulating butyric acid levels in aged mice. eLife 2025; 13:RP98714. [PMID: 39907694 PMCID: PMC11798573 DOI: 10.7554/elife.98714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025] Open
Abstract
Gout is a prevalent form of inflammatory arthritis that occurs due to high levels of uric acid in the blood leading to the formation of urate crystals in and around the joints, particularly affecting the elderly. Recent research has provided evidence of distinct differences in the gut microbiota of patients with gout and hyperuricemia compared to healthy individuals. However, the link between gut microbiota and age-related gout remained underexplored. Our study found that gut microbiota plays a crucial role in determining susceptibility to age-related gout. Specifically, we observed that age-related gut microbiota regulated the activation of the NLRP3 inflammasome pathway and modulated uric acid metabolism. More scrutiny highlighted the positive impact of 'younger' microbiota on the gut microbiota structure of old or aged mice, enhancing butanoate metabolism and butyric acid content. Experimentation with butyrate supplementation indicated that butyric acid exerts a dual effect, inhibiting inflammation in acute gout and reducing serum uric acid levels. These insights emphasize the potential of gut microbiome rejuvenation in mitigating senile gout, unraveling the intricate dynamics between microbiota, aging, and gout. It potentially serves as a therapeutic target for senile gout-related conditions.
Collapse
Affiliation(s)
- Ning Song
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin UniversityChangchunChina
| | - Hang Gao
- Department of Bone and Joint Surgery, No 1 Hospital of Jilin UniversityChangchunChina
| | - Jianhao Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin UniversityChangchunChina
| | - Yi Liu
- Department of Bone and Joint Surgery, No 1 Hospital of Jilin UniversityChangchunChina
| | - Mingze Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin UniversityChangchunChina
| | - Zhiming Ma
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin UniversityChangchunChina
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin UniversityChangchunChina
| | - Wenlong Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin UniversityChangchunChina
| |
Collapse
|
32
|
Redruello-Requejo M, del Mar Blaya M, González-Reguero D, Robas-Mora M, Arranz-Herrero J, Partearroyo T, Varela-Moreiras G, Penalba-Iglesias D, Jiménez-Gómez P, Reche-Sainz P. Cross-Sectional Comparative Analysis of Gut Microbiota in Spanish Adolescents with Mediterranean and Western Diets. Nutrients 2025; 17:388. [PMID: 39940246 PMCID: PMC11820480 DOI: 10.3390/nu17030388] [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: 12/23/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/14/2025] Open
Abstract
Dietary patterns, such as the Mediterranean diet (MD) and the Western diet (WD), influence gut microbiota composition and functionality, which play important roles in energy metabolism and nutrient absorption. OBJECTIVES A descriptive cross-sectional study was designed to evaluate the gut microbiota of 19 Spanish adolescents and to investigate the association of MD and ultra-processed food (UPF) intake with microbial diversity and community structure. METHODS Functional diversity of gut microbiota was evaluated using Biolog EcoPlates, taxonomic composition was assessed with 16S rRNA sequencing via MinION, and phenotypic responses to antibiotics were analyzed using the cenoantibiogram technique under aerobic and anaerobic conditions. RESULTS Adolescents with higher adherence to the MD exhibited greater functional diversity, as per the Shannon-Weaver index. In addition, this group showed higher abundance of bacterial genera previously described as beneficial, such as Paraclostridium, Anaerobutyricum, Romboutsia, and Butyricicoccus. In contrast, adolescents reporting greater UPF intakes had a microbiota composition similar to those with low adherence to the MD, characterized by decreased abundance of beneficial genera. Regarding antibiotic resistance, significant differences were only observed under anaerobic conditions, with individuals with low adherence to the MD showing more sensitivity for most antibiotics tested. CONCLUSIONS These results suggest that the MD promotes a healthier and more balanced gut environment, potentially improving metabolic functions in adolescents. Despite the lack of differences in α-diversity, comparisons of microbial community structure between adolescents following the MD and those with high UPF (characteristic of the WD) showed clear differences in terms of β-diversity. These findings suggest that dietary patterns influence the composition of the gut microbiota in a more complex manner, beyond just taxonomic richness. The outcomes of this exploratory study highlight opportunities for future research to deepen understanding of the long-term health implications of these dietary patterns, as well as the mechanisms regulating the composition, functionality, and phenotypic responses to antibiotics of gut microbial communities.
Collapse
Affiliation(s)
- Marina Redruello-Requejo
- Grupo USP-CEU de Excelencia “Nutrición para la vida (Nutrition for Life)”, Ref: E02/0720, Department of Pharmaceutical and Health Sciences, Faculty of Pharmacy, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (T.P.); (G.V.-M.)
- Instituto Universitario CEU Alimentación y Sociedad, Faculty of Pharmacy, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - María del Mar Blaya
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
| | - Daniel González-Reguero
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
| | - Marina Robas-Mora
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
| | - Javier Arranz-Herrero
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Medicine Faculty, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - Teresa Partearroyo
- Grupo USP-CEU de Excelencia “Nutrición para la vida (Nutrition for Life)”, Ref: E02/0720, Department of Pharmaceutical and Health Sciences, Faculty of Pharmacy, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (T.P.); (G.V.-M.)
- Instituto Universitario CEU Alimentación y Sociedad, Faculty of Pharmacy, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - Gregorio Varela-Moreiras
- Grupo USP-CEU de Excelencia “Nutrición para la vida (Nutrition for Life)”, Ref: E02/0720, Department of Pharmaceutical and Health Sciences, Faculty of Pharmacy, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (T.P.); (G.V.-M.)
