1
|
Sun Y, Lu J, Tung Lau EY, Zeng Y, Lam Li SW, Au TH, Ye S, Zhou T, Chan FK, Liang JQ. Fusobacterium nucleatum enhances cholesterol biosynthesis in colorectal cancer via miR-130a-3p-mediated AMPK inhibition, a process counteracted by butyrate. Cancer Lett 2025:217810. [PMID: 40414519 DOI: 10.1016/j.canlet.2025.217810] [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: 03/01/2025] [Revised: 04/29/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
Fusobacterium nucleatum (Fn) has been implicated in various diseases, including colorectal cancer (CRC). This study elucidates Fn's contribution to cholesterol synthesis and the underlying link with CRC, as well as butyrate's counteracting effects in this process. Cells and mouse models were treated with Fn followed/accompanied by butyrate treatments to investigate the interplay between butyrate and Fn's oncogenic properties. Transcriptomics analysis pinpointed Fn's profound impact on cholesterol biosynthesis genes and pathways. Fn treatment upregulated the expression of genes involved in cholesterol synthesis (FDPS, FDFT1, and SQLE) and increased SREBF2 activity in cells and mouse intestines, elevating cholesterol levels in cells, intestines, and sera. Fn upregulated miR-130a-3p, identified through transcriptomics and target prediction, through nuclear factor-κB activation. miR-130a-3p subsequently downregulated AMPKα/β1 expression to activate SREBF2 and upregulate cholesterol biosynthesis genes. These effects were predominantly mitigated by butyrate. Notably, analysis of TCGA data revealed that fusobacterial abundance correlated significantly with the expression of FDPS, FDFT1, SQLE, and AMPKα/β1 in CRC. Fn abundance and miRNA expression in human stools were quantified using qPCR and RT-qPCR. Fecal miR-130a-3p levels increased progressively from normal subjects through adenoma patients to CRC patients, correlating significantly with fecal Fn abundance. Additionally, heightened fecal Fn abundance was associated with an increased incidence of hypercholesterolemia in CRC patients. Fn promotes cholesterol biosynthesis by upregulating miR-130a-3p, which downregulates AMPK proteins and activates SREBF2. This study highlights the influence of gut bacteria on host cholesterol synthesis. Targeted modulation of gut microbiota to reduce cholesterol may represent a promising preventive strategy for CRC.
Collapse
Affiliation(s)
- Yuting Sun
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jiawei Lu
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Effie Yin Tung Lau
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Yao Zeng
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Sarah Wing Lam Li
- Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Ting Hei Au
- Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Silin Ye
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Tingyu Zhou
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Francis Kl Chan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jessie Qiaoyi Liang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
2
|
Luo X, Wang K, Jiang C. Gut microbial enzymes and metabolic dysfunction-associated steatohepatitis: Function, mechanism, and therapeutic prospects. Cell Host Microbe 2025:S1931-3128(25)00153-2. [PMID: 40425014 DOI: 10.1016/j.chom.2025.04.020] [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: 02/10/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent liver disease worldwide. The liver communicates with the intestine, in large part through the gut microbiota. Microbial enzymes are key mediators that affect the progression of MASLD and the more severe metabolic dysfunction-associated steatohepatitis (MASH). These enzymes contribute to the metabolism or biosynthesis of steroids, fatty acids, amino acids, ethanol, choline, and intestinal hormones that contribute to disease progression. Additionally, dysbiosis and functional alterations in the microbiota compromise the intestinal barrier, increasing its permeability to bacterial metabolites and liver exposure to microbial-associated molecular patterns (MAMPs), thereby exacerbating liver inflammation and fibrosis. Furthermore, functional alterations in the gut microbiota can modulate intestinal signaling pathways through metabolites or gut hormones, subsequently affecting hepatic metabolism. A deeper understanding of the roles of the gut microbiota and microbial enzymes in MASH will facilitate the development of personalized treatments targeting specific gut microbes or functional enzymes.
Collapse
Affiliation(s)
- Xi Luo
- Department of Physiology and Pathophysiology, Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Kai Wang
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China.
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing 100191, China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
3
|
Netter U, Bisht V, Gaurav A, Sharma R, Ghosh A, Bisht VS, Ambatipudi K, Sharma HP, Mohanty S, Loat S, Sarkar M, Tahlan K, Navani NK. Discovery and mechanistic characterization of a probiotic-origin 3β-OH-Δ 5-6-cholesterol-5β-reductase directly converting cholesterol to coprostanol. FEBS J 2025. [PMID: 40364602 DOI: 10.1111/febs.70131] [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: 12/05/2024] [Revised: 03/21/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025]
Abstract
Cholesterol serves as a fundamental molecule in various structural and biochemical pathways; however, high cholesterol levels are linked to cardiovascular diseases. Some selected strains of Lactobacilli are known for modulating cholesterol levels. However, the molecular mechanism underlying cholesterol transformation by lactobacilli has remained elusive. This study describes the discovery and function of a microbial 3β-OH-Δ5-6-cholesterol-5β-reductase (5βChR) from Limosilactobacillus fermentum NKN51, which directly converts cholesterol to coprostanol, thereby unraveling this longstanding mystery. Protein engineering of the reductase enzyme identified the cholesterol and NADPH interacting amino acid residues, detailing the catalytic mechanism of 5βChR. Phylogenetic analyses highlight the prevalence of 5βChRs in gut commensal lactobacilli, which share a common evolutionary origin with plant 5β reductases. Meta-analysis of microbiomes from healthy individuals underscores the importance of 5βChR homologs, while a cohort study demonstrates an inverse association between 5βChR abundance and diabetes. The discovery of the 5βChR enzyme and its molecular mechanism in cholesterol metabolism paves the way for a better understanding of the gut-associated microbiome and the design of practical applications to ameliorate dyslipidemia.
Collapse
Affiliation(s)
- Urmila Netter
- Chemical Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Vishakha Bisht
- Chemical Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Amit Gaurav
- Chemical Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Rekha Sharma
- Chemical Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Avik Ghosh
- Chemical Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Vinod Singh Bisht
- Proteomics and Lipidomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Kiran Ambatipudi
- Proteomics and Lipidomics Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| | - Hanuman Prasad Sharma
- Centralized Core Research Facility, All India Institute of Medical Sciences, New Delhi, India
| | - Sujata Mohanty
- Stem Cell Facility (DBT-Centre of Excellence for Stem Cell Research), All India Institute of Medical Sciences, New Delhi, India
| | - Shubham Loat
- ICAR-National Research Centre on Yak, Dirang, India
| | - Mihir Sarkar
- ICAR-National Research Centre on Yak, Dirang, India
| | - Kapil Tahlan
- Department of Biology, Memorial University of Newfoundland and Labrador, St. John's, Canada
| | - Naveen K Navani
- Chemical Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology, Roorkee, India
| |
Collapse
|
4
|
Arp G, Levy S, Hall B. Linking bacterial androgen production and prostate cancer. Nat Microbiol 2025; 10:1038-1039. [PMID: 40259018 DOI: 10.1038/s41564-025-01996-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Affiliation(s)
- Gabriela Arp
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, MD, USA
| | - Sophia Levy
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, MD, USA
| | - Brantley Hall
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, MD, USA.
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, College Park, MD, USA.
| |
Collapse
|
5
|
Xiong S. Gut-Microbiota-Driven Lipid Metabolism: Mechanisms and Applications in Swine Production. Metabolites 2025; 15:248. [PMID: 40278377 PMCID: PMC12029090 DOI: 10.3390/metabo15040248] [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: 03/12/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: The gut microbiota plays a pivotal role in host physiology through metabolite production, with lipids serving as essential biomolecules for cellular structure, metabolism, and signaling. This review aims to elucidate the interactions between gut microbiota and lipid metabolism and their implications for enhancing swine production. Methods: We systematically analyzed current literature on microbial lipid metabolism, focusing on mechanistic studies on microbiota-lipid interactions, key regulatory pathways in microbial lipid metabolism, and multi-omics evidence (metagenomic/metabolomic) from swine models. Results: This review outlines the structural and functional roles of lipids in bacterial membranes and examines the influence of gut microbiota on the metabolism of key lipid classes, including cholesterol, bile acids, choline, sphingolipids, and fatty acids. Additionally, we explore the potential applications of microbial lipid metabolism in enhancing swine production performance. Conclusions: Our analysis establishes a scientific framework for microbiota-based strategies to optimize lipid metabolism. The findings highlight potential interventions to improve livestock productivity through targeted manipulation of gut microbial communities.
Collapse
Affiliation(s)
- Shuqi Xiong
- National Key Laboratory of Pig Genetic Improvement and Germplasm Innovation, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang 330045, China
| |
Collapse
|
6
|
Lu X, Xu Y, Liu Y, Li F, Feng Q, Gao C, Liu D, Zhou L, Yang H, Zhang J, Cui F, Chen Q. Neutrophil Depletion Reduced the Relative Abundance of Unsaturated Long-Chain Fatty Acid Synthesis Microbiota and Intestinal Lipid Absorption. Cell Biochem Funct 2025; 43:e70060. [PMID: 40016914 DOI: 10.1002/cbf.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/08/2025] [Accepted: 02/12/2025] [Indexed: 03/01/2025]
Abstract
As immune cells, neutrophils serve as the first line of defense against infections; however, the mechanism by which neutrophils regulate lipid metabolism is unknown. The neutrophil depletion group was treated with 100 μg InVivoMAb anti-mouse Ly6G 6 times, whereas the control group mice were intraperitoneally injected with the same quantity of InVivoMAb rat IgG2a. Body fat content, triglycerides (TGs), total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C) in the jejunum and ileum, as well as 9 long-chain fatty acids (LCFAs) in the intestinal contents were significantly decreased. Furthermore, genes involved in the absorption of lipids in each segment of the intestine also showed decreased expression. Neutrophil-depletion and control models were administered 25 μCi of 3H-cholesterol by gavage. The distribution of 3H cholesterol in the intestinal segment, heart, liver, serum, and feces was not altered by anti-Ly6G antibodies. Metagenomics was applied to investigate uncultured microorganisms in the intestinal contents to identify bacteria containing lipid metabolism genes. At the species level, 12 bacteria were involved in unsaturated LCFA synthesis, among which 2 increased and 10 decreased. The overall relative abundance of these bacteria decreased from 3.102% to 0.734%. Many genes involved in lipid metabolism were also reduced as a result, such as fatty acid synthase and peroxisome proliferator-activated receptor γ. In conclusion, neutrophil depletion does not affect intestinal lipid absorption in the diet but leads to a decrease in the overall relative abundance of gut bacteria involved in unsaturated LCFA synthesis. Consequently, intestinal lipid synthesis and absorption are reduced.
Collapse
Affiliation(s)
- Xingyu Lu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Yike Xu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Yitong Liu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Fang Li
- School of Biology & Basic Medical Sciences, Soochow University, Suzhou, China
| | - Qiong Feng
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Chun Gao
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dan Liu
- Health Management Center, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Zhou
- Nutriology Department, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haizhen Yang
- Health Management Center, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ji Zhang
- Ophthalmology Department, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Fengmei Cui
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Qiu Chen
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Collaborative Innovation Center of Radiological Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| |
Collapse
|
7
|
Ji Y, Sun H, Wang Y, Li Y, Piao R, Bu L, Xu H. Characterizing the oral and gastrointestinal microbiome associated with healthy aging: insights from long-lived populations in Northeastern China. GeroScience 2025; 47:2275-2292. [PMID: 39505797 PMCID: PMC11978580 DOI: 10.1007/s11357-024-01419-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 10/29/2024] [Indexed: 11/08/2024] Open
Abstract
The oral and gastrointestinal (GI) tract microbiota in humans is susceptible to geographical influences and represents vital factors impacting healthy aging. The northeastern region of China, characterized by distinct dietary and climatic conditions, significantly influences the human microbiome composition. However, the microbial structure of the entire long-lived population in this area has not been evaluated. This study recruited a cohort of 142 individuals aged 55-102 residing in Northeast China, and their oral and gut microbiota were evaluated using full-length 16S rRNA gene amplicon sequencing. The results indicate that the oral and GI tract microbiota of long-lived individuals showed reduced microbial taxonomic richness and evenness compared to sub-longevity individuals. With aging, the core species experience a gradual decline in abundance, while subordinate species show an increase. The long-lived population exhibited a heightened ability to enrich beneficial bacteria including Akkermansia, Alistipes, Parabacteroides, and Eubacterium coprostanoligenes in the GI tract, which are associated with host metabolism and have the potential to act as probiotics, reducing the risks of unhealthy aging in the northeast population. Bifidobacterium sp. and Lactobacillus salivarius have been found to coexist in both the oral cavity and the GI tract of long-lived individuals. We hypothesize that beneficial bacterial taxa from the oral cavity colonize the GI tract more extensively in long-lived individuals compared to those with a shorter lifespan. These findings pave the way for identifying probiotic strains that can promote healthy aging in Northeast China.
Collapse
Affiliation(s)
- Yue Ji
- Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China
| | - Hao Sun
- CAS Key Laboratory of Forest Ecology and Management, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, 110016, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yingda Wang
- Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China
| | - Yanhui Li
- Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China
| | - Rennv Piao
- Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China
| | - Li Bu
- Jinqiu Hospital of Liaoning Province, Shenyang, 110016, China.
| | - Hui Xu
- CAS Key Laboratory of Forest Ecology and Management, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, 110016, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
8
|
Chen L, Wang X, Wang S, Liu W, Song Z, Liao H. The impact of gut microbiota on the occurrence, treatment, and prognosis of ischemic stroke. Neurobiol Dis 2025; 207:106836. [PMID: 39952411 DOI: 10.1016/j.nbd.2025.106836] [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: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025] Open
Abstract
Ischemic stroke (IS) is a cerebrovascular disease that predominantly affects middle-aged and elderly populations, exhibiting high mortality and disability rates. At present, the incidence of IS is increasing annually, with a notable trend towards younger affected individuals. Recent discoveries concerning the "gut-brain axis" have established a connection between the gut and the brain. Numerous studies have revealed that intestinal microbes play a crucial role in the onset, progression, and outcomes of IS. They are involved in the entire pathophysiological process of IS through mechanisms such as chronic inflammation, neural regulation, and metabolic processes. Although numerous studies have explored the relationship between IS and intestinal microbiota, comprehensive analyses of specific microbiota is relatively scarce. Therefore, this paper provides an overview of the typical changes in gut microbiota following IS and investigates the role of specific microorganisms in this context. Additionally, it presents a comprehensive analysis of post-stroke microbiological therapy and the relationship between IS and diet. The aim is to identify potential microbial targets for therapeutic intervention, as well as to highlight the benefits of microbiological therapies and the significance of dietary management. Overall, this paper seeks to provide key strategies for the treatment and management of IS, advocating for healthy diets and health programs for individuals. Meanwhile, it may offer a new perspective on the future interdisciplinary development of neurology, microbiology and nutrition.
Collapse
Affiliation(s)
- Liying Chen
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Xi Wang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Shiqi Wang
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | - Weili Liu
- Geriatric Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China
| | | | - Huiling Liao
- Neurology Department, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
9
|
Yan Z, Guan G, Jia H, Li H, Zhuoga S, Zheng S. The association between gut microbiota and accelerated aging and frailty: a Mendelian randomization study. Aging Clin Exp Res 2025; 37:82. [PMID: 40074999 PMCID: PMC11903541 DOI: 10.1007/s40520-025-02971-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 02/12/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND The recent observational studies have unveiled the correlation between the composition and dynamic alterations of the gut microbiome and aging; however, the causal relationship remains uncertain. AIMS The objective of this study is to investigate the causal relationship between the gut microbiome and accelerated aging as well as frailty, from a genetic perspective. METHODS We obtained data on the gut microbiome, intrinsic epigenetic age acceleration, and Frailty Index from published large-scale genome-wide association studies. A two-sample Mendelian randomization analysis was conducted primarily using inverse variance weighting model. We utilized the MR-Egger intercept analysis, IVW method, the Cochran Q test, and the leave-one-out analysis to assess the robustness of the results. RESULTS IVW analysis indicated a potential association between Peptococcus (OR: 1.231, 95% CI 1.013-1.497, P = 0.037), Dialister (OR: 1.447, 95% CI 1.078-1.941, P = 0.014) and Subdoligranulum (OR: 1.538, 95% CI 1.047-2.257, P = 0.028) with intrinsic epigenetic age acceleration; while Prevotella 7 (OR: 0.792, 95% CI 0.672-0.935, P = 0.006) was associated with a potential protective effect. Allisonella (OR: 1.033, 95% CI 1.005-1.063, P = 0.022), Howardella (OR: 1.026, 95% CI 1.002-1.050, P = 0.031) and Eubacterium coprostanoligenes (OR: 1.037, 95% CI 1.001-1.073, P = 0.042) were associated with an increased risk of frailty; conversely, Flavonifractor (OR: 0.954, 95% CI 0.920-0.990, P = 0.012) and Victivallis (OR: 0.984, 95% CI 0.968-1.000, P = 0.049) appeared to exhibit a potential protective effect against frailty. CONCLUSION The findings of this study provide further evidence for the genetic correlation between gut microbiota and accelerated aging as well as frailty, enhancing the understanding of the role of gut microbiota in aging-related processes. However, the underlying mechanisms and potential clinical applications require further investigation before any targeted interventions can be developed.
Collapse
Affiliation(s)
- Zhiliang Yan
- Department of Gastroenterology, Huadong Hospital, Fudan University, Shanghai, China
| | - Guoyu Guan
- Department of Geriatrics, Huadong Hospital, Fudan University, Shanghai, China
| | - Hanqi Jia
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, China
| | - Hanyu Li
- Department of Gastroenterology, Huadong Hospital, Fudan University, Shanghai, China
| | - Sangdan Zhuoga
- Department of Gastroenterology, Huadong Hospital, Fudan University, Shanghai, China
| | - Songbai Zheng
- Department of Gastroenterology, Huadong Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Tang J, Xu W, Yu Y, Yin S, Ye BC, Zhou Y. The role of the gut microbial metabolism of sterols and bile acids in human health. Biochimie 2025; 230:43-54. [PMID: 39542125 DOI: 10.1016/j.biochi.2024.11.003] [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: 04/22/2024] [Revised: 10/31/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Sterols and bile acids are vital signaling molecules that play key roles in systemic functions, influencing the composition of the human gut microbiota, which maintains a symbiotic relationship with the host. Additionally, gut microbiota-encoded enzymes catalyze the conversion of sterols and bile acids into various metabolites, significantly enhancing their diversity and biological activities. In this review, we focus on the microbial transformations of sterols and bile acids in the gut, summarize the relevant bacteria, genes, and enzymes, and review the relationship between the sterols and bile acids metabolism of gut microbiota and human health. This review contributes to a deeper understanding of the crucial roles of sterols and bile acids metabolism by gut microbiota in human health, offering insights for further investigation into the interactions between gut microbiota and the host.
Collapse
Affiliation(s)
- Jiahui Tang
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Wenwu Xu
- Institute of Animal Husbandry and Veterinary Medicine, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yangfan Yu
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Shengxiang Yin
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Bang-Ce Ye
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Yunyan Zhou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China.
| |
Collapse
|
11
|
Gu J, Mu W, Xu Y, Nie Y. From discovery to application: Enabling technology-based optimizing carbonyl reductases biocatalysis for active pharmaceutical ingredient synthesis. Biotechnol Adv 2025; 79:108496. [PMID: 39647674 DOI: 10.1016/j.biotechadv.2024.108496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/04/2024] [Accepted: 11/30/2024] [Indexed: 12/10/2024]
Abstract
The catalytic conversion of chiral alcohols and corresponding carbonyl compounds by carbonyl reductases (alcohol dehydrogenases), which are NAD(P) or NAD(P)H-dependent oxidoreductases, has attracted considerable attention. However, existing carbonyl reductases are insufficient to meet the demands of diverse industrial applications; hence, new enzymes with functions that can expand the toolbox of biocatalysts are urgently required. Developing precisely controlled chiral biocatalysts is of great significance for the efficient development of a broad spectrum of active pharmaceutical ingredients via biosynthesis. In this review, we summarized methods for discovering novel natural carbonyl reductases from various perspectives. Furthermore, advances in protein engineering, utilizing known sequence and structural information as well as catalytic dynamics mechanisms to improve potential functions, are also addressed. The exponential growth in data-driven tools over the past decade has made it possible to de novo design carbonyl reductases. Additionally, various applications of these high-performance carbonyl reductases and different strategies for coenzyme regeneration involving photocatalysis during the reaction process were reviewed. These advancements will bring new opportunities and challenges to the fields of green chemistry and biosynthesis in the future.
Collapse
Affiliation(s)
- Jie Gu
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wanmeng Mu
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China; State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yan Xu
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China; State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yao Nie
- Lab of Brewing Microbiology and Applied Enzymology, School of Biotechnology and Key laboratory of Industrial Biotechnology of Ministry of Education, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
12
|
Luo J, Wang Y. Precision Dietary Intervention: Gut Microbiome and Meta-metabolome as Functional Readouts. PHENOMICS (CHAM, SWITZERLAND) 2025; 5:23-50. [PMID: 40313608 PMCID: PMC12040796 DOI: 10.1007/s43657-024-00193-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/25/2024] [Accepted: 08/02/2024] [Indexed: 05/03/2025]
Abstract
Gut microbiome, the group of commensals residing within the intestinal tract, is closely associated with dietary patterns by interacting with food components. The gut microbiome is modifiable by the diet, and in turn, it utilizes the undigested food components as substrates and generates a group of small molecule-metabolites that addressed as "meta-metabolome" in this review. Profiling and mapping of meta-metabolome could yield insightful information at higher resolution and serve as functional readouts for precision nutrition and formation of personalized dietary strategies. For assessing the meta-metabolome, sample preparation is important, and it should aim for retrieval of gut microbial metabolites as intact as possible. The meta-metabolome can be investigated via untargeted and targeted meta-metabolomics with analytical platforms such as nuclear magnetic resonance spectroscopy and mass spectrometry. Employing flux analysis with meta-metabolomics using available database could further elucidate metabolic pathways that lead to biomarker discovery. In conclusion, integration of gut microbiome and meta-metabolomics is a promising supplementary approach to tailor precision dietary intervention. In this review, relationships among diet, gut microbiome, and meta-metabolome are elucidated, with an emphasis on recent advances in alternative analysis techniques proposed for nutritional research. We hope that this review will provide information for establishing pipelines complementary to traditional approaches for achieving precision dietary intervention.
Collapse
Affiliation(s)
- Jing Luo
- Chair of Nutrition and Immunology, TUM School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
- TUMCREATE, 1 Create Way, #10-02 CREATE Tower, Singapore, 138602 Singapore
| | - Yulan Wang
- Singapore Phenome Centre, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 636921 Singapore
| |
Collapse
|
13
|
Liu Y, Li H, Sun T, Sun G, Jiang B, Liu M, Wang Q, Li T, Cao J, Zhao L, Xiao F, Zhao F, Cui H. Gut microbiome and metabolome characteristics of patients with cholesterol gallstones suggest the preventive potential of prebiotics. IMETA 2025; 4:e70000. [PMID: 40027485 PMCID: PMC11865347 DOI: 10.1002/imt2.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 03/05/2025]
Abstract
Cholesterol gallstones (CGS) still lack effective noninvasive treatment. The etiology of experimentally proven cholesterol stones remains underexplored. This cross-sectional study aims to comprehensively evaluate potential biomarkers in patients with gallstones and assess the effects of microbiome-targeted interventions in mice. Microbiome taxonomic profiling was conducted on 191 samples via V3-V4 16S rRNA sequencing. Next, 60 samples (30 age- and sex-matched CGS patients and 30 controls) were selected for metagenomic sequencing and fecal metabolite profiling via liquid chromatography-mass spectrometry. Microbiome and metabolite characterizations were performed to identify potential biomarkers for CGS. Eight-week-old male C57BL/6J mice were given a lithogenic diet for 8 weeks to promote gallstone development. The causal relationship was examined through monocolonization in antibiotics-treated mice. The effects of short-chain fatty acids such as sodium butyrate, sodium acetate (NaA), sodium propionate, and fructooligosaccharides (FOS) on lithogenic diet-induced gallstones were investigated in mice. Gut microbiota and metabolites exhibited distinct characteristics, and selected biomarkers demonstrated good diagnostic performance in distinguishing CGS patients from healthy controls. Multi-omics data indicated associations between CGS and pathways involving butanoate and propanoate metabolism, fatty acid biosynthesis and degradation pathways, taurine and hypotaurine metabolism, and glyoxylate and dicarboxylate metabolism. The incidence of gallstones was significantly higher in the Clostridium glycyrrhizinilyticum group compared to the control group in mice. The grade of experimental gallstones in control mice was significantly higher than in mice treated with NaA and FOS. FOS could completely inhibit the formation of gallstones in mice. This study characterized gut microbiome and metabolome alterations in CGS. C. glycyrrhizinilyticum contributed to gallstone formation in mice. Supplementing with FOS could serve as a potential approach for managing CGS by altering the composition and functionality of gut microbiota.
Collapse
Affiliation(s)
- Ye Liu
- Peking University Fifth School of Clinical MedicineBeijing Hospital, National Center of GerontologyBeijingChina
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Hexin Li
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Tianhan Sun
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Gaoyuan Sun
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Boyue Jiang
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Meilan Liu
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Qing Wang
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Tong Li
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Jianfu Cao
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Li Zhao
- Department of Gastroenterology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingChina
| | - Fei Xiao
- Peking University Fifth School of Clinical MedicineBeijing Hospital, National Center of GerontologyBeijingChina
- Clinical Biobank, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Beijing Hospital, National Center of Gerontology, National Health Commission, Institute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Fangqing Zhao
- Institute of Zoology, Chinese Academy of SciencesBeijingChina
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, Beijing Institutes of Life ScienceChinese Academy of SciencesBeijingChina
| | - Hongyuan Cui
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| |
Collapse
|
14
|
Uthaiah NM, Venkataramareddy SR, Mudhol S, Sheikh AY. EPA-rich Nannochloropsis oceanica biomass regulates gut microbiota, alleviates inflammation and ameliorates liver fibrosis in rats. Food Res Int 2025; 202:115733. [PMID: 39967180 DOI: 10.1016/j.foodres.2025.115733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/14/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025]
Abstract
Omega-3 fatty acids are believed to show anti-fibrotic effects by lowering inflammation and regulating the gut microflora. Marine microalgae are an alternative, sustainable source of omega-3 fatty acids to the conventionally used fish oil. Microalgae N. oceanica is a promising source of EPA, one of the essential omega-3 PUFAs. Current study investigates the inhibitory effects of EPA rich N. oceanica biomass against CCl4 induced liver fibrosis in rats. Here, we studied the anti-fibrotic effects in N. oceanica biomass fed groups: T1 - Low dose (4.16 mg/kg EPA), T2 - Medium dose (8.33 mg/kg EPA) and T3 - High dose (16.66 mg/kg EPA), when compared to fish oil fed group (FO - 16.66 mg/kg EPA) as a positive control. The elevated levels of serum liver biomarker enzymes and cholesterol induced by CCl4 showed a significant reduction in T3. Histopathological analysis showed the protective effects of biomass feeding on inflammation and hepatocyte degeneration. In addition, the abundance of the SCFA producing bacteria like Blautia argi, Romboutsia ilealis, Romboutsia timonensis, Stomatobaculum longum and Limosilactobacillus reuteri markedly increased in the PUFA fed groups. The cholesterol metabolising bacteria Eubacterium coprostanoligenes showed a noteworthy increase upon PUFA administration. Overall results indicate that the ameliorative effects observed upon administration of N. oceanica biomass were comparable to FO in a dose dependent manner. Therefore, we can conclude that N. oceanica biomass supplementation is associated with the alleviation of liver fibrosis in rats.