- Instituto Universitario CEU Alimentación y Sociedad, Faculty of Pharmacy, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain
| | - Diana Penalba-Iglesias
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
| | - Pedro Jiménez-Gómez
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
| | - Paloma Reche-Sainz
- Department of Pharmaceutical and Health Sciences, San Pablo University, CEU Universities, Urbanización Montepríncipe, 28660 Boadilla del Monte, Spain; (M.d.M.B.); (D.G.-R.); (M.R.-M.); (J.A.-H.); (D.P.-I.); (P.R.-S.)
| |
Collapse
|
33
|
Zhang T, Chang M, Hou X, Yan M, Zhang S, Song W, Sheng Q, Yuan Y, Yue T. Apple polyphenols prevent patulin-induced intestinal damage by modulating the gut microbiota and metabolism of the gut-liver axis. Food Chem 2025; 463:141049. [PMID: 39260178 DOI: 10.1016/j.foodchem.2024.141049] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/06/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
Patulin (PAT), a foodborne toxin, causes severe intestinal damage. To mitigate this health threat, mice were pretreated with apple polyphenols (AP) in their drinking water (0.01 % and 0.05 %) for eight weeks, followed by exposure to PAT during the last two weeks. Subsequently, histopathological and biochemical evaluations of intestinal tissues were conducted, alongside assessments of alterations in gut microbiota, colonic content metabolome, and hepatic metabolome. Consequently, AP alleviated PAT-induced villus and crypt injury, mucus depletion, GSH level decline, GSH-Px and SOD activity reduction, and MPO activity elevation. Notably, AP counteracted PAT-mediated microbiota disruptions and promoted the abundance of beneficial bacteria (Dubosiella, Akkermansia, Lachnospiraceae, and Lactobacillus). Furthermore, AP counteracted PAT-induced metabolic disorders in the colonic contents and liver. Ultimately, AP prevented intestinal injury by regulating the gut microbiota and amino acid, purine, butanoate, and glycerophospholipid metabolism in the gut-liver axis. These results underscore the potential of AP to prevent foodborne toxin-induced intestinal damage.
Collapse
Affiliation(s)
- Ting Zhang
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Min Chang
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Xiaohui Hou
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Min Yan
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Shirui Zhang
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Wei Song
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Qinglin Sheng
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China
| | - Yahong Yuan
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China.
| | - Tianli Yue
- College of Food Science and Technology, Northwest University, Xi'an 710069, Shaanxi, China; Laboratory of Nutritional and Healthy Food-Individuation Manufacturing Engineering, Xi'an 710069, Shaanxi, China; Research Center of Food Safety Risk Assessment and Control, Xi'an 710069, Shaanxi, China.
| |
Collapse
|
34
|
Zheng ZL, Zheng QF, Wang LQ, Liu Y. Bowel preparation before colonoscopy: Consequences, mechanisms, and treatment of intestinal dysbiosis. World J Gastroenterol 2025; 31:100589. [PMID: 39811511 PMCID: PMC11684204 DOI: 10.3748/wjg.v31.i2.100589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/22/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
The term "gut microbiota" primarily refers to the ecological community of various microorganisms in the gut, which constitutes the largest microbial community in the human body. Although adequate bowel preparation can improve the results of colonoscopy, it may interfere with the gut microbiota. Bowel preparation for colonoscopy can lead to transient changes in the gut microbiota, potentially affecting an individual's health, especially in vulnerable populations, such as patients with inflammatory bowel disease. However, measures such as oral probiotics may ameliorate these adverse effects. We focused on the bowel preparation-induced changes in the gut microbiota and host health status, hypothesized the factors influencing these changes, and attempted to identify measures that may reduce dysbiosis, thereby providing more information for individualized bowel preparation for colonoscopy in the future.
Collapse
Affiliation(s)
- Ze-Long Zheng
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Qing-Fan Zheng
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Li-Qiang Wang
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Yi Liu
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
35
|
Qiu J, Wu S, Huang R, Liao Z, Pan X, Zhao K, Peng Y, Xiang S, Cao Y, Ma Y, Xiao Z. Effects of antibiotic therapy on the early development of gut microbiota and butyrate-producers in early infants. Front Microbiol 2025; 15:1508217. [PMID: 39839108 PMCID: PMC11748296 DOI: 10.3389/fmicb.2024.1508217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/09/2024] [Indexed: 01/23/2025] Open
Abstract
Background Antibiotics, as the most commonly prescribed class of drugs in neonatal intensive care units, have an important impact on the developing neonatal gut microbiota. Therefore, comprehending the effects of commonly used antibiotic therapy on the gut microbiota and butyrate-producers in early infants could provide information for therapeutic decision-making in the NICU. Objectives To explore the effects of antibiotic therapy on the early development of gut microbiota and butyrate-producers in early infants. Methods A total of 72 infants were included in the study. We performed 16S rRNA sequencing on stool swab samples collected from neonatal intensive care unit patients who received amoxicillin-clavulanic acid (AC, n = 10), moxalactam (ML, n = 28) and non-antibiotics (NA, n = 34). We then compared the taxonomic composition between treatment regimens, focusing on differences in butyrate-producers. Results Our study showed that there were significant differences in Shannon index (p = 0.033) and Beta diversity (p = 0.014) among the three groups. At the family level, compared with the other two groups, the relative abundance of Clostridiaceae (p < 0.001) and Veillonellaceae (p = 0.004) were significantly higher, while the relative abundance of Enterococcidae (p < 0.001) was significantly lower in the NA group. The relative abundance of Enterobacteriaceae (p = 0.022) in the AC group was greater than that in the other two groups. Additionally, butyrate-producers (p < 0.001), especially Clostridiaceae (p < 0.001), were noticeably more abundant in the NA group. The relative abundance of Clostridiaceae and butyrate-producers were the lowest in the ML group (p < 0.001). Conclusion We found that antibiotic therapy had an adverse impact on the initial development of gut microbiota and leaded to a reduction in the abundance of butyrate-producers, particularly Clostridiaceae. Furthermore, moxalactam had a more pronounced effect on the gut microbiota compared to amoxicillin-clavulanic acid.