Collapse
Affiliation(s)
- Nethravathy Malachira Uthaiah
- Plant Cell Biotechnology Department, CSIR - Central Food Technological Research Institute, Mysuru 570020 Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sreedhar Reddampalli Venkataramareddy
- Plant Cell Biotechnology Department, CSIR - Central Food Technological Research Institute, Mysuru 570020 Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Seema Mudhol
- Department of Biochemistry, CSIR - Central Food Technological Research Institute, Mysuru 570020 Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Azam Yakub Sheikh
- Plant Cell Biotechnology Department, CSIR - Central Food Technological Research Institute, Mysuru 570020 Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
15
|
Zheng L, Ye ZY, Ma JJ. Effect of cholesterol metabolism on hepatolithiasis. World J Gastroenterol 2025; 31:99960. [PMID: 39777239 PMCID: PMC11684189 DOI: 10.3748/wjg.v31.i1.99960] [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: 08/04/2024] [Revised: 10/03/2024] [Accepted: 11/05/2024] [Indexed: 12/09/2024] Open
Abstract
Surgical intervention is currently the primary treatment for hepatolithiasis; however, some patients still experience residual stones and high recurrence rates after surgery. Cholesterol metabolism seems to play an important role in hepatolithiasis pathogenesis. A high cholesterol diet is one of the significant reasons for the increasing incidence of hepatolithiasis. Therefore, regular diet and appropriate medical intervention are crucial measures to prevent hepatolithiasis and reduce recurrence rate after surgery. Reducing dietary cholesterol and drugs that increase cholesterol stone solubility are key therapeutic approaches in treating hepatolithiasis. This article discusses the cholesterol metabolic pathways related to the pathogenesis of hepatolithiasis, as well as food intake and targeted therapeutic drugs.
Collapse
Affiliation(s)
- Lin Zheng
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215 Heping West Road, Shijiazhuang 050000, Hebei Province, China
| | - Zi-Yu Ye
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215 Heping West Road, Shijiazhuang 050000, Hebei Province, China
| | - Jun-Ji Ma
- Department of Gastroenterology, The Second Hospital of Hebei Medical University, Hebei Key Laboratory of Gastroenterology, Hebei Institute of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, No. 215 Heping West Road, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|
16
|
Chou JC, Dassama LMK. Lipid Trafficking in Diverse Bacteria. Acc Chem Res 2025; 58:36-46. [PMID: 39680024 PMCID: PMC11713862 DOI: 10.1021/acs.accounts.4c00540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
Lipids are essential for life and serve as cell envelope components, signaling molecules, and nutrients. For lipids to achieve their required functions, they need to be correctly localized. This requires the action of transporter proteins and an energy source. The current understanding of bacterial lipid transporters is limited to a few classes. Given the diversity of lipid species and the predicted existence of specific lipid transporters, many more transporters await discovery and characterization. These proteins could be prime targets for modulators that control bacterial cell proliferation and pathogenesis. One overarching goal of our research is to understand the molecular mechanisms of bacterial metabolite trafficking, including lipids, and to leverage that understanding to identify or engineer inhibitory ligands. In recent years, our work has revealed two novel lipid transport systems in bacteria: bacterial sterol transporters (Bst) A, B, and C in Methylococcus capsulatus and the TatT proteins in Enhygromyxa salina and Treponema pallidum. Both systems are composed of transporters bioinformatically identified as being involved in the transport of other metabolites, but substrates were never revealed. However, the genetic colocalization of the genes encoding BstABC with sterol biosynthetic enzymes in M. capsulatus suggested that they might recognize sterols as substrates. Also, homologues of TatTs are present in diverse bacteria but are overrepresented in bacteria deficient in de novo lipid synthesis or residing in nutrient-poor environments; we reasoned that these proteins might facilitate the transport of lipids. Our efforts to reveal the substrate scope of two TatT proteins revealed their engagement with long-chain fatty acids. Enabling the discovery of the BstABC system and the TatT proteins were bioinformatic analyses, quantitative measurements of protein-ligand equilibrium affinities, and high-resolution structural studies that provided remarkable insights into ligand binding cavities and the structural basis for ligand interaction. These approaches, in particular our bioinformatics and structural work, highlighted the diversity of protein sequence and structures amenable to lipid engagement. These observations allowed the hypothesis that lipid handling proteins, in general and especially so in the bacterial domain, can have diverse amino acid compositions and three-dimensional structures. As such, bioinformatics geared at identifying them in poorly characterized genomes is likely to miss many candidates that diverge from well-characterized family members. This realization spurred efforts to understand the unifying features in all of the lipid handling proteins we have characterized to date. To do this, we inspected the ligand binding sites of the proteins: they were remarkably hydrophobic and sometimes displayed a dichotomy of hydrophobic and hydrophilic amino acids, akin to the ligands that they accommodate in those cavities. Because of this, we reasoned that the physicochemical features of ligand binding cavities could be accurate predictors of a protein's propensity to bind lipids. This finding was leveraged to create structure-based lipid-interacting pocket predictor (SLiPP), a machine-learning algorithm capable of identifying ligand cavities with physico-chemical features consistent with those of known lipid binding sites. SLiPP is especially useful in poorly annotated genomes (such as with bacterial pathogens), where it could reveal candidate proteins to be targeted for the development of antimicrobials.
Collapse
Affiliation(s)
- Jonathan
Chiu-Chun Chou
- Department
of Chemistry and Sarafan ChEM-H Institute, Stanford University, Stanford, California 94305, United States
| | - Laura M. K. Dassama
- Department
of Chemistry and Sarafan ChEM-H Institute, Stanford University, Stanford, California 94305, United States
- Department
of Microbiology and Immunology, Stanford
School of Medicine, Stanford, California 94305, United States
| |
Collapse
|
17
|
Xu K, Motiwala Z, Corona-Avila I, Makhanasa D, Alkahalifeh L, Khan MW. The Gut Microbiome and Its Multifaceted Role in Cancer Metabolism, Initiation, and Progression: Insights and Therapeutic Implications. Technol Cancer Res Treat 2025; 24:15330338251331960. [PMID: 40208053 PMCID: PMC12032467 DOI: 10.1177/15330338251331960] [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: 10/30/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 04/11/2025] Open
Abstract
This review summarizes the intricate relationship between the microbiome and cancer initiation and development. Microbiome alterations impact metabolic pathways, immune responses, and gene expression, which can accelerate or mitigate cancer progression. We examine how dysbiosis affects tumor growth, metastasis, and treatment resistance. Additionally, we discuss the potential of microbiome-targeted therapies, such as probiotics and fecal microbiota transplants, to modulate cancer metabolism. These interventions offer the possibility of reversing or controlling cancer progression, enhancing the efficacy of traditional treatments like chemotherapy and immunotherapy. Despite promising developments, challenges remain in identifying key microbial species and pathways and validating microbiome-targeted therapies through large-scale clinical trials. Nonetheless, the intersection of microbiome research and cancer initiation and development presents an exciting frontier for innovative therapies. This review offers a fresh perspective on cancer initiation and development by integrating microbiome insights, highlighting the potential for interdisciplinary research to enhance our understanding of cancer progression and treatment strategies.
Collapse
Affiliation(s)
- Kai Xu
- Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
| | - Zainab Motiwala
- Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, India
| | - Irene Corona-Avila
- Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
| | - Dhruvi Makhanasa
- Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
| | | | - Md. Wasim Khan
- Division of Endocrinology, Diabetes and Metabolism, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
18
|
Strandberg TE, Kovanen PT, Lloyd-Jones DM, Raal FJ, Santos RD, Watts GF. Drugs for dyslipidaemia: the legacy effect of the Scandinavian Simvastatin Survival Study (4S). Lancet 2024; 404:2462-2475. [PMID: 39577453 DOI: 10.1016/s0140-6736(24)02089-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 11/24/2024]
Abstract
Since the discovery of statins and the Scandinavian Simvastatin Survival Study (4S) results three decades ago, remarkable advances have been made in the treatment of dyslipidaemia, a major risk factor for atherosclerotic cardiovascular disease. Safe and effective statins remain the cornerstone of therapeutic approach for this indication, including for children with genetic dyslipidaemia, and are one of the most widely prescribed drugs in the world. However, despite the affordability of generic statins, they remain underutilised worldwide. The use of ezetimibe to further decrease plasma LDL cholesterol and the targeting of other atherogenic lipoproteins, such as triglyceride-rich lipoproteins and lipoprotein(a), are likely to be required to further reduce atherosclerotic cardiovascular disease events. Drugs directed at these lipoproteins, including gene silencing and editing methods that durably suppress the production of proteins, such as PCSK9 and ANGPTL3, open novel therapeutic options to further reduce the development of atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Timo E Strandberg
- University of Helsinki and Helsinki University Hospital, Helsinki, Finland; University of Oulu, Center for Life Course Health Research, Oulu, Finland.
| | | | - Donald M Lloyd-Jones
- Department of Preventive Medicine and Department of Medicine (Cardiology), Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Frederick J Raal
- Division of Endocrinology and Metabolism, University of the Witwatersrand, Johannesburg, South Africa
| | - Raul D Santos
- Academic Research Organization, Hospital Israelita Albert Einstein, São Paulo, Brazil; Lipid Clinic Heart Institute (InCor) University of São Paulo Medical School Hospital, São Paulo, Brazil
| | - Gerald F Watts
- School of Medicine, University of Western Australia, Perth, WA, Australia; Cardiometabolic Service, Department of Cardiology and Internal Medicine, Royal Perth Hospital, Perth, WA, Australia
| |
Collapse
|
19
|
Mahayri TM, Atallah E, Fliegerová KO, Mrázek J, Piccolo G, Bovera F, Moniello G. Inclusion of Tenebrio molitor larvae meal in the diet of barbary partridge (Alectoris barbara) improves caecal bacterial diversity and composition. Sci Rep 2024; 14:29600. [PMID: 39609484 PMCID: PMC11604920 DOI: 10.1038/s41598-024-80341-1] [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: 06/07/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
In this study, we investigated the influence of the inclusion of Tenebrio molitor (TM) larvae meal in the diet on the diversity and structure of the bacterial community in the caecal content of Barbary partridges. A total of 36 partridges, selected randomly for slaughter from 54 animals, were divided equally into three treatment groups, including the control group (C) with a diet containing corn-soybean meal and two experimental groups, in which 25% (TM25) and 50% (TM50) of the soybean meal protein was replaced by the meal from TM larvae. After slaughtering, the bacterial community of the 30 caecal samples (10 samples per each experimental group) was analysed by high-throughput sequencing using the V4-V5 region of the 16 S rRNA gene. Alpha diversity showed a higher diversity richness in the TM50 group. Beta diversity showed statistical dissimilarities among the three groups. Firmicutes was the dominant phylum regardless of the diet, with the predominant families Ruminococcaceae and Lachnospiraceae. Clostridia and Faecalibacterium were decreased in both TM groups, Lachnospiraceae was suppressed in the TM50 group, but still this class, genus and family were abundantly present in all samples. Several potentially beneficial genera, such as Bacillus, Ruminococcaceae UCG-009, Oscillibacter and UC1-2E3 (Lachnospiraceae) were increased in the TM50 group. The results showed a beneficial effect of the T. molitor larvae meal on the caecal microbiota of Barbary partridges, particularly in the TM50 group, which showed an increase in bacterial diversity.
Collapse
Affiliation(s)
- Tiziana Maria Mahayri
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics, Czech Academy of Science, Prague, 14220, Czech Republic
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy
| | - Elie Atallah
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy
- Department of Veterinary Medicine and Animal Sciences, University of Milan, via dell'Università 6, Lodi, 26900, Italy
| | - Kateřina Olša Fliegerová
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics, Czech Academy of Science, Prague, 14220, Czech Republic.
| | - Jakub Mrázek
- Laboratory of Anaerobic Microbiology, Institute of Animal Physiology and Genetics, Czech Academy of Science, Prague, 14220, Czech Republic
| | - Giovanni Piccolo
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, via F. Delpino, 1, Napoli, 80137, Italy.
| | - Fulvia Bovera
- Department of Veterinary Medicine and Animal Production, University of Napoli Federico II, via F. Delpino, 1, Napoli, 80137, Italy
| | - Giuseppe Moniello
- Department of Veterinary Medicine, University of Sassari, Sassari, 07100, Italy
| |
Collapse
|
20
|
Zahn LE, Gannon PM, Rajakovich LJ. Iron-sulfur cluster-dependent enzymes and molybdenum-dependent reductases in the anaerobic metabolism of human gut microbes. Metallomics 2024; 16:mfae049. [PMID: 39504489 PMCID: PMC11574389 DOI: 10.1093/mtomcs/mfae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
Metalloenzymes play central roles in the anaerobic metabolism of human gut microbes. They facilitate redox and radical-based chemistry that enables microbial degradation and modification of various endogenous, dietary, and xenobiotic nutrients in the anoxic gut environment. In this review, we highlight major families of iron-sulfur (Fe-S) cluster-dependent enzymes and molybdenum cofactor-containing enzymes used by human gut microbes. We describe the metabolic functions of 2-hydroxyacyl-CoA dehydratases, glycyl radical enzyme activating enzymes, Fe-S cluster-dependent flavoenzymes, U32 oxidases, and molybdenum-dependent reductases and catechol dehydroxylases in the human gut microbiota. We demonstrate the widespread distribution and prevalence of these metalloenzyme families across 5000 human gut microbial genomes. Lastly, we discuss opportunities for metalloenzyme discovery in the human gut microbiota to reveal new chemistry and biology in this important community.