Collapse
Affiliation(s)
- Jun Qiu
- The School of Pediatrics, Hengyang Medical School, University of South China, Hunan Children’s Hospital, Hengyang, Hunan, China
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, China
| | - Sha Wu
- The School of Pediatrics, Hengyang Medical School, University of South China, Hunan Children’s Hospital, Hengyang, Hunan, China
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, China
| | - Ruiwen Huang
- Department of Neonatology, Hunan Children's Hospital, Changsha, Hunan, China
| | - Zhenyu Liao
- Department of Neonatology, Hunan Children's Hospital, Changsha, Hunan, China
| | - Xiongfeng Pan
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, China
| | - Kunyan Zhao
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, China
- The School of Public Health, University of South China, Hengyang, China
| | - Yunlong Peng
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, China
- Department of Epidemiology and Health Statistics, Medical College of Soochow University, Suzhou, China
| | - Shiting Xiang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, China
| | - Yunhui Cao
- The School of Pediatrics, Hengyang Medical School, University of South China, Hunan Children’s Hospital, Hengyang, Hunan, China
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, China
| | - Ye Ma
- Department of Neonatology, Hunan Children's Hospital, Changsha, Hunan, China
| | - Zhenghui Xiao
- The School of Pediatrics, Hengyang Medical School, University of South China, Hunan Children’s Hospital, Hengyang, Hunan, China
- Department of Emergency Center, Hunan Children’s Hospital, Changsha, China
| |
Collapse
|
36
|
Li X, Lin H, Peng J, Gong J. Exploring the mediating role of blood metabolites in the relationship between gut microbiota and gastric cancer risk: a Mendelian randomization study. Front Cell Infect Microbiol 2025; 14:1453286. [PMID: 39839262 PMCID: PMC11747456 DOI: 10.3389/fcimb.2024.1453286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Background Prior studies have established correlations between gut microbiota (GM) dysbiosis, circulating metabolite alterations, and gastric cancer (GC) risk. However, the causal nature of these associations remains uncertain. Methods We utilized summary data from genome-wide association studies (GWAS) on GM (European, n=8,956), blood metabolites (European, n=120,241; East Asian, n=4,435), and GC (European, n=476,116; East Asian, n=167,122) to perform a bidirectional Mendelian randomization (MR) analysis, investigating the causal effects of GM and metabolites on GC risk. Additionally, we conducted mediation analysis (two-step MR) to identify potential metabolite mediators in the GM-GC relationship. Results We identified twelve negative and seven positive associations between specific GM taxa and GC risk. For blood metabolites, seven traits were found to be significantly associated with reduced GC risk in the European population, with these findings subsequently validated in the East Asian cohort. Three GM taxa showed potential causal associations with five metabolic traits: the Bacteroidia class and Bacteroidales order were positively correlated with five metabolites (all P < 0.013), while Bacteroides OTU97_27 exhibited a negative correlation with one metabolite (P = 0.007). Two-step MR analysis indicated that total lipids in intermediate-density lipoprotein (IDL), IDL particle concentration, phospholipids in medium low-density lipoprotein (LDL), phospholipids in small LDL, and free cholesterol in small LDL indirectly influenced the association between Bacteroidia class/Bacteroidales order and GC, with mediation proportions of 1.71% (P = 0.048), 1.69% (P = 0.048), 2.05% (P = 0.045), 1.85% (P = 0.048), and 1.99% (P = 0.045), respectively. Conclusion The present study provides suggestive evidence of a causal relationship between specific GM, blood metabolites, and GC risk, potentially offering new insights into GC etiology.
Collapse
Affiliation(s)
- Xiaocheng Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of General Surgery, The First Affiliated Hospital of Hunan University of Medicine, Huaihua, Hunan, China
| | - Huapeng Lin
- Department of Gastroenterology and Hepatology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Digestive Diseases Research and Clinical Translation, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Gut Microecology and Associated Major Diseases Research, Shanghai Jiao Tong University, Shanghai, China
| | - Jing Peng
- Department of General Surgery, The First Affiliated Hospital of Hunan University of Medicine, Huaihua, Hunan, China
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
37
|
Saban Güler M, Arslan S, Ağagündüz D, Cerqua I, Pagano E, Berni Canani R, Capasso R. Butyrate: A potential mediator of obesity and microbiome via different mechanisms of actions. Food Res Int 2025; 199:115420. [PMID: 39658184 DOI: 10.1016/j.foodres.2024.115420] [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: 04/18/2024] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024]
Abstract
Butyrate, a short-chain fatty acid, is a crucial product of gut microbial fermentation with significant implications for various metabolic and physiological processes. Dietary sources of butyrate are limited, primarily derived from the fermentation of dietary fibers by butyrate-producing gut bacteria. Butyrate exerts its effects primarily as a histone deacetylase (HDAC) inhibitor and through signaling pathways involving G protein-coupled receptors (GPCRs). Its diverse benefits include promoting gut health, enhancing energy metabolism, and potentially alleviating complications associated with obesity. However, the exact role of butyrate in obesity is still under investigation, with a limited number of human trials necessitating further research to determine its efficacy and safety profile. Moreover, butyrate impact on the gut-brain axis and its modulation of microbiome effect on behavior highlight its broader importance in regulating host physiology. A thorough understanding of the metabolic pathways and mechanisms of butyrate is essential for developing targeted interventions for metabolic disorders. Continued research is crucial to fully realize its therapeutic potential and optimize its clinical applications in human health. In summary, this review illuminates the multifaceted role of butyrate as a potential mediator of obesity and related metabolic changes.
Collapse
Affiliation(s)
- Meryem Saban Güler
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Turkey
| | - Sabriye Arslan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Turkey
| | - Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Turkey.
| | - Ida Cerqua
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Ester Pagano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science and ImmunoNutritionLab at CEINGE Biotechnologies Research Center and Task Force for Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Portici, 80055 Naples, Italy.
| |
Collapse
|
38
|
Huang X, Geng H, Liang C, Xiong X, Du X, Zhuan Q, Liu Z, Meng L, Zhou D, Zhang L, Fu X, Qi X, Hou Y. Leonurine restrains granulosa cell ferroptosis through SLC7A11/GPX4 axis to promote the treatment of polycystic ovary syndrome. Free Radic Biol Med 2025; 226:330-347. [PMID: 39547522 DOI: 10.1016/j.freeradbiomed.2024.11.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/02/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine disorder marked by ovarian dysfunction and metabolic abnormality. This study explores the therapeutic potential of leonurine (SCM-198) in PCOS. Our results show that SCM-198 treatment significantly improved ovarian function, hormone disorders and insulin resistance while reducing granulosa cell ferroptosis. This study provides the first evidence that SCM-198 modulates the gut microbiota composition, increases the abundance of Christensenella minuta, and boosts butyrate levels. Transcriptomic and metabolomic analyses revealed that PCOS patients exhibit granulosa cell ferroptosis and decreased butyrate levels in follicular fluid. Butyrate was shown to alleviate ferroptosis in granulosa cells via the SLC7A11/TXNRD1/GPX4 pathway, as confirmed in vitro with KGN cells. The therapeutic mechanism of SCM-198 in the management of PCOS via the gut microbiota-ovary axis involves the enhancement of gut microbiota and its metabolites. This intervention improves ovarian function and alleviates PCOS symptoms by targeting ferroptosis in granulosa cells.
Collapse
Affiliation(s)
- Xiaohan Huang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Hucheng Geng
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Chunxiao Liang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xianglei Xiong
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Xingzhu Du
- State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Qingrui Zhuan
- State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Zhiqiang Liu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Lin Meng
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Dan Zhou
- State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Luyao Zhang
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xiangwei Fu
- State Key Laboratory of Animal Biotech Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xinyu Qi
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
| | - Yunpeng Hou
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China.
| |
Collapse
|
39
|
Chen J, Song Y, Zeng W, Wang L, Qin J, Fang L, Ding Y. RESEARCH PROGRESS ON THE ROLE OF GUT MICROBIOTA AND ITS METABOLITES IN THE OCCURRENCE AND DEVELOPMENT OF SEPTIC-ASSOCIATED LIVER INJURY. Shock 2025; 63:4-10. [PMID: 39158846 DOI: 10.1097/shk.0000000000002441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
ABSTRACT Sepsis is a life-threatening organ dysfunction that occurs due to a dysregulated host response to infection. Septic-associated liver injury (SALI) has been closely linked to the prognosis and mortality of sepsis. Recent investigations have delved into the gut-liver axis and its association with SALI, identifying its pivotal role in the gut microbiota. Bacterial translocation and the onset of SALI can occur due to an imbalance in the gut microbiota, impairing the function of the gut barrier. Moreover, their metabolites might exacerbate or initiate SALI by modulating immune responses. Nevertheless, interventions to restore the balance of the gut microbiota, such as the administration of probiotics, fecal microbiota transplantation, or dietary adjustments, may ameliorate SALI and enhance the prognosis and survival rates of septic patients. This review aimed to elucidate the function of the gut microbiota in the genesis and procession of SALI and its potential therapeutic value, offering a deeper understanding of the pathogenesis and therapeutic avenues for SALI.
Collapse
Affiliation(s)
- Jiangtao Chen
- Department of Intensive Care Unit, Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yu Song
- Department of Hepatology, Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Wenqing Zeng
- Department of Intensive Care Unit, Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Lei Wang
- Department of Intensive Care Unit, Affiliated Hospital of Hangzhou Normal University, Hangzhou, Zhejiang Province, China
| | - Jinyan Qin
- Department of Intensive Care Unit, Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Lexin Fang
- Department of Intensive Care Unit, Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yueping Ding
- Department of Intensive Care Unit, Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| |
Collapse
|
40
|
Connolly KR, Sweeney T, O’Doherty JV. Sustainable Nutritional Strategies for Gut Health in Weaned Pigs: The Role of Reduced Dietary Crude Protein, Organic Acids and Butyrate Production. Animals (Basel) 2024; 15:66. [PMID: 39795009 PMCID: PMC11718951 DOI: 10.3390/ani15010066] [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: 12/02/2024] [Revised: 12/21/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Weaning in piglets presents significant physiological and immunological challenges, including gut dysbiosis and increased susceptibility to post-weaning diarrhoea (PWD). Abrupt dietary, environmental, and social changes during this period disrupt the intestinal barrier and microbiota, often necessitating antimicrobial use. Sustainable dietary strategies are critical to addressing these issues while reducing reliance on antimicrobials. Reducing dietary crude protein mitigates the availability of undigested proteins for pathogenic bacteria, lowering harmful by-products like ammonia and branched-chain fatty acids, which exacerbate dysbiosis. Organic acid supplementation improves gastric acidification, nutrient absorption, and microbial balance, while also serving as an energy-efficient alternative to traditional grain preservation methods. Increasing intestinal butyrate, a key short-chain fatty acid with anti-inflammatory and gut-protective properties, is particularly promising. Butyrate strengthens intestinal barrier integrity by upregulating tight junction proteins, reduces inflammation by modulating cytokine responses, and promotes anaerobic microbial stability. Exogenous butyrate supplementation via salts provides immediate benefits, while endogenous stimulation through prebiotics (e.g., resistant starch) and probiotics promotes sustained butyrate production. These interventions selectively enhance butyrate-producing bacteria such as Roseburia and Faecalibacterium prausnitzii, further stabilising the gut microbiota. Integrating these strategies can enhance gut integrity, microbial resilience, and immune responses in weaned piglets. Their combination offers a sustainable, antimicrobial-free approach to improving health and productivity in modern pig production systems.