Collapse
Affiliation(s)
- Leah E Zahn
- Department of Chemistry, University of Washington, Seattle, United States
| | - Paige M Gannon
- Department of Chemistry, University of Washington, Seattle, United States
| | | |
Collapse
|
21
|
Guarner F, Bustos Fernandez L, Cruchet S, Damião A, Maruy Saito A, Riveros Lopez JP, Rodrigues Silva L, Valdovinos Diaz MA. Gut dysbiosis mediates the association between antibiotic exposure and chronic disease. Front Med (Lausanne) 2024; 11:1477882. [PMID: 39568738 PMCID: PMC11576192 DOI: 10.3389/fmed.2024.1477882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/22/2024] [Indexed: 11/22/2024] Open
Abstract
Antibiotics are safe, effective drugs and continue to save millions of lives and prevent long-term illness worldwide. A large body of epidemiological, interventional and experimental evidence shows that exposure to antibiotics has long-term negative effects on human health. We reviewed the literature data on the links between antibiotic exposure, gut dysbiosis, and chronic disease (notably with regard to the "developmental origins of health and disease" ("DOHaD") approach). Molecular biology studies show that the systemic administration of antibiotic to infants has a rapid onset but also often a long-lasting impact on the microbial composition of the gut. Along with other environmental factors (e.g., an unhealthy "Western" diet and sedentary behavior), antibiotics induce gut dysbiosis, which can be defined as the disruption of a previously stable, functionally complete microbiota. Gut dysbiosis many harmful long-term effects on health. Associations between early-life exposure to antibiotics have been reported for chronic diseases, including inflammatory bowel disease, celiac disease, some cancers, metabolic diseases (obesity and type 2 diabetes), allergic diseases, autoimmune disorders, atherosclerosis, arthritis, and neurodevelopmental, neurodegenerative and other neurological diseases. In mechanistic terms, gut dysbiosis influences chronic disease through direct effects on mucosal immune and inflammatory pathways, plus a wide array of direct or indirect effects of short-chain fatty acids, the enteric nervous system, peristaltic motility, the production of hormones and neurotransmitters, and the loss of intestinal barrier integrity (notably with leakage of the pro-inflammatory endotoxin lipopolysaccharide into the circulation). To mitigate dysbiosis, the administration of probiotics in patients with chronic disease is often (but not always) associated with positive effects on clinical markers (e.g., disease scores) and biomarkers of inflammation and immune activation. Meta-analyses are complicated by differences in probiotic composition, dose level, and treatment duration, and large, randomized, controlled clinical trials are lacking in many disease areas. In view of the critical importance of deciding whether or not to prescribe antibiotics (especially to children), we suggest that the DOHaD concept can be logically extended to "gastrointestinal origins of health and disease" ("GOHaD") or even "microbiotic origins of health and disease" ("MOHaD").
Collapse
Affiliation(s)
| | - Luis Bustos Fernandez
- Centro Medico Bustos Fernandez, Instituto de Gastroenterologia, Buenos Aires, Argentina
| | - Sylvia Cruchet
- Institute of Nutrition and Food Technology, Universidad de Chile, Santiago, Chile
| | - Adérson Damião
- Department of Gastroenterology, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Aldo Maruy Saito
- Catedra de Pediatria, Hospital Cayetano Heredia, Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | |
Collapse
|
22
|
Skolnick S. Prospecting for para-endogenous anxiolytics in the human microbiome: Some promising pathways. Pharmacol Biochem Behav 2024; 244:173842. [PMID: 39069097 DOI: 10.1016/j.pbb.2024.173842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
The gut microbiome is a vast, variable, and largely unexplored component of human biology that sits at the intersection of heritable and environmental factors, and represents a rich source of novel chemistry that is already known to be compatible with the human body. This alone would make it a promising place to search for new therapeutics, but recent work has also identified gut microbiome abnormalities in patients with a number of psychiatric disorders, including anxiety disorders-suggesting that not only treatments, but cures may lie therein. Here, we'll discuss two known "para-endogenous" anxiolytics-γ-hydroxybutyrate and the neurosteroid allopregnanolone-which have recently been discovered to be produced by the microbiome.
Collapse
Affiliation(s)
- Stephen Skolnick
- Seed Health, Inc., 2100 Abbot Kinney Blvd, Unit G, Venice, CA 90291, United States of America; Swiss Institute for Allergy & Asthma Research, Herman-Burchard Strasse 9, 7265 Davos Wolfgang, Graubünden, Switzerland.
| |
Collapse
|
23
|
Hermanson JB, Tolba SA, Chrisler EA, Leone VA. Gut microbes, diet, and genetics as drivers of metabolic liver disease: a narrative review outlining implications for precision medicine. J Nutr Biochem 2024; 133:109704. [PMID: 39029595 PMCID: PMC11480923 DOI: 10.1016/j.jnutbio.2024.109704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 07/21/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is rapidly increasing in prevalence, impacting over a third of the global population. The advanced form of MASLD, Metabolic dysfunction-associated steatohepatitis (MASH), is on track to become the number one indication for liver transplant. FDA-approved pharmacological agents are limited for MASH, despite over 400 ongoing clinical trials, with only a single drug (resmetirom) currently on the market. This is likely due to the heterogeneous nature of disease pathophysiology, which involves interactions between highly individualized genetic and environmental factors. To apply precision medicine approaches that overcome interpersonal variability, in-depth insights into interactions between genetics, nutrition, and the gut microbiome are needed, given that each have emerged as dynamic contributors to MASLD and MASH pathogenesis. Here, we discuss the associations and molecular underpinnings of several of these factors individually and outline their interactions in the context of both patient-based studies and preclinical animal model systems. Finally, we highlight gaps in knowledge that will require further investigation to aid in successfully implementing precision medicine to prevent and alleviate MASLD and MASH.
Collapse
Affiliation(s)
- Jake B Hermanson
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Samar A Tolba
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA; Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Evan A Chrisler
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Vanessa A Leone
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
24
|
Culp EJ, Nelson NT, Verdegaal AA, Goodman AL. Microbial transformation of dietary xenobiotics shapes gut microbiome composition. Cell 2024; 187:6327-6345.e20. [PMID: 39321800 PMCID: PMC11531382 DOI: 10.1016/j.cell.2024.08.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 02/23/2024] [Accepted: 08/20/2024] [Indexed: 09/27/2024]
Abstract
Diet is a major determinant of gut microbiome composition, and variation in diet-microbiome interactions may contribute to variation in their health consequences. To mechanistically understand these relationships, here we map interactions between ∼150 small-molecule dietary xenobiotics and the gut microbiome, including the impacts of these compounds on community composition, the metabolic activities of human gut microbes on dietary xenobiotics, and interindividual variation in these traits. Microbial metabolism can toxify and detoxify these compounds, producing emergent interactions that explain community-specific remodeling by dietary xenobiotics. We identify the gene and enzyme responsible for detoxification of one such dietary xenobiotic, resveratrol, and demonstrate that this enzyme contributes to interindividual variation in community remodeling by resveratrol. Together, these results systematically map interactions between dietary xenobiotics and the gut microbiome and connect toxification and detoxification to interpersonal differences in microbiome response to diet.
Collapse
Affiliation(s)
- Elizabeth J Culp
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Nora T Nelson
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Andrew A Verdegaal
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Andrew L Goodman
- Department of Microbial Pathogenesis and Microbial Sciences Institute, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
25
|
Hakkak R, Korourian S, Li W, Spray B, Twaddle NC, Randolph CE, Børsheim E, Robeson II MS. Dietary soy protein reverses obesity-induced liver steatosis and alters fecal microbial composition independent of isoflavone level. Front Nutr 2024; 11:1487859. [PMID: 39529929 PMCID: PMC11551038 DOI: 10.3389/fnut.2024.1487859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Metabolic dysfunction-associated steatotic liver disease (MASLD) is a major public health concern that is exacerbated by the obesity pandemic. Dietary interventions have the potential to alleviate obesity-associated MASLD through variable mechanisms, including optimizing the gut microbiota. Previously, we reported that soy protein concentrate (SPC) with low or high levels of isoflavone (LIF or HIF) protected young obese Zucker rats from developing liver steatosis. The current study was designed to test whether SPC-LIF and SPC-HIF diets would reverse liver steatosis and alter fecal microbial composition in adult obese Zucker rats with existing steatosis. Methods Six-week-old male obese Zucker rats (n = 26) were fed a casein control diet (CAS) for 8 weeks and 7 rats were randomly selected and sacrificed to confirm liver steatosis. The remaining rats were randomly assigned to receive CAS, SPC-LIF, or SPC-HIF diet (n = 6-7/group) for an additional 10 weeks. Results Compared to CAS diet, feeding SPC-LIF and SPC-HIF diets resulted in significantly lower liver weight, liver steatosis score, and liver microvesicular score (p < 0.05), but did not lead to difference in body weight, liver macrovesicular score, serum ALT, or serum AST. Isoflavone levels (e.g., LIF vs. HIF) did not affect any of these measurements except in the SPC-HIF group, which had an additional decrease in liver weight (p < 0.05) compared to the SPC-LIF group. The SPC-HIF group also had significantly higher levels of the aglycone forms of daidzein, genistein, and equol as well as the total levels of daidzein, genistein, and equol compared to SPC-LIF or CAS diet fed rats (p < 0.05). The distribution of microbial communities based on measures of beta diversity of both SPC-LIF and SPC-HIF groups were significantly different to that of the CAS group (p ≤ 0.005). Alpha-diversity did not differ between any of the groups. Conclusion Taken together, dietary soy protein can reverse liver steatosis in adult Zucker rats, and the reversal of steatosis is accompanied by alterations in gut microbial composition.
Collapse
Affiliation(s)
- Reza Hakkak
- Department of Dietetics and Nutrition, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children’s Research Institute, Little Rock, AR, United States
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Soheila Korourian
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Wei Li
- Department of Dietetics and Nutrition, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Beverly Spray
- Division of Biostatistics Core, Arkansas Children’s Research Institute, Little Rock, AR, United States
| | - Nathan C. Twaddle
- Division of Biochemical Toxicology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR, United States
| | | | - Elisabet Børsheim
- Arkansas Children’s Research Institute, Little Rock, AR, United States
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Arkansas Children’s Nutrition Center, Little Rock, AR, United States
| | - Michael S. Robeson II
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
26
|
Šardzíková S, Gajewska M, Gałka N, Štefánek M, Baláž A, Garaiová M, Holič R, Świderek W, Šoltys K. Can longer lifespan be associated with gut microbiota involvement in lipid metabolism? FEMS Microbiol Ecol 2024; 100:fiae135. [PMID: 39354675 PMCID: PMC11503954 DOI: 10.1093/femsec/fiae135] [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: 12/01/2023] [Revised: 08/25/2024] [Accepted: 09/30/2024] [Indexed: 10/03/2024] Open
Abstract
Biological aging is linked to altered body composition and reduced neuroactive steroid hormones like dehydroepiandrosterone sulfate (DHEAS), which can stimulate the GABA signaling pathway via gut microbiota. Our study examined the association of gut microbiota with lifespan in mice through comprehensive analysis of its composition and functional involvement in cholesterol sulfate, a precursor of DHEAS, metabolism. We used 16S rRNA and metagenomic sequencing, followed by metabolic pathway prediction and thin layer chromatography and MALDI-TOF cholesterol sulfate identification. Significant increases in bacteria such as Bacteroides, typical for long-lived and Odoribacter and Colidextribacter, specific for short-lived mice were detected. Furthermore, for males (Rikenella and Alloprevotella) and females (Lactobacillus and Bacteroides), specific bacterial groups emerged as predictors (AUC = 1), highlighting sex-specific patterns. Long-lived mice showed a strong correlation of Bacteroides (0.918) with lipid and steroid hormone metabolism, while a negative correlation of GABAergic synapse with body weight (-0.589). We found that several Bacteroides species harboring the sulfotransferase gene and gene cluster for sulfonate donor synthesis are involved in converting cholesterol to cholesterol sulfate, significantly higher in the feces of long-lived individuals. Overall, we suggest that increased involvement of gut bacteria, mainly Bacteroides spp., in cholesterol sulfate synthesis could ameliorate aging through lipid metabolism.
Collapse
Affiliation(s)
- Sára Šardzíková
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia
| | - Marta Gajewska
- Institute of Animal Sciences, Department of Animal Genetics and Conservation, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Norbert Gałka
- Institute of Animal Sciences, Department of Animal Genetics and Conservation, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Matúš Štefánek
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia
| | - Andrej Baláž
- Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava, 842 48 Bratislava, Slovakia
| | - Martina Garaiová
- Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska Cesta 9, 84005 Bratislava, Slovakia
| | - Roman Holič
- Institute of Animal Biochemistry and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dubravska Cesta 9, 84005 Bratislava, Slovakia
| | - Wiesław Świderek
- Institute of Animal Sciences, Department of Animal Genetics and Conservation, Warsaw University of Life Sciences, Ciszewskiego 8, 02-786 Warsaw, Poland
| | - Katarína Šoltys
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, 842 15 Bratislava, Slovakia
| |
Collapse
|
27
|
Qu W, Xu Y, Yang J, Shi H, Wang J, Yu X, Chen J, Wang B, Zhuoga D, Luo M, Liu R. Berberine alters the gut microbiota metabolism and impairs spermatogenesis. Acta Biochim Biophys Sin (Shanghai) 2024; 57:569-581. [PMID: 39420836 PMCID: PMC12040761 DOI: 10.3724/abbs.2024174] [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: 03/19/2024] [Accepted: 07/21/2024] [Indexed: 10/19/2024] Open
Abstract
Berberine (BBR) is used to treat diarrhea clinically. However, its reproductive toxicity is unclear. This study aims to investigate the impact of BBR on the male reproductive system. Intragastric BBR administration for 14 consecutive days results in a significant decrease in the serum testosterone concentration, epididymal sperm concentration, mating rate and fecundity of male mice. Testicular treatment with testosterone propionate (TP) partially reverses the damage caused by BBR to the male reproductive system. Mechanistically, the decrease in Muribaculaceae abundance in the gut microbiota of mice is the principal cause of the BBR-induced decrease in the sperm concentration. Both fecal microbiota transplantation (FMT) and polyethylene glycol (PEG) treatment demonstrate that Muribaculaceae is necessary for spermatogenesis. The intragastric administration of Muribaculaceae intestinale to BBR-treated mice restores the sperm concentration and testosterone levels. Metabolomic analysis reveals that BBR affects arginine and proline metabolism, of which ornithine level is downregulated. Combined analysis via 16S rRNA metagenomics sequencing and metabolomics shows that Muribaculaceae regulates ornithine level. The transcriptomic results of the testes indicate that the expressions of genes related to the low-density lipoprotein receptor (LDLR)-mediated testosterone synthesis pathway decrease after BBR administration. The transcriptional activity of the Ldlr gene in TM3 cells is increased with increased ornithine supplementation in the culture media, leading to increased testosterone synthesis. Overall, this study reveals an association between a BBR-induced decrease in Muribaculaceae abundance and defective spermatogenesis, providing a prospective therapeutic approach for addressing infertility-related decreases in serum testosterone triggered by changes in the gut microbiota composition.