Collapse
Affiliation(s)
- Kathryn Ruth Connolly
- School of Agriculture and Food Science, University College Dublin, Belfield, D04 W6F6 Dublin, Ireland;
| | - Torres Sweeney
- School of Veterinary Medicine, University College Dublin, Belfield, D04 W6F6 Dublin, Ireland;
| | - John V. O’Doherty
- School of Agriculture and Food Science, University College Dublin, Belfield, D04 W6F6 Dublin, Ireland;
| |
Collapse
|
41
|
Spagnolo P, Tweddell D, Cela E, Daley M, Clarson C, Rupar CA, Stranges S, Bravo M, Cepinskas G, Fraser DD. Metabolomic signature of pediatric diabetic ketoacidosis: key metabolites, pathways, and panels linked to clinical variables. Mol Med 2024; 30:250. [PMID: 39707182 DOI: 10.1186/s10020-024-01046-9] [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] [Accepted: 12/12/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Diabetic ketoacidosis (DKA) is a serious complication of type 1 diabetes (T1D), arising from relative insulin deficiency and leading to hyperglycemia, ketonemia, and metabolic acidosis. Early detection and treatment are essential to prevent severe outcomes. This pediatric case-control study utilized plasma metabolomics to explore metabolic alterations associated with DKA and to identify predictive metabolite patterns. METHODS We examined 34 T1D participants, including 17 patients admitted with severe DKA and 17 age- and sex-matched individuals in insulin-controlled states. A total of 215 plasma metabolites were analyzed using proton nuclear magnetic resonance and direct-injection liquid chromatography/mass spectrometry. Multivariate statistical methods, machine learning techniques, and bioinformatics were employed for data analysis. RESULTS After adjusting for multiple comparisons, 65 metabolites were found to differ significantly between the groups (28 increased and 37 decreased). Metabolomics profiling demonstrated 100% accuracy in differentiating severe DKA from insulin-controlled states. Random forest analysis indicated that classification accuracy was primarily influenced by changes in ketone bodies, acylcarnitines, and phosphatidylcholines. Additionally, groups of metabolites (ranging in number from 8 to 18) correlated with key clinical and biochemical variables, including pH, bicarbonate, glucose, HbA1c, and Glasgow Coma Scale scores. CONCLUSIONS These findings underscore significant metabolic disturbances in severe DKA and their associations with critical clinical indicators. Future investigations should explore if metabolic alterations in severe DKA can identify patients at increased risk of complications and/or guide future therapeutic interventions.
Collapse
Affiliation(s)
- Paolo Spagnolo
- Medicine, Campus Bio-Medico University of Rome, Via Alvaro del Portillo 21, 00128, Rome, Italy
| | - David Tweddell
- Computer Science, Western University, London, ON, N6A 3K7, Canada
| | - Enis Cela
- Physiology & Pharmacology, Western University, London, ON, N6A 3K7, Canada
| | - Mark Daley
- Computer Science, Western University, London, ON, N6A 3K7, Canada
- Epidemiology and Biostatistics, Western University, London, ON, N6G 2M1, Canada
| | - Cheril Clarson
- Pediatrics, Western University, London, ON, N6A 3K7, Canada
| | - C Anthony Rupar
- Pediatrics, Western University, London, ON, N6A 3K7, Canada
- Biochemistry, Western University, London, ON, N6A 3K7, Canada
| | - Saverio Stranges
- Epidemiology and Biostatistics, Western University, London, ON, N6G 2M1, Canada
- Family Medicine, Western University, London, ON, N6G 2M1, Canada
- Clinical Medicine and Surgery, University of Naples Federico II, Naples, 80131, Italy
- Medicine, Western University, London, ON, N6A 3K7, Canada
| | - Michael Bravo
- Emergency Department, Hospital for Sick Children, Toronto, ON, M5G 1X8, Canada
| | - Gediminas Cepinskas
- Medical Biophysics, Western University, London, ON, N6A 3K7, Canada
- Anatomy and Cell Biology, Western University, London, ON, N6A 3K7, Canada
- London Health Sciences Centre Research Institute, London, ON, N6C 2R5, Canada
| | - Douglas D Fraser
- Physiology & Pharmacology, Western University, London, ON, N6A 3K7, Canada.
- Pediatrics, Western University, London, ON, N6A 3K7, Canada.
- London Health Sciences Centre Research Institute, London, ON, N6C 2R5, Canada.
- Clinical Neurological Sciences, Western University, London, ON, N6A 3K7, Canada.
- Child Health Research Institute, London, ON, N6C 4V3, Canada.
- A5-132, Victoria Research Laboratories, London Health Sciences Centre, Victoria Campus, 800 Commissioners Road E, London, ON, N6A 5W9, Canada.
| |
Collapse
|
42
|
Lu LL, Liu LZ, Li L, Hu YY, Xian XH, Li WB. Sodium butyrate improves cognitive dysfunction in high-fat diet/ streptozotocin-induced type 2 diabetic mice by ameliorating hippocampal mitochondrial damage through regulating AMPK/PGC-1α pathway. Neuropharmacology 2024; 261:110139. [PMID: 39233201 DOI: 10.1016/j.neuropharm.2024.110139] [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: 07/15/2024] [Revised: 08/17/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
Cognitive dysfunction is an important comorbidity of type 2 diabetes mellitus (T2DM). Sodium butyrate (NaB) is a short-chain fatty acid and has an effect improving T2DM-associated cognitive dysfunction. Using a high-fat diet (HFD)/streptozotocin (STZ)-induced T2DM mouse model, the present study investigated the mechanism involved in the beneficial effect of butyrate on diabetic cognitive dysfunction, with a focus on ameliorating mitochondrial damage through regulating the adenosine monophosphate-activated protein kinase/peroxisome proliferator-activated receptor gamma coactivator 1α (AMPK/PGC-1α) pathway considering the important role of mitochondrial impairments in the occurrence of T2DM-associated cognitive dysfunction. We found, based on reconfirmation of the improvement of NaB on cognitive impairment, that NaB treatment improved damaged synaptic structural plasticity including the decrease in dendritic spine density and downregulation in the expression of postsynaptic density protein 95 and synaptophysin in the hippocampus in the model mice. NaB treatment also ameliorated mitochondrial ultrastructural damage, increased mitochondrial membrane potential and adenosine 5'-triphosphate content, and improved mitochondrial biogenesis and dynamics in the model mice. Furthermore, the expression of phosphorylated AMPK and PGC-1α was upregulated after NaB treatment in the model mice. In particular, the above beneficial effects of NaB were blocked by the inhibition of either AMPK or PGC-1α. In conclusion, NaB treatment improved cognitive impairment and damaged synaptic structural plasticity in the hippocampus by ameliorating damage to mitochondrial morphology and function through regulating the AMPK/PGC-1α pathway in HFD/STZ-induced T2DM mice.
Collapse
Affiliation(s)
- Li-Li Lu
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, PR China; Department of Pathology, The Third Hospital of Shijiazhuang, 15 Tiyu South Avenue, Shijiazhuang, 050011, PR China
| | - Li-Zhe Liu
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, PR China
| | - Li Li
- Central Laboratory, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, 050000, PR China
| | - Yu-Yan Hu
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, PR China
| | - Xiao-Hui Xian
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, PR China.
| | - Wen-Bin Li
- Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang, 050017, PR China.
| |
Collapse
|
43
|
Chen W, Zhou Z, Qi R, Zhou J, Liang H, Huang P, Zou Z, Dong L, Li H, Du B, Li P. Ameliorative effects of Trichosanthes kirilowii Maxim. seed oil on hyperlipidemia rats associated with the regulation of gut microbiology and metabolomics. Food Res Int 2024; 197:115141. [PMID: 39593355 DOI: 10.1016/j.foodres.2024.115141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 11/28/2024]
Abstract
The mechanisms underlying the ameliorative effects of polyunsaturated fatty acids (PUFAs) on metabolic disorders induced by a high-fat diet (HFD) remain poorly unclear. In this study, we investigated the anti-hyperlipidemic effects of Trichosanthes kirilowii Maxim. (T. kirilowii) seed oil rich in conjugated linolenic acid in HFD-induced hyperlipidemic rats, by the gut microbiome, cecum bile acids (BAs), and serum metabolomics. The results showed that T. kirilowii seed oil improved dyslipidemia, hepatic steatosis, oxidative stress, and inflammatory responses in HFD-induced rats. Meanwhile, T. kirilowii seed oil inhibited sterol regulatory element-binding protein 1c (SREBP-1c) mediated fatty acid synthesis and upregulated cholesterol 7-alpha hydroxylase (CYP7A1) mediated hepatic cholesterol metabolism to exert hypolipidemic effects. The administration of high dose T. kirilowii seed oil (THD) improved gut microbiota dysbiosis, increased the relative abundance of beneficial bacteria Romboutsia and unidentified_Oscillospiraceae, and decreased the relative abundance of Christensenellaceae_R-7 group, Phascolarctobacterium, and Bacteroides in HFD-induced rats. T. kirilowii seed oil reduced the accumulation of cecum primary BAs in HFD-induced rats. In addition, THD reversed the HFD-induced changes in 24 serum metabolites including leucine, isoleucine, acetylcarnitine, and glucose. Metabolic pathway enrichment analysis of the differential metabolites revealed that valine, leucine and isoleucine metabolism, butanoate metabolism, citrate cycle, and glycolysis were potential metabolic pathways involved in the anti-hyperlipidemic effects of T. kirilowii seed oil. In conclusion, this study found that dietary T. kirilowii seed oil alleviated gut microbiota dysbiosis and improved metabolic disorders in hyperlipidemic rats. This provides new insights into the anti-hyperlipidemic mechanism by which other families of PUFAs are derived from different plants.
Collapse
Affiliation(s)
- Weili Chen
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Zhangbao Zhou
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Ruida Qi
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Jun Zhou
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Huiying Liang
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Pinxi Huang
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Zebin Zou
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Ling Dong
- Institute of Horticulture, Anhui Academy of Agricultural Sciences, Hefei, Anhui 230031, China
| | - Hua Li
- Anhui Youyu Kuayue Food Development Co., Ltd, Anqing, Anhui 246300, China
| | - Bing Du
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, China
| | - Pan Li
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.
| |
Collapse
|
44
|
Yang Q, Wang Z, Liu M, Gan L. Causal Relationship Between Gut Microbiota and Leukemia: Future Perspectives. Oncol Ther 2024; 12:663-683. [PMID: 39217582 PMCID: PMC11573970 DOI: 10.1007/s40487-024-00300-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
The gut microbiota plays a crucial role in maintaining homeostasis in the human gastrointestinal tract. Numerous studies have shown a strong association between the gut microbiota and the emergence and progression of various diseases. Leukemia is one of the most common hematologic malignancies. Although standardized protocols and expert consensus have been developed for routine diagnosis and treatment, limitations remain due to individual differences. Nevertheless, a large number of studies have established a link between the gut microbiota and leukemia, with disturbances in the gut microbiota directly or indirectly affecting the development of leukemia. However, the causal relationship between the two remains unclear, and studying and exploring the causal relationship may open up entirely new avenues and protocols for use in the prevention and/or treatment of leukemia, offering new insights into diagnosis and treatment. In this review, the intricate relationship between the gut microbiota and leukemia is explored in depth, including causal associations, metabolite effects, therapeutic applications, and complications. Based on the characteristics of the gut microbiota, the future applications and prospects of gut microbiota are discussed to provide useful information for clinical treatment of leukemia.