Collapse
Affiliation(s)
- Wei Qu
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhan430071China
| | - Yumin Xu
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhan430071China
| | - Jing Yang
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhan430071China
| | - Hanqing Shi
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhan430071China
| | - Junli Wang
- Reproductive Medicine Centerthe Affiliated Hospital of Youjiang Medical University for NationalitiesBaise53300China
| | - Xinnai Yu
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhan430071China
| | - Jiemin Chen
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhan430071China
| | - Binyi Wang
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhan430071China
| | - Deqing Zhuoga
- Institute of Livestock ResearchTibet Academy of Agriculture and Animal Husbandry ScienceLhasa850000China
| | - Mengcheng Luo
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhan430071China
| | - Rong Liu
- Hubei Provincial Key Laboratory of Developmentally Originated DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhan430071China
| |
Collapse
|
28
|
Fan L, Liu B, Wang Y, Tang B, Xu T, Fu J, Wang C, Liu Y, Ge L, Wei H, Ren W. Intestinal Lactobacillus murinus-derived small RNAs target porcine polyamine metabolism. Proc Natl Acad Sci U S A 2024; 121:e2413241121. [PMID: 39361652 PMCID: PMC11474053 DOI: 10.1073/pnas.2413241121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/21/2024] [Indexed: 10/05/2024] Open
Abstract
Gut microbiota plays a vital role in host metabolism; however, the influence of gut microbes on polyamine metabolism is unknown. Here, we found germ-free models possess elevated polyamine levels in the colon. Mechanistically, intestinal Lactobacillus murinus-derived small RNAs in extracellular vesicles down-regulate host polyamine metabolism by targeting the expression of enzymes in polyamine metabolism. In addition, Lactobacillus murinus delays recovery of dextran sodium sulfate-induced colitis by reducing polyamine levels in mice. Notably, a decline in the abundance of small RNAs was observed in the colon of mice with colorectal cancer (CRC) and human CRC specimens, accompanied by elevated polyamine levels. Collectively, our study identifies a specific underlying mechanism used by intestinal microbiota to modulate host polyamine metabolism, which provides potential intervention for the treatment of polyamine-associated diseases.
Collapse
Affiliation(s)
- Lijuan Fan
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou510642, China
- National Center of Technology Innovation for Pigs, Chongqing402460, China
| | - Bingnan Liu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou510642, China
- National Center of Technology Innovation for Pigs, Chongqing402460, China
| | - Youxia Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou510642, China
- National Center of Technology Innovation for Pigs, Chongqing402460, China
| | - Bin Tang
- Department of Clinical Laboratory, Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, Jiangjin, Chongqing402260, China
| | - Tianqi Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou225009, China
| | - Jian Fu
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou510642, China
- National Center of Technology Innovation for Pigs, Chongqing402460, China
| | - Chuanlong Wang
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou510642, China
- National Center of Technology Innovation for Pigs, Chongqing402460, China
| | - Yuan Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou225009, China
| | - Liangpeng Ge
- National Center of Technology Innovation for Pigs, Chongqing Academy of Animal Sciences, Key Laboratory of Pig Industry Science, Ministry of Agriculture, Chongqing402460, China
| | - Hong Wei
- State Key Laboratory of Agricultural Microbiology, College of Animal Sciences and Technology, Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan430070, China
| | - Wenkai Ren
- State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou510642, China
- National Center of Technology Innovation for Pigs, Chongqing402460, China
| |
Collapse
|
29
|
Van Hul M, Cani PD, Petitfils C, De Vos WM, Tilg H, El-Omar EM. What defines a healthy gut microbiome? Gut 2024; 73:1893-1908. [PMID: 39322314 PMCID: PMC11503168 DOI: 10.1136/gutjnl-2024-333378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 08/16/2024] [Indexed: 09/27/2024]
Abstract
The understanding that changes in microbiome composition can influence chronic human diseases and the efficiency of therapies has driven efforts to develop microbiota-centred therapies such as first and next generation probiotics, prebiotics and postbiotics, microbiota editing and faecal microbiota transplantation. Central to microbiome research is understanding how disease impacts microbiome composition and vice versa, yet there is a problematic issue with the term 'dysbiosis', which broadly links microbial imbalances to various chronic illnesses without precision or definition. Another significant issue in microbiome discussions is defining 'healthy individuals' to ascertain what characterises a healthy microbiome. This involves questioning who represents the healthiest segment of our population-whether it is those free from illnesses, athletes at peak performance, individuals living healthily through regular exercise and good nutrition or even elderly adults or centenarians who have been tested by time and achieved remarkable healthy longevity.This review advocates for delineating 'what defines a healthy microbiome?' by considering a broader range of factors related to human health and environmental influences on the microbiota. A healthy microbiome is undoubtedly linked to gut health. Nevertheless, it is very difficult to pinpoint a universally accepted definition of 'gut health' due to the complexities of measuring gut functionality besides the microbiota composition. We must take into account individual variabilities, the influence of diet, lifestyle, host and environmental factors. Moreover, the challenge in distinguishing causation from correlation between gut microbiome and overall health is presented.The review also highlights the resource-heavy nature of comprehensive gut health assessments, which hinders their practicality and broad application. Finally, we call for continued research and a nuanced approach to better understand the intricate and evolving concept of gut health, emphasising the need for more precise and inclusive definitions and methodologies in studying the microbiome.
Collapse
Affiliation(s)
- Matthias Van Hul
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
| | - Patrice D Cani
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
- Institute of Experimental and Clinical Research (IREC), UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Camille Petitfils
- Louvain Drug Research Institute (LDRI), Metabolism and Nutrition research group (MNUT), UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO) department, WEL Research Institute, Wavre, Belgium
| | - Willem M De Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medizinische Universitat Innsbruck, Innsbruck, Austria
| | - Emad M El-Omar
- Microbiome Research Centre, St George and Sutherland Clinical Campuses, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
30
|
Arp G, Jiang A, Dufault-Thompson K, Levy S, Zhong A, Wassan JT, Grant M, Li Y, Hall B, Jiang X. Gut Bacteria Encode Reductases that Biotransform Steroid Hormones. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.04.616736. [PMID: 39803498 PMCID: PMC11722256 DOI: 10.1101/2024.10.04.616736] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The metabolism of steroids by the gut microbiome affects hormone homeostasis, impacting host development, mental health, and reproductive functions. In this study, we identify the Δ4-3-ketosteroid 5β-reductase, 3β-hydroxysteroid dehydrogenase/Δ5-4 isomerase, and Δ6-3-ketosteroid reductase enzyme families encoded by common human gut bacteria. Through phylogenetic reconstruction and mutagenesis, We show that 5β-reductase and Δ6-3-ketosteroid reductase have evolved to specialize in converting diverse 3-keto steroid hormones into their 5β- and Δ6-reduced derivatives. We also find that the novel 3β-hydroxysteroid dehydrogenase/Δ5-4 isomerase is fused with 5β-reductase in multiple species, streamlining the multi-step conversion of pregnenolone, a steroid hormone precursor, into epipregnanolone. Through metagenomic analysis, we reveal that these enzymes are prevalent in healthy populations, being enriched in females over males. These findings provide the molecular basis for studying microbial steroid metabolism in the gut, offering insights into its potential impact on hormonal health in hosts, especially in the context of women's health.
Collapse
Affiliation(s)
- Gabriela Arp
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, Maryland, USA
| | - Angela Jiang
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, Maryland, USA
- National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Sophia Levy
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, Maryland, USA
| | - Aoshu Zhong
- Division of Molecular and Cellular Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Jyotsna Talreja Wassan
- National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
- Department of Computer Science, Maitreyi College University Of Delhi, Delhi, India
| | - Maggie Grant
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, Maryland, USA
| | - Yue Li
- Department of Chemistry and Biochemistry, University of Maryland, College Park, College Park, Maryland, USA
| | - Brantley Hall
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, College Park, Maryland, USA
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, College Park, Maryland, USA
| | - Xiaofang Jiang
- National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
31
|
Wyatt M, Choudhury A, Von Dohlen G, Heileson JL, Forsse JS, Rajakaruna S, Zec M, Tfaily MM, Greathouse L. Randomized control trial of moderate dose vitamin D alters microbiota stability and metabolite networks in healthy adults. Microbiol Spectr 2024; 12:e0008324. [PMID: 39189761 PMCID: PMC11448053 DOI: 10.1128/spectrum.00083-24] [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: 01/10/2024] [Accepted: 07/10/2024] [Indexed: 08/28/2024] Open
Abstract
Evidence indicates that both vitamin D and the gut microbiome are involved in the process of colon carcinogenesis. However, it is unclear what effects supplemental vitamin D3 has on the gut microbiome and its metabolites in healthy adults. We conducted a double-blind, randomized, placebo-controlled trial to identify the acute and long-term microbiota structural and metabolite changes that occur in response to a moderate dose (4,000 IU) of vitamin D3 for 12 weeks in healthy adults. Our results demonstrated a significant increase in serum 25-hydroxy-vitamin D (25(OH)D) in the treatment group compared to placebo (P < 0.0001). Vitamin D3 significantly increased compositional similarity (P < 0.0001) in the treatment group, and enriched members of the Bifidobacteriaceae family. We also identified a significant inverse relationship between the percent change in serum 25(OH)D and microbial stability in the treatment group (R = -0.52, P < 0.019). Furthermore, vitamin D3 supplementation resulted in notable metabolic shifts, in addition to resulting in a drastic rewiring of key gut microbial-metabolic associations. In conclusion, we show that a moderate dose of vitamin D3 among healthy adults has unique acute and persistent effects on the fecal microbiota, and suggest novel mechanisms by which vitamin D may affect the host-microbiota relationship. IMPORTANCE Preventative measures to reduce the rise in early-onset colorectal cancer are of critical need. Both vitamin D, dietary and serum levels, and the gut microbiome are implicated in the etiology of colorectal cancer. By understanding the intimate relationship between vitamin D, the gut microbiome, and its metabolites, we may be able to identify key mechanisms that can be targeted for intervention, including inflammation and metabolic dysfunction. Furthermore, the similarity of vitamin D to cholesterol, which is metabolized by the gut microbiome, gives precedence to its ability to produce metabolites that can be further studied and leveraged for controlling colorectal cancer incidence and mortality.
Collapse
Affiliation(s)
- Madhur Wyatt
- Human Health Performance and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
| | - Ankan Choudhury
- Human Science and Design, Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
| | - Gabriella Von Dohlen
- Human Science and Design, Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
| | - Jeffery L. Heileson
- Human Health Performance and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
- Nutrition Services Division, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Jeffrey S. Forsse
- Human Health Performance and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
- Department of Biology, Baylor University, Waco, Texas, USA
| | - Sumudu Rajakaruna
- Department of Environmental Science, University of Arizona, Tucson, Arizona, USA
- BIO5 Institute, The University of Arizona, Tucson, Arizona, USA
| | - Manja Zec
- Department of Environmental Science, University of Arizona, Tucson, Arizona, USA
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Malak M. Tfaily
- Department of Environmental Science, University of Arizona, Tucson, Arizona, USA
- BIO5 Institute, The University of Arizona, Tucson, Arizona, USA
| | - Leigh Greathouse
- Human Science and Design, Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
- Department of Biology, Baylor University, Waco, Texas, USA
| |
Collapse
|
32
|
Yan G, Qin Z, Liu A, Huang Z, Wang X, Zhang S, Xie X, Huang X, Chen J, Li Y, Xie Q, Liu Y, Su Z, Xie J. Sulfonation metabolism in the gut microbiota is the main metabolic pathway of cholesterol in hypercholesterolemic mice. Food Funct 2024; 15:9750-9765. [PMID: 39238326 DOI: 10.1039/d4fo02312a] [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: 09/07/2024]
Abstract
The interactions between dietary cholesterol and intestinal microbiota strongly affect host health. Sulfonation is a major conjugating pathway responsible for regulating the chemical and functional homeostasis of endogenous and exogenous molecules. However, the role of cholesterol sulfonation metabolism in the host remains unclear. This work was designed to profile cholesterol-specific host-microbe interaction and conversion focusing on cholesterol sulfonation metabolism. Results indicated that the serum and fecal cholesterol sulfate (CHS) levels were significantly higher than those of total bile acid (TBA) levels in hypercholesterolemic mice. Deletion of the gut microbiota by antibiotics could dramatically increase total cholesterol (TC) levels but it decreased CHS levels in a pseudo-germ-free (PGF) mouse host. 16S rRNA gene sequencing assay and correlation analysis between the abundance of various intestinal bacteria (phylum and class) and the CHS/TC ratio showed that the intestinal genera Bacteroides contributed essentially to cholesterol sulfonation metabolism. These results were further confirmed in an in situ and ex vivo mouse intestinal model, which indicated that the sulfonation metabolism rate of cholesterol could reach 42% under high cholesterol conditions. These findings provided new evidence that the sulfonation metabolic pathway dominated cholesterol metabolism in hypercholesterolemic mice and microbial conversion of cholesterol-to-CHS was of vital importance for cholesterol-lowering by Bacteroides. This suggested that the gut microbiota could regulate cholesterol metabolism and that it was feasible to reduce cholesterol levels by dietary interventions involving the gut microbiota.
Collapse
Affiliation(s)
- Guangtao Yan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Zehui Qin
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, PR China
| | - Aitong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Ziwei Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Xinhong Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Shanli Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Xiaolin Xie
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Xiaoqi Huang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Qingfeng Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China.
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China.
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
| | - Jianhui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China.