Collapse
Affiliation(s)
- Qiang Yang
- Mianyang Central Hospital, Fucheng District, Mianyang City, 621000, Sichuan Province, China
| | - Zexin Wang
- Mianyang Central Hospital, Fucheng District, Mianyang City, 621000, Sichuan Province, China.
| | - Miao Liu
- Mianyang Central Hospital, Fucheng District, Mianyang City, 621000, Sichuan Province, China
| | - Lingling Gan
- Mianyang Central Hospital, Fucheng District, Mianyang City, 621000, Sichuan Province, China
| |
Collapse
|
45
|
Berriel Diaz M, Rohm M, Herzig S. Cancer cachexia: multilevel metabolic dysfunction. Nat Metab 2024; 6:2222-2245. [PMID: 39578650 DOI: 10.1038/s42255-024-01167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024]
Abstract
Cancer cachexia is a complex metabolic disorder marked by unintentional body weight loss or 'wasting' of body mass, driven by multiple aetiological factors operating at various levels. It is associated with many malignancies and significantly contributes to cancer-related morbidity and mortality. With emerging recognition of cancer as a systemic disease, there is increasing awareness that understanding and treatment of cancer cachexia may represent a crucial cornerstone for improved management of cancer. Here, we describe the metabolic changes contributing to body wasting in cachexia and explain how the entangled action of both tumour-derived and host-amplified processes induces these metabolic changes. We discuss energy homeostasis and possible ways that the presence of a tumour interferes with or hijacks physiological energy conservation pathways. In that context, we highlight the role played by metabolic cross-talk mechanisms in cachexia pathogenesis. Lastly, we elaborate on the challenges and opportunities in the treatment of this devastating paraneoplastic phenomenon that arise from the complex and multifaceted metabolic cross-talk mechanisms and provide a status on current and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Chair Molecular Metabolic Control, Technical University of Munich, Munich, Germany.
| |
Collapse
|
46
|
He Z, Xiong H, Liu L, Li Q, Wu K, Deng X, Yang L, Xiao Q, Deng X. Influence of Postoperative Insulin Resistance on Short-Term Outcomes of Radical Gastrectomy for Gastric Cancer: A Microbiome and Metabolome-Based Prospective Cohort Study. Ann Surg Oncol 2024; 31:8638-8650. [PMID: 39222298 DOI: 10.1245/s10434-024-16125-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Gastrectomy is one of the main treatment modalities for gastric cancer (GC) and induces pathophysiological changes that significantly affect patients' postoperative recovery. In this study, we investigated the relationships between altered insulin resistance (IR), inflammation, and gut microbiota associated with gastrectomy. PATIENTS AND METHODS This study was a single-center prospective cohort investigation involving 60 patients with GC who underwent gastrectomy between May 2023 and April 2024. Monitoring encompassed IR, inflammation, and nutrition-related markers via blood assays, while gut microbiota analysis employed high-throughput sequencing, and short-chain fatty acids (SCFAs) were examined through targeted metabolomics. The study is registered under the number ChiCTR2300075653. RESULTS The patients exhibited a significant increase in post-gastrectomy IR markers (P < 0.001), accompanied by elevated inflammation markers (P < 0.001), and also showed decreased nutrition-related indicators (P < 0.001). Notable alterations were observed in the gut microbiota, including reductions in Bifidobacterium and Faecalibacterium, an increase in Streptococcus, and a noteworthy decrease in fecal butyrate. Patients with postoperative IR exhibited poorer inflammation markers (P < 0.05), nutritional indicators (P < 0.05), and postoperative recovery parameters (P < 0.05). Furthermore, significant negative correlations were observed between IR and Bifidobacterium, Faecalibacterium, as well as butyrate. CONCLUSIONS Patients with GC post-gastrectomy displayed heightened IR, exacerbated inflammation, and compromised nutritional status. Disturbed gut microbiota and reduced fecal butyrate were observed. Gut microbiota and metabolite butyrate production may be predictors of postoperative IR and short-term outcomes in patients with GC.
Collapse
Affiliation(s)
- Zhipeng He
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Huan Xiong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Lulin Liu
- Department of Vascular Surgery, Heyuan Hospital of Guangdong Provincial People's Hospital, Heyuan, People's Republic of China
| | - Qiang Li
- Department of Vascular Surgery, Tengzhou Central People's Hospital, Tengzhou, People's Republic of China
| | - Kai Wu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Xi Deng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China
| | - Liang Yang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China.
| | - Qun Xiao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan College of Traditional Chinese Medicine, Zhuzhou, People's Republic of China.
| | - Xiaorong Deng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People's Republic of China.
| |
Collapse
|
47
|
Yang C, Liu L, Du Y, Zhao L, Liu L, Yang X, Zhao Y. Summer-autumn tea promotes adipocyte browning and thermogenesis in association with gut microbiota regulation in high-fat diet-fed mice. Food Funct 2024; 15:11458-11471. [PMID: 39479981 DOI: 10.1039/d4fo03826f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
This study revealed for the first time the anti-obesity effect of summer-autumn tea aqueous extract (SATE) and its underlying mechanism. High-fat diet (HFD)-fed C57BL/6J mice were treated with or without 400 mg kg-1 SATE for 12 weeks, and administration of SATE significantly ameliorated glucolipid metabolism disorder and induced beige-fat development and brown adipose tissue (BAT)-derived non-shivering thermogenesis via the AMPK-PGC-1α-UCP1 signal axis in HFD-fed mice. 16S rDNA-based microbiota and targeted metabolomics analyses indicated that SATE improved intestinal microbiota dysbiosis and microbial metabolism abnormality caused by HFD, reflected by a dramatic increase in the relative abundance of Muribaculaceae, Bifidobacterium and Odoribacter and production of short-chain fatty acids (SCFAs). Interestingly, SATE-induced thermogenesis was highly correlated with the reconstruction of the gut microbiome and the formation of SCFAs. These findings suggest that SATE has the potential to alleviate obesity by activating adipose browning and thermogenesis in association with the reconstruction of the gut microbiota and its metabolites, providing a theoretical foundation for summer-autumn tea as a functional tea to prevent obesity.