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, PR China
| |
Collapse
|
33
|
Chen X, Chen X, Yan D, Zhang N, Fu W, Wu M, Ge F, Wang J, Li X, Geng M, Wang J, Tang D, Liu J. GV-971 prevents severe acute pancreatitis by remodeling the microbiota-metabolic-immune axis. Nat Commun 2024; 15:8278. [PMID: 39333064 PMCID: PMC11436807 DOI: 10.1038/s41467-024-52398-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 09/02/2024] [Indexed: 09/29/2024] Open
Abstract
Despite recent advances, severe acute pancreatitis (SAP) remains a lethal inflammation with limited treatment options. Here, we provide compelling evidence of GV-971 (sodium oligomannate), an anti-Alzheimer's medication, as being a protective agent in various male mouse SAP models. Microbiome sequencing, along with intestinal microbiota transplantation and mass cytometry technology, unveil that GV-971 reshapes the gut microbiota, increasing Faecalibacterium populations and modulating both peripheral and intestinal immune systems. A metabolomics analysis of cecal contents from GV-971-treated SAP mice further identifies short-chain fatty acids, including propionate and butyrate, as key metabolites in inhibiting macrophage M1 polarization and subsequent lethal inflammation by blocking the MAPK pathway. These findings suggest GV-971 as a promising therapeutic for SAP by targeting the microbiota metabolic immune axis.
Collapse
Affiliation(s)
- Xi Chen
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xin Chen
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ding Yan
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Na Zhang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Wen Fu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Meixuan Wu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Feifei Ge
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiangtuan Wang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiaofen Li
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Meiyu Geng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jinheng Wang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Jinbao Liu
- Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Disease, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
34
|
Han S, Zhuang J, Song Y, Wu X, Yu X, Tao Y, Chu J, Qu Z, Wu Y, Han S, Yang X. Gut microbial subtypes and clinicopathological value for colorectal cancer. Cancer Med 2024; 13:e70180. [PMID: 39234654 PMCID: PMC11375334 DOI: 10.1002/cam4.70180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 08/03/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Gut bacteria are related to colorectal cancer (CRC) and its clinicopathologic characteristics. OBJECTIVE To develop gut bacterial subtypes and explore potential microbial targets for CRC. METHODS Stool samples from 914 volunteers (376 CRCs, 363 advanced adenomas, and 175 normal controls) were included for 16S rRNA sequencing. Unsupervised learning was used to generate gut microbial subtypes. Gut bacterial community composition and clustering effects were plotted. Differences of gut bacterial abundance were analyzed. Then, the association of CRC-associated bacteria with subtypes and the association of gut bacteria with clinical information were assessed. The CatBoost models based on gut differential bacteria were constructed to identify the diseases including CRC and advanced adenoma (AA). RESULTS Four gut microbial subtypes (A, B, C, D) were finally obtained via unsupervised learning. The characteristic bacteria of each subtype were Escherichia-Shigella in subtype A, Streptococcus in subtype B, Blautia in subtype C, and Bacteroides in subtype D. Clinical information (e.g., free fatty acids and total cholesterol) and CRC pathological information (e.g., tumor depth) varied among gut microbial subtypes. Bacilli, Lactobacillales, etc., were positively correlated with subtype B. Positive correlation of Blautia, Lachnospiraceae, etc., with subtype C and negative correlation of Coriobacteriia, Coriobacteriales, etc., with subtype D were found. Finally, the predictive ability of CatBoost models for CRC identification was improved based on gut microbial subtypes. CONCLUSION Gut microbial subtypes provide characteristic gut bacteria and are expected to contribute to the diagnosis of CRC.
Collapse
Affiliation(s)
- Shuwen Han
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
- Institut Catholique de Lille, Junia (ICL), Université Catholique de Lille, Laboratoire Interdisciplinaire des Transitions de Lille (LITL), Lille, France
| | - Jing Zhuang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Yifei Song
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Xinyue Wu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Xiaojian Yu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Ye Tao
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Shanghai Biozeron Biotechnology Co., Ltd., Shanghai, China
| | - Jian Chu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Zhanbo Qu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Yinhang Wu
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| | - Shugao Han
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xi Yang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, Huzhou, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer, Huzhou, China
| |
Collapse
|
35
|
Furuichi M, Kawaguchi T, Pust MM, Yasuma-Mitobe K, Plichta DR, Hasegawa N, Ohya T, Bhattarai SK, Sasajima S, Aoto Y, Tuganbaev T, Yaginuma M, Ueda M, Okahashi N, Amafuji K, Kiridoshi Y, Sugita K, Stražar M, Avila-Pacheco J, Pierce K, Clish CB, Skelly AN, Hattori M, Nakamoto N, Caballero S, Norman JM, Olle B, Tanoue T, Suda W, Arita M, Bucci V, Atarashi K, Xavier RJ, Honda K. Commensal consortia decolonize Enterobacteriaceae via ecological control. Nature 2024; 633:878-886. [PMID: 39294375 PMCID: PMC11424487 DOI: 10.1038/s41586-024-07960-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 08/19/2024] [Indexed: 09/20/2024]
Abstract
Persistent colonization and outgrowth of potentially pathogenic organisms in the intestine can result from long-term antibiotic use or inflammatory conditions, and may perpetuate dysregulated immunity and tissue damage1,2. Gram-negative Enterobacteriaceae gut pathobionts are particularly recalcitrant to conventional antibiotic treatment3,4, although an emerging body of evidence suggests that manipulation of the commensal microbiota may be a practical alternative therapeutic strategy5-7. Here we isolated and down-selected commensal bacterial consortia from stool samples from healthy humans that could strongly and specifically suppress intestinal Enterobacteriaceae. One of the elaborated consortia, comprising 18 commensal strains, effectively controlled ecological niches by regulating gluconate availability, thereby re-establishing colonization resistance and alleviating Klebsiella- and Escherichia-driven intestinal inflammation in mice. Harnessing these activities in the form of live bacterial therapies may represent a promising solution to combat the growing threat of proinflammatory, antimicrobial-resistant Enterobacteriaceae infection.
Collapse
Affiliation(s)
- Munehiro Furuichi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Takaaki Kawaguchi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Marie-Madlen Pust
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Keiko Yasuma-Mitobe
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Damian R Plichta
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Naomi Hasegawa
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Takashi Ohya
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Shakti K Bhattarai
- Department of Microbiology and Physiological Systems, Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Satoshi Sasajima
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Yoshimasa Aoto
- JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo, Japan
| | - Timur Tuganbaev
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Human Biology Microbiome Quantum Research Center (Bio2Q), Keio University, Tokyo, Japan
| | - Mizuki Yaginuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Ueda
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo, Japan
| | - Nobuyuki Okahashi
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Osaka, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Kimiko Amafuji
- JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kiridoshi
- JSR-Keio University Medical and Chemical Innovation Center, Keio University School of Medicine, Tokyo, Japan
| | - Kayoko Sugita
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Martin Stražar
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julian Avila-Pacheco
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kerry Pierce
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Clary B Clish
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ashwin N Skelly
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Masahira Hattori
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Cooperative Major in Advanced Health Science, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Nobuhiro Nakamoto
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | - Takeshi Tanoue
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Wataru Suda
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Cooperative Major in Advanced Health Science, Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Makoto Arita
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Human Biology Microbiome Quantum Research Center (Bio2Q), Keio University, Tokyo, Japan
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan
| | - Vanni Bucci
- Department of Microbiology and Physiological Systems, Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Koji Atarashi
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Human Biology Microbiome Quantum Research Center (Bio2Q), Keio University, Tokyo, Japan
| | - Ramnik J Xavier
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA.
| | - Kenya Honda
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan.
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
- Human Biology Microbiome Quantum Research Center (Bio2Q), Keio University, Tokyo, Japan.
| |
Collapse
|
36
|
Zhang S, Hou R, Sun C, Huang Q, Lin L, Li H, Liu S, Cheng Y, Xu X. Metabolic activity of gut microbial enrichment cultures from different marine species and their transformation abilities to plastic additives. ENVIRONMENT INTERNATIONAL 2024; 190:108882. [PMID: 38996798 DOI: 10.1016/j.envint.2024.108882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024]
Abstract
The role of the gut microbiota in host physiology has been previously elucidated for some marine organisms, but little information is available on their metabolic activity involved in transformation of environmental pollutants. This study assessed the metabolic profiles of the gut microbial cultures from grouper (Epinephelus coioides), green mussel (Perna viridis) and giant tiger prawn (Penaeus monodon) and investigated their transformation mechanisms to typical plastic additives. Community-level physiological profiling analysis confirmed the utilization profiles of the microbial cultures including carbon sources of carbohydrates, amines, carboxylic acids, phenolic compounds, polymers and amino acids, and the plastic additives of organophosphate flame retardants, tetrabromobisphenol A derivates and bisphenols. Using in vitro incubation, triphenyl phosphate (TPHP) was found to be rapidly metabolized into diphenyl phosphate by the gut microbiota as a representative ester-containing plastic additive, whereas the transformation of BPA (a representative phenol) was relatively slower. Interestingly, all three kinds of microbial cultures efficiently transformed the hepatic metabolite of BPA (BPA-G) back to BPA, thereby increasing its bioavailability in the body. The specific enzyme analysis confirmed the ability of the gut microbiota to perform the metabolic reactions. The results of 16S rRNA sequencing and network analysis revealed that the genera Escherichia-Shigella, Citrobacter, and Anaerospora were functional microbes, and their collaboration with fermentative microbes played pivotal roles in the transformation of the plastic additives. The structure-specific transformations by the gut microbiota and their distinct bioavailability deserve more attention in the future.
Collapse
Affiliation(s)
- Siqi Zhang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Rui Hou
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China.
| | - Chuansheng Sun
- Marine College, Shandong University, Weihai 264209, China
| | - Qianyi Huang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lang Lin
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Hengxiang Li
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Shan Liu
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Yuanyue Cheng
- State Key Laboratory of Tropical Oceanography, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Xiangrong Xu
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China; Guangxi Laboratory on the Study of Coral Reefs in the South China Sea, Coral Reef Research Center of China, School of Marine Sciences, Guangxi University, Nanning 530004, China.
| |
Collapse
|
37
|
Liedike B, Khatib M, Tabarsi B, Harris M, Wilson SL, Ortega-Santos CP, Mohr AE, Vega-López S, Whisner CM. Evaluating the Effects of Corn Flour Product Consumption on Cardiometabolic Outcomes and the Gut Microbiota in Adults with Elevated Cholesterol: A Randomized Crossover. J Nutr 2024; 154:2437-2447. [PMID: 38880174 PMCID: PMC11923428 DOI: 10.1016/j.tjnut.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/24/2024] [Accepted: 06/12/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Consumption of whole grains is associated with a reduction in chronic diseases and offers benefits for cardiovascular health and metabolic regulation. The relationship between whole-grain corn and corn bran with the gut microbiota (GM) remains an area of growing interest, particularly regarding their influence on cardiometabolic health. OBJECTIVES To investigate the effects of different corn flours on cardiometabolic outcomes and GM changes in adults with elevated low-density lipoprotein cholesterol (LDL cholesterol) concentrations. METHODS In this crossover study, 36 adults with LDL cholesterol above 110 mg/dL consumed 48 g/d of 3 corn flour types for 4 wk: whole-grain corn meal, refined corn meal (RCM), and a blend of RCM and corn bran (RCM + B). We assessed the impact on cardiometabolic markers [LDL cholesterol, high-density lipoprotein cholesterol (HDL cholesterol), total cholesterol, and triglycerides)] and GM composition and estimated function. Statistical analyses included mixed-effects modeling and responder (>5% decrease in LDL cholesterol) analysis to evaluate changes in GM related to lipid profile improvements. RESULTS Of the 3 corn flour types, only RCM + B significantly decreased LDL cholesterol over time (-10.4 ± 3.6 mg/dL, P = 0.005) and marginally decreased total cholesterol (-9.2 ± 3.9 mg/dL, P = 0.072) over time. There were no significant effects on HDL cholesterol or triglyceride concentrations. No significant changes were observed in GM alpha diversity, whereas beta diversity metrics indicated individual variability. Two genera, unclassified Lachnospiraceae and Agathobaculum (Padj ≤ 0.096), differed significantly by treatment, but only Agathobaculum remained significantly elevated in the whole-grain corn meal, compared to RCM and RCM + B, after adjustment for multiple comparisons. CONCLUSIONS The type of corn flour, particularly RCM + B, notably influenced LDL cholesterol concentrations in adults with elevated LDL cholesterol. This study suggests that incorporating milled fractions (e.g., bran) of whole-grain corn with refined corn flour may be a viable alternative to supplementing manufactured grain products with isolated or synthetic fibers for improved metabolic health. This trial was registered at clinicaltrials.gov as NCT03967990.
Collapse
Affiliation(s)
- Bethany Liedike
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States
| | - Maissa Khatib
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States
| | - Baharak Tabarsi
- Community Health Center, Valleywise Health, Phoenix, AZ, United States
| | - Michelle Harris
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States
| | - Shannon L Wilson
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States
| | - Carmen P Ortega-Santos
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States; Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington, DC, United States
| | - Alex E Mohr
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States; Center for Health Through Microbiomes, The Biodesign Institute, Arizona State University, Tempe, AZ, United States
| | - Sonia Vega-López
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States; Southwest Interdisciplinary Research Center, Arizona State University, Phoenix, AZ, United States.
| | - Corrie M Whisner
- College of Health Solutions, Arizona State University, Phoenix, AZ, United States; Center for Health Through Microbiomes, The Biodesign Institute, Arizona State University, Tempe, AZ, United States.
| |
Collapse
|
38
|
Schneider E, O'Riordan KJ, Clarke G, Cryan JF. Feeding gut microbes to nourish the brain: unravelling the diet-microbiota-gut-brain axis. Nat Metab 2024; 6:1454-1478. [PMID: 39174768 DOI: 10.1038/s42255-024-01108-6] [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: 11/01/2023] [Accepted: 07/15/2024] [Indexed: 08/24/2024]
Abstract
The prevalence of brain disorders, including stress-related neuropsychiatric disorders and conditions with cognitive dysfunction, is rising. Poor dietary habits contribute substantially to this accelerating trend. Conversely, healthy dietary intake supports mood and cognitive performance. Recently, the communication between the microorganisms within the gastrointestinal tract and the brain along the gut-brain axis has gained prominence as a potential tractable target to modulate brain health. The composition and function of the gut microbiota is robustly influenced by dietary factors to alter gut-brain signalling. To reflect this interconnection between diet, gut microbiota and brain functioning, we propose that a diet-microbiota-gut-brain axis exists that underpins health and well-being. In this Review, we provide a comprehensive overview of the interplay between diet and gut microbiota composition and function and the implications for cognition and emotional functioning. Important diet-induced effects on the gut microbiota for the development, prevention and maintenance of neuropsychiatric disorders are described. The diet-microbiota-gut-brain axis represents an uncharted frontier for brain health diagnostics and therapeutics across the lifespan.
Collapse
Affiliation(s)
| | | | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
39
|
Hu L, Hu B, zhang L, Hu Y, Zhang Y, Zhang R, Yu H, Liu D, Wang X, Lin O, Gong Y, Zhang Y, Li C, Li J. Role of gut microbiota and metabolomics in the lipid-lowering efficacy of statins among Chinese patients with coronary heart disease and hypercholesterolemia. Front Cell Infect Microbiol 2024; 14:1408581. [PMID: 39119290 PMCID: PMC11306155 DOI: 10.3389/fcimb.2024.1408581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/01/2024] [Indexed: 08/10/2024] Open
Abstract
Background Statins, being the primary pharmacological intervention for hypercholesterolemia, exhibit a notable degree of interpatient variability in their effectiveness, which may be associated with gut microbiota. This study sought to identify the biomarkers for evaluating differences in statin efficacy. Methods A quasi case-control study was conducted among participants with hypercholesterolemia and coronary heart disease taking rosuvastatin essential. According to the level of low density lipoprotein cholesterol (LDL-C), participants was divided into the "Up to standard" (US) group and the "Below standard" (BS) group. 16S rDNA sequencing and untargeted metabolomics were applied to detected the information of gut microbiota and related metabolites. Results A total of 8 US and 8 BS group matched by age and sex were included in the final analysis. 16S rDNA sequencing results indicated that the characteristic strains of the US group were f-Eubacterium_coprostanoligenes and g-Papillibacter, while the characteristic flora of the BS group were o-C0119, g-Pseudolabrys, s-Dyella-Marensis and f-Xanthobacaceae. Metabolomic results suggested that the levels of chenodeoxycholic acid-3-β-D-glucuronide, 1-methylnicotinamide and acetoacetate in stool samples of the US group were significantly higher than those of the BS group. By identifying the differentially abundant bacterial taxa, the gut microbiota could modulate the efficacy of statins through producing enzymes involved in cholesterol metabolism. Conclusions The findings suggest that the difference in statin efficacy may be related to gut microbiota strains that can produce short-chain fatty acids and secondary bile acids and affect the efficacy of statins by regulating the activities of cholesterol metabolite-related proteins. Metabolites related to short-chain fatty acids and secondary bile acids in the gut are expected to be biomarkers indicating the efficacy of statins.
Collapse
Affiliation(s)
- Lihua Hu
- Department of Cardiology, Peking University First Hospital, Beijing, China
- Institute of Cardiovascular Disease, Peking University First Hospital, Beijing, China
| | - Boxian Hu
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, China
| | - Long zhang
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Yuhong Hu
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, China
| | - Yali Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Ruihang Zhang
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, China
| | - Hongxi Yu
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, China
| | - Dan Liu
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, China
| | - Xiaolei Wang
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, China
| | - Ouya Lin
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, China
| | - Yanjun Gong
- Department of Cardiology, Peking University First Hospital, Beijing, China
| | - Yan Zhang
- Department of Cardiology, Peking University First Hospital, Beijing, China
- Institute of Cardiovascular Disease, Peking University First Hospital, Beijing, China
| | - Cheng Li
- School of Engineering Medicine of Beihang University and Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of China, Beijing, China
| | - Jianping Li
- Department of Cardiology, Peking University First Hospital, Beijing, China
- Institute of Cardiovascular Disease, Peking University First Hospital, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- National Health Commission (NHC) Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing, China
| |
Collapse
|
40
|
Potrykus M, Czaja-Stolc S, Stankiewicz M, Szymański M, Łoniewski I, Kaska Ł, Proczko-Stepaniak M. Preoperative Multistrain Probiotic Supplementation Does Not Affect Body Weight Changes or Cardiometabolic Risk Factors in Bariatrics: Randomized, Double-Blind, Placebo-Controlled Clinical Trial. Nutrients 2024; 16:2055. [PMID: 38999802 PMCID: PMC11243469 DOI: 10.3390/nu16132055] [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: 06/01/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
Emerging evidence suggests that microbiota plays a crucial role in the development, progression, and therapeutic options in obesity and its comorbidities. This study assessed preoperative probiotic therapy's impact on bariatric treatment outcomes. A 12-week randomized, double-blind, placebo-controlled trial with 48 patients undergoing bariatric surgery was conducted. Participants received probiotics-Sanprobi Barrier-which contained nine strains of bacteria: Bifidobacterium bifidum W23, Bifidobacterium lactis W51 and W52, Lactobacillus acidophilus W37, Levilactobacillus brevis W63, Lacticaseibacillus casei W56, Ligilactobacillus salivarius W24, Lactococcus lactis W19, and Lactococcus lactis W58. Primary outcomes included excess body weight loss, body weight loss, and excess body mass index loss, with secondary objectives focusing on metabolic profiles. Surgical treatment of obesity significantly improved anthropometric and metabolic parameters. No significant differences were observed in primary outcomes or in secondary outcomes between groups at any time point post-surgery. Preoperative probiotics administration did not affect clinical outcomes 1, 3, or 6 months following bariatric surgery.
Collapse
Affiliation(s)
- Marta Potrykus
- Department of Oncological, Transplant, and General Surgery, Medical University of Gdansk, 80-211 Gdańsk, Poland; (M.P.); (M.S.); (M.P.-S.)
| | - Sylwia Czaja-Stolc
- Department of Clinical Nutrition and Dietetics, Medical University of Gdansk, 80-211 Gdańsk, Poland;
| | - Marta Stankiewicz
- Department of Clinical Nutrition and Dietetics, Medical University of Gdansk, 80-211 Gdańsk, Poland;
| | - Michał Szymański
- Department of Oncological, Transplant, and General Surgery, Medical University of Gdansk, 80-211 Gdańsk, Poland; (M.P.); (M.S.); (M.P.-S.)
| | - Igor Łoniewski
- Sanprobi sp. z o.o. sp. k., 70-535 Szczecin, Poland;
- Department of Biochemical Science, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland
| | - Łukasz Kaska
- Independent Public Health Care Center of the Ministry of Internal Affairs and Administration, 80-210 Gdańsk, Poland;
| | - Monika Proczko-Stepaniak
- Department of Oncological, Transplant, and General Surgery, Medical University of Gdansk, 80-211 Gdańsk, Poland; (M.P.); (M.S.); (M.P.-S.)
| |
Collapse
|
41
|
Hou G, Wei L, Li R, Chen F, Yin J, Huang X, Yin Y. Lactobacillus delbrueckii Ameliorated Blood Lipids via Intestinal Microbiota Modulation and Fecal Bile Acid Excretion in a Ningxiang Pig Model. Animals (Basel) 2024; 14:1801. [PMID: 38929420 PMCID: PMC11201289 DOI: 10.3390/ani14121801] [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: 04/26/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Lactobacillus delbrueckii intervention can regulate body lipid metabolism, but the underlying mechanism remains unclear. Our study investigated the effects of L. delbrueckii on serum lipid levels, tissular fat metabolism and deposition, bile acid metabolism, and gut microbiota in Ningxiang pigs. Ninety-six pigs were divided into two groups and fed basal diets containing either 0 (CON) or 0.1% L. delbrueckii (LD) for 60 days. Dietary L. delbrueckii promoted fecal total bile acid (TBA) excretion and increased hepatic enzyme activities related to cholesterol and bile synthesis but decreased hepatic and serum lipid concentrations. L. delbrueckii downregulated gene expression associated with fatty acid synthesis but upregulated gene expression related to lipolysis and β-fatty acid oxidation in liver and subcutaneous fat. L. delbrueckii elevated gut Lactobacillus abundance and colonic short-chain fatty acid (SCFA)-producing bacteria but declined the abundance of some pathogenic bacteria. These findings demonstrated that L. delbrueckii modulated intestinal microbiota composition and facilitated fecal TBA excretion to regulate hepatic fat metabolism, which resulted in less lipid deposition in the liver and reduced levels of serum lipids.
Collapse
Affiliation(s)
- Gaifeng Hou
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (G.H.); (Y.Y.)
| | - Liangkai Wei
- Hunan Co-Innovation Center of Safety Animal Production, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (L.W.); (J.Y.); (X.H.)
| | - Rui Li
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (G.H.); (Y.Y.)
| | - Fengming Chen
- Academician Workstation, Changsha Medical University, Changsha 410219, China;
| | - Jie Yin
- Hunan Co-Innovation Center of Safety Animal Production, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (L.W.); (J.Y.); (X.H.)
| | - Xingguo Huang
- Hunan Co-Innovation Center of Safety Animal Production, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (L.W.); (J.Y.); (X.H.)
| | - Yulong Yin
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; (G.H.); (Y.Y.)
| |
Collapse
|
42
|
Chen C, Han P, Qing Y. Metabolic heterogeneity in tumor microenvironment - A novel landmark for immunotherapy. Autoimmun Rev 2024; 23:103579. [PMID: 39004158 DOI: 10.1016/j.autrev.2024.103579] [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: 01/31/2024] [Revised: 04/10/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
The surrounding non-cancer cells and tumor cells that make up the tumor microenvironment (TME) have various metabolic rhythms. TME metabolic heterogeneity is influenced by the intricate network of metabolic control within and between cells. DNA, protein, transport, and microbial levels are important regulators of TME metabolic homeostasis. The effectiveness of immunotherapy is also closely correlated with alterations in TME metabolism. The response of a tumor patient to immunotherapy is influenced by a variety of variables, including intracellular metabolic reprogramming, metabolic interaction between cells, ecological changes within and between tumors, and general dietary preferences. Although immunotherapy and targeted therapy have made great strides, their use in the accurate identification and treatment of tumors still has several limitations. The function of TME metabolic heterogeneity in tumor immunotherapy is summarized in this article. It focuses on how metabolic heterogeneity develops and is regulated as a tumor progresses, the precise molecular mechanisms and potential clinical significance of imbalances in intracellular metabolic homeostasis and intercellular metabolic coupling and interaction, as well as the benefits and drawbacks of targeted metabolism used in conjunction with immunotherapy. This offers insightful knowledge and important implications for individualized tumor patient diagnosis and treatment plans in the future.
Collapse
Affiliation(s)
- Chen Chen
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China
| | - Peng Han
- Harbin Medical University Cancer Hospital, Harbin 150081, Heilongjiang, China.
| | - Yanping Qing
- The First Affiliated Hospital of Ningbo University, Ningbo 315211, Zhejiang, China.
| |
Collapse
|
43
|
Wang M, Guo W, Chen JF. Caffeine: a potential mechanism for anti-obesity. Purinergic Signal 2024:10.1007/s11302-024-10022-1. [PMID: 38802651 DOI: 10.1007/s11302-024-10022-1] [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: 03/05/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Obesity refers to the excessive accumulation of fat caused by a long-term imbalance between energy intake (EI) and energy expenditure (EE). Over recent years, obesity has become a major public health challenge. Caffeine is a natural product that has been demonstrated to exert anti-obesity effects; however, the mechanisms responsible for the effect of caffeine on weight loss have yet to be fully elucidated. Most obesity-related deaths are due to cardiovascular disease. Recent research has demonstrated that caffeine can reduce the risk of death from cardiovascular disease; thus, it can be hypothesized that caffeine may represent a new therapeutic agent for weight loss. In this review, we synthesize data arising from clinical and animal studies over the last decade and discuss the potential mechanisms by which caffeine may induce weight loss, focusing particularly on increasing energy consumption, suppressing appetite, altering lipid metabolism, and influencing the gut microbiota. Finally, we summarize the major challenges associated with caffeine and anti-obesity research and highlight possible directions for future research and development.
Collapse
Affiliation(s)
- Meng Wang
- International Joint Research Center on Purinergic Signaling, School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Wei Guo
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiang-Fan Chen
- The Molecular Neuropharmacology Laboratory and the Eye-Brain Research Center, State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
44
|
Li S, Li S, Liu S, Lu S, Li J, Cheng S, Zhang S, Huang S, Li J, Jian F. Portulaca oleracea exhibited anti-coccidian activity, fortified the gut microbiota of Hu lambs. AMB Express 2024; 14:50. [PMID: 38700828 PMCID: PMC11068709 DOI: 10.1186/s13568-024-01705-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 04/13/2024] [Indexed: 05/06/2024] Open
Abstract
Coccidia of the genus Eimeria are important pathogens that cause coccidiosis in livestock and poultry. Due to the expansion of intensive farming, coccidiosis has become more difficult to control. In addition, the continued use of anti-coccidiosis drugs has led to drug resistance and residue. Some herbs used in traditional Chinese medicine (TCM) have been shown to alleviate the clinical symptoms of coccidiosis, while enhancing immunity and growth performance (GP) of livestock and poultry. Previous in vitro and in vivo studies have reported that the TCM herb Portulaca oleracea exhibited anti-parasitic activities. In total, 36 female Hu lambs were equally divided into six treatment groups: PL (low-dose P. oleracea), PH (high-dose P. oleracea), PW (P. oleracea water extract), PE (P. oleracea ethanol extract), DIC (diclazuril), and CON (control). The treatment period was 14 days. The McMaster counting method was used to evaluate the anti-coccidiosis effects of the different treatments. Untargeted metabolomics and 16S rRNA gene sequencing were used to investigate the effects of treatment on the gut microbiota (GM) and GP. The results showed that P. oleracea ameliorated coccidiosis, improved GP, increased the abundances of beneficial bacteria, and maintained the composition of the GM, but failed to completely clear coccidian oocysts. The Firmicutes to Bacteroides ratio was significantly increased in the PH group. P. oleracea increased metabolism of tryptophan as well as some vitamins and cofactors in the GM and decreased the relative content of arginine, tryptophan, niacin, and other nutrients, thereby promoting intestinal health and enhancing GP. As an alternative to the anti-coccidiosis drug DIC, P. oleracea effectively inhibited growth of coccidia, maintained the composition of the GM, promoted intestinal health, and increased nutrient digestibility.