Collapse
Affiliation(s)
- Chengcheng Yang
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Luyao Liu
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Yao Du
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Lu Zhao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Lu Liu
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Xingbin Yang
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Yan Zhao
- Key Laboratory of Ministry of Education for Medicinal Resource and Natural Pharmaceutical Chemistry, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
48
|
Jørgensen KS, Pedersen SS, Hjorth SA, Billestrup N, Prause M. Protection of beta cells against cytokine-induced apoptosis by the gut microbial metabolite butyrate. FEBS J 2024. [PMID: 39569473 DOI: 10.1111/febs.17334] [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: 07/04/2024] [Revised: 08/28/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
Type 1 diabetes (T1D) is characterized by immune cell infiltration in the islets of Langerhans, leading to the destruction of insulin-producing beta cells. This destruction is driven by secreted cytokines and cytotoxic T cells inducing apoptosis in beta cells. Butyrate, a metabolite produced by the gut microbiota, has been shown to have various health benefits, including anti-inflammatory and anti-diabetic effects. In this study, we investigated the potential protective effects of butyrate on cytokine-induced apoptosis in beta cells and explored the underlying mechanisms. Insulin-secreting INS-1E cells and isolated mouse islets were treated with interleukin-1beta (IL-1β) or a combination of IL-1β and interferon-gamma (IFN-γ) in the presence or absence of butyrate. We analyzed apoptosis, nitric oxide (NO) levels, expression of stress-related genes, and immune cell migration. Our results demonstrated that butyrate significantly attenuated cytokine-induced apoptosis in both INS-1E cells and mouse islets, accompanied by a reduction in NO levels. Butyrate also decreased the expression of endoplasmic reticulum (ER) stress markers such as Chop, phosphorylated eIF2α and Atf4, as well as some pro-apoptotic genes including Dp5 and Puma. Butyrate reduced the cytokine-induced expression of the chemokine genes Cxcl1 and Cxcl10 in mouse islets, as well as the chemotactic activity of THP-1 monocytes toward conditioned media from IL-1β-exposed islets. In conclusion, these findings indicate that butyrate protects beta cells from cytokine-induced apoptosis and ER stress, suggesting its potential as a therapeutic agent to prevent beta cell destruction in T1D.
Collapse
Affiliation(s)
- Kasper Suhr Jørgensen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Signe Schultz Pedersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Siv Annegrethe Hjorth
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Nils Billestrup
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Michala Prause
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
49
|
Yi B, Su K, Cai YL, Chen XL, Bao Y, Wen ZY. Liraglutide ameliorates diabetic kidney disease by modulating gut microbiota and L-5-Oxoproline. Eur J Pharmacol 2024; 983:176905. [PMID: 39154828 DOI: 10.1016/j.ejphar.2024.176905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 08/20/2024]
Abstract
The gut microbiome-metabolites-kidney axis is a potential target for treating diabetic kidney disease (DKD). Our previous study found that Liraglutide attenuated DKD in rats by decreasing renal tubular ectopic lipid deposition (ELD) and serum metabolites levels, including L-5-Oxoproline (5-OP). However, the response of gut microbiome-metabolites-kidney axis to Liraglutide in DKD rats and the effect of 5-OP on ELD remain unknown. In this study, Sprague-Dawley rats were used as an animal model of DKD. They were subjected to a high fat diet, streptozotocin and uninephrectomy, followed by Liraglutide treatment (0.4 mg/kg d). Additionally, HK-2 cells were incubated with 30 mM glucose and 200 μM palmitate for 24h, and exposed to different concentrations of 5-OP. In DKD rats, Liraglutide dramatically improved the renal tubule structure. It increased the Simpson index (F = 4.487, p = 0.035) and reduced the Actinobacteria-to-Bacteroidetes ratio (F = 6.189, p = 0.014). At the genus level, Liraglutide increased the relative abundance of Clostridium, Oscillospira, Sarcina, SMB53, and 02d06 while decreasing that of Allobaculum. Meanwhile, 13 metabolites were significantly altered after Liraglutide treatment. Multi-omics analysis found that 5-OP levels were positively correlated with Clostridium abundance but negatively correlated with renal injury related indicators. In HK-2 cells, 5-OP significantly reduced the ELD in a dose-dependent manner through inhibiting the expression of SREBP1 and FAS. Overall, the renoprotective effect of Liraglutide in DKD rats is linked to the improvement of the gut microbiota composition and increased serum 5-OP levels, which may reduce ELD in renal tubular cells by lowering lipid synthesis.
Collapse
Affiliation(s)
- Bo Yi
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ke Su
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Yu-Li Cai
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiao-Ling Chen
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yan Bao
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Zhong-Yuan Wen
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
50
|
Pan X, Song Y, Liang Y, Feng G, Wang Z. Roseburia intestinalis: A possible target for vascular calcification. Heliyon 2024; 10:e39865. [PMID: 39524709 PMCID: PMC11550659 DOI: 10.1016/j.heliyon.2024.e39865] [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: 01/21/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
With the advancement of metagenomics and metabolomics techniques, the crucial role of the gut microbiome in intestinal, cardiovascular, and metabolic disorders has been extensively explored. Vascular calcification (VC) is common in atherosclerosis, hypertension, diabetes mellitus, and chronic kidney disease. Moreover, it is a significant cause of cardiovascular diseases and mortality. Roseburia intestinalis, as a promising candidate for the next generation of probiotics, plays a substantial role in inhibiting the systemic inflammatory response and holds great potential in the treatment of intestinal diseases, cardiovascular diseases, and metabolic disorders. Its primary metabolite, butyrate, acts on specific receptors (GPR43, GPR41, GPR109a). It enters cells via transporters (MCT1, SMCT1), affecting gene expression through HDACs, PPARγ and Nrf2, promoting energy metabolism and changing the concentration of other metabolites (including AGEs, LPS, BHB) in the circulation to affect the body's life activities. In this paper, we focus on the possible mechanism of the primary metabolite butyrate of Roseburia intestinalis in inhibiting VC, which may become a potential therapeutic target for the treatment of VC and the ways to enhance its effect.
Collapse
Affiliation(s)
- Xinyun Pan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| | - Yunjian Song
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| | - Yapeng Liang
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Guoquan Feng
- Department of Imaging, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| |
Collapse
|