Collapse
Affiliation(s)
- Shiheng Li
- College of Animal Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, People's Republic of China
| | - Senyang Li
- College of Animal Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, People's Republic of China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, 450046, Henan, China
| | - Shuaiqi Liu
- College of Animal Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, People's Republic of China
| | - Shunli Lu
- College of Animal Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, People's Republic of China
| | - Jing Li
- College of Animal Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, People's Republic of China
| | - Shuqi Cheng
- College of Animal Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, People's Republic of China
| | - Sumei Zhang
- College of Animal Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, People's Republic of China
| | - Shucheng Huang
- College of Animal Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, People's Republic of China
| | - Junqiang Li
- College of Animal Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, People's Republic of China
| | - Fuchun Jian
- College of Animal Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China.
- Key Laboratory of Quality and Safety Control of Poultry Products, Ministry of Agriculture and Rural Affairs, Zhengzhou, Henan, People's Republic of China.
| |
Collapse
|
45
|
Chen L, Xu R, Zhu J. Lipidome isotope labelling of gut microbes (LILGM): A method of discovering endogenous microbial lipids. Talanta 2024; 271:125730. [PMID: 38310758 DOI: 10.1016/j.talanta.2024.125730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/22/2024] [Accepted: 01/28/2024] [Indexed: 02/06/2024]
Abstract
Lipidomics analysis of gut microbiome has become critical in recent surge of extensive human disease studies that investigate microbiome contributions. However, challenges remain in comprehending the origins of thousands of lipid species produced by the diverse microbes. Here, we proposed the development and utilization of a liquid chromatography-mass spectrometry-based approach, named lipidome isotope labelling of gut microbes (LILGM), which enables confident detection and identification of endogenous gut microbial lipidome via 13C/15N labeling strategy and high-resolution mass spectrometry. Our method leveraged in vitro microbial cultures and stable isotope-labeled 13C and 15N, allowing a reasonable degree of isotope incorporation into microbial lipids over short-term of inoculation. We then systematically detected the mass spectral patterns of 182 labeled lipid species by our in-house data analysis pipeline. Further bioinformatics analyses confidently identified biologically relevant microbial lipids from lipid classes such as diacylglycerols (DGs), fatty acids (FAs), phosphatidylglycerols (PGs), and phosphatidylethanolamines (PEs) that may have profound impacts to human physiology. Our study also demonstrated the application of LILGM by showcasing the confident detection of dysregulated microbial lipids post antibiotic perturbation. The debiased sparse partial correlation analysis provides insights into lipid metabolism intricacies. Overall, our method can provide unambiguous analyses to the endogenous microbial lipids in given biological context, and can also instantly reflect the lipidomic changes of gut microbes in response to environmental factors. We believe our LILGM approach has the potential to provide new body of knowledge by combining promising analytical approaches for sensitive and specific lipid detection to support functional microbiome studies.
Collapse
Affiliation(s)
- Li Chen
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Rui Xu
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Jiangjiang Zhu
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH, 43210, USA; James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
46
|
Ridlon JM, Gaskins HR. Another renaissance for bile acid gastrointestinal microbiology. Nat Rev Gastroenterol Hepatol 2024; 21:348-364. [PMID: 38383804 PMCID: PMC11558780 DOI: 10.1038/s41575-024-00896-2] [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] [Accepted: 01/16/2024] [Indexed: 02/23/2024]
Abstract
The field of bile acid microbiology in the gastrointestinal tract is going through a current rebirth after a peak of activity in the late 1970s and early 1980s. This renewed activity is a result of many factors, including the discovery near the turn of the century that bile acids are potent signalling molecules and technological advances in next-generation sequencing, computation, culturomics, gnotobiology, and metabolomics. We describe the current state of the field with particular emphasis on questions that have remained unanswered for many decades in both bile acid synthesis by the host and metabolism by the gut microbiota. Current knowledge of established enzymatic pathways, including bile salt hydrolase, hydroxysteroid dehydrogenases involved in the oxidation and epimerization of bile acid hydroxy groups, the Hylemon-Bjӧrkhem pathway of bile acid C7-dehydroxylation, and the formation of secondary allo-bile acids, is described. We cover aspects of bile acid conjugation and esterification as well as evidence for bile acid C3-dehydroxylation and C12-dehydroxylation that are less well understood but potentially critical for our understanding of bile acid metabolism in the human gut. The physiological consequences of bile acid metabolism for human health, important caveats and cautionary notes on experimental design and interpretation of data reflecting bile acid metabolism are also explored.
Collapse
Affiliation(s)
- Jason M Ridlon
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Center for Advanced Study, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Microbiology & Immunology, Virginia Commonwealth University, Richmond, VA, USA.
| | - H Rex Gaskins
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Biomedical and Translational Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
47
|
Zhang S, Ren X, Zhang B, Lan T, Liu B. A Systematic Review of Statins for the Treatment of Nonalcoholic Steatohepatitis: Safety, Efficacy, and Mechanism of Action. Molecules 2024; 29:1859. [PMID: 38675679 PMCID: PMC11052408 DOI: 10.3390/molecules29081859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the liver component of a cluster of conditions, while its subtype, nonalcoholic steatohepatitis (NASH), emerges as a potentially progressive liver disorder that harbors the risk of evolving into cirrhosis and culminating in hepatocellular carcinoma (HCC). NASH and cardiovascular disease (CVD) have common risk factors, but compared to liver-related causes, the most common cause of death in NASH patients is CVD. Within the pharmacological armamentarium, statins, celebrated for their lipid-modulating prowess, have now garnered attention for their expansive therapeutic potential in NASH. Evidence from a plethora of studies suggests that statins not only manifest anti-inflammatory and antifibrotic properties but also impart a multifaceted beneficial impact on hepatic health. In this review, we used "statin", "NAFLD", "NASH", and "CVD" as the major keywords and conducted a literature search using the PubMed and Web of Science databases to determine the safety and efficacy of statins in patients and animals with NASH and NAFLD, and the mechanism of statin therapy for NASH. Simultaneously, we reviewed the important role of the intestinal microbiota in statin therapy for NASH, as it is hoped that statins will provide new insights into modulating the harmful inflammatory microbiota in the gut and reducing systemic inflammation in NASH patients.
Collapse
Affiliation(s)
- Shiqin Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (S.Z.); (X.R.); (B.Z.)
| | - Xiaoling Ren
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (S.Z.); (X.R.); (B.Z.)
| | - Bingzheng Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (S.Z.); (X.R.); (B.Z.)
| | - Tian Lan
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (S.Z.); (X.R.); (B.Z.)
- Department of Pharmacology, College of Pharmacy, Harbin Medical University, Harbin 150086, China
| | - Bing Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China; (S.Z.); (X.R.); (B.Z.)
| |
Collapse
|
48
|
Li C, Stražar M, Mohamed AMT, Pacheco JA, Walker RL, Lebar T, Zhao S, Lockart J, Dame A, Thurimella K, Jeanfavre S, Brown EM, Ang QY, Berdy B, Sergio D, Invernizzi R, Tinoco A, Pishchany G, Vasan RS, Balskus E, Huttenhower C, Vlamakis H, Clish C, Shaw SY, Plichta DR, Xavier RJ. Gut microbiome and metabolome profiling in Framingham heart study reveals cholesterol-metabolizing bacteria. Cell 2024; 187:1834-1852.e19. [PMID: 38569543 PMCID: PMC11071153 DOI: 10.1016/j.cell.2024.03.014] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 01/23/2024] [Accepted: 03/11/2024] [Indexed: 04/05/2024]
Abstract
Accumulating evidence suggests that cardiovascular disease (CVD) is associated with an altered gut microbiome. Our understanding of the underlying mechanisms has been hindered by lack of matched multi-omic data with diagnostic biomarkers. To comprehensively profile gut microbiome contributions to CVD, we generated stool metagenomics and metabolomics from 1,429 Framingham Heart Study participants. We identified blood lipids and cardiovascular health measurements associated with microbiome and metabolome composition. Integrated analysis revealed microbial pathways implicated in CVD, including flavonoid, γ-butyrobetaine, and cholesterol metabolism. Species from the Oscillibacter genus were associated with decreased fecal and plasma cholesterol levels. Using functional prediction and in vitro characterization of multiple representative human gut Oscillibacter isolates, we uncovered conserved cholesterol-metabolizing capabilities, including glycosylation and dehydrogenation. These findings suggest that cholesterol metabolism is a broad property of phylogenetically diverse Oscillibacter spp., with potential benefits for lipid homeostasis and cardiovascular health.
Collapse
Affiliation(s)
- Chenhao Li
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Center for Computational and Integrative Biology and Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Ahmed M T Mohamed
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Center for Computational and Integrative Biology and Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Tina Lebar
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Shijie Zhao
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julia Lockart
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Andrea Dame
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Eric M Brown
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Qi Yan Ang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Dallis Sergio
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Rachele Invernizzi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Center for Computational and Integrative Biology and Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Antonio Tinoco
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | | | - Ramachandran S Vasan
- Boston University and NHLBI's Framingham Heart Study, Framingham, MA, USA; Sections of Preventive Medicine and Epidemiology and Cardiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA; University of Texas School of Public Health, San Antonio, TX, USA
| | - Emily Balskus
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Curtis Huttenhower
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Hera Vlamakis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Clary Clish
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Stanley Y Shaw
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Center for Computational and Integrative Biology and Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
49
|
Zhou X, Shen X, Johnson JS, Spakowicz DJ, Agnello M, Zhou W, Avina M, Honkala A, Chleilat F, Chen SJ, Cha K, Leopold S, Zhu C, Chen L, Lyu L, Hornburg D, Wu S, Zhang X, Jiang C, Jiang L, Jiang L, Jian R, Brooks AW, Wang M, Contrepois K, Gao P, Rose SMSF, Tran TDB, Nguyen H, Celli A, Hong BY, Bautista EJ, Dorsett Y, Kavathas PB, Zhou Y, Sodergren E, Weinstock GM, Snyder MP. Longitudinal profiling of the microbiome at four body sites reveals core stability and individualized dynamics during health and disease. Cell Host Microbe 2024; 32:506-526.e9. [PMID: 38479397 PMCID: PMC11022754 DOI: 10.1016/j.chom.2024.02.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/23/2024] [Accepted: 02/20/2024] [Indexed: 03/26/2024]
Abstract
To understand the dynamic interplay between the human microbiome and host during health and disease, we analyzed the microbial composition, temporal dynamics, and associations with host multi-omics, immune, and clinical markers of microbiomes from four body sites in 86 participants over 6 years. We found that microbiome stability and individuality are body-site specific and heavily influenced by the host. The stool and oral microbiome are more stable than the skin and nasal microbiomes, possibly due to their interaction with the host and environment. We identify individual-specific and commonly shared bacterial taxa, with individualized taxa showing greater stability. Interestingly, microbiome dynamics correlate across body sites, suggesting systemic dynamics influenced by host-microbial-environment interactions. Notably, insulin-resistant individuals show altered microbial stability and associations among microbiome, molecular markers, and clinical features, suggesting their disrupted interaction in metabolic disease. Our study offers comprehensive views of multi-site microbial dynamics and their relationship with host health and disease.
Collapse
Affiliation(s)
- Xin Zhou
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Center for Genomics and Personalized Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford, CA 94305, USA; The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Xiaotao Shen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Center for Genomics and Personalized Medicine, Stanford, CA 94305, USA
| | - Jethro S Johnson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Oxford Centre for Microbiome Studies, Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Headington, Oxford OX3 7FY, UK
| | - Daniel J Spakowicz
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | | | - Wenyu Zhou
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Center for Genomics and Personalized Medicine, Stanford, CA 94305, USA
| | - Monica Avina
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alexander Honkala
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Healthcare Innovation Labs, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR 97239, USA
| | - Faye Chleilat
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shirley Jingyi Chen
- Stanford Healthcare Innovation Labs, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kexin Cha
- Stanford Healthcare Innovation Labs, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shana Leopold
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Chenchen Zhu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lei Chen
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Shanghai Institute of Immunology, Shanghai Jiao Tong University, Shanghai 200240, PRC
| | - Lin Lyu
- Shanghai Institute of Immunology, Shanghai Jiao Tong University, Shanghai 200240, PRC
| | - Daniel Hornburg
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Si Wu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinyue Zhang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chao Jiang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, PRC
| | - Liuyiqi Jiang
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, PRC
| | - Lihua Jiang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Andrew W Brooks
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Meng Wang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kévin Contrepois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peng Gao
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | - Hoan Nguyen
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | - Alessandra Celli
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bo-Young Hong
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Woody L Hunt School of Dental Medicine, Texas Tech University Health Science Center, El Paso, TX 79905, USA
| | - Eddy J Bautista
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Corporación Colombiana de Investigación Agropecuaria (Agrosavia), Headquarters-Mosquera, Cundinamarca 250047, Colombia
| | - Yair Dorsett
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Medicine, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Paula B Kavathas
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yanjiao Zhou
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA; Department of Medicine, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Erica Sodergren
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032, USA
| | | | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Center for Genomics and Personalized Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford, CA 94305, USA; Stanford Healthcare Innovation Labs, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
50
|
Xu Z, Mak JWY, Lin Y, Yang K, Liu Q, Zhang F, Lau L, Tang W, Ching JY, Tun HM, Chan P, Chan FKL, Ng SC. Mixed-donor faecal microbiota transplantation was associated with increased butyrate-producing bacteria for obesity. Gut 2024; 73:875-878. [PMID: 37001978 DOI: 10.1136/gutjnl-2022-328993] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/23/2023] [Indexed: 06/19/2023]
Affiliation(s)
- Zhilu Xu
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Joyce Wing Yan Mak
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yu Lin
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Keli Yang
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qin Liu
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Fen Zhang
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Louis Lau
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Whitney Tang
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jessica Yl Ching
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Hein M Tun
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
- The Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Paul Chan
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Francis K L Chan
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Siew C Ng
- Microbiota I-Center (MagIC), Hong Kong SAR, China; Department of Medicine and Therapeutics, Institute of Digestive Disease, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|