1
|
Thibaut MM, Roumain M, Piron E, Gillard J, Loriot A, Neyrinck AM, Rodriguez J, Massart I, Thissen JP, Huot JR, Pin F, Bonetto A, Delzenne NM, Muccioli GG, Bindels LB. The microbiota-derived bile acid taurodeoxycholic acid improves hepatic cholesterol levels in mice with cancer cachexia. Gut Microbes 2025; 17:2449586. [PMID: 39780051 PMCID: PMC11730681 DOI: 10.1080/19490976.2025.2449586] [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: 06/11/2024] [Revised: 11/20/2024] [Accepted: 12/30/2024] [Indexed: 01/11/2025] Open
Abstract
Alterations in bile acid profile and pathways contribute to hepatic inflammation in cancer cachexia, a syndrome worsening the prognosis of cancer patients. As the gut microbiota impinges on host metabolism through bile acids, the current study aimed to explore the functional contribution of gut microbial dysbiosis to bile acid dysmetabolism and associated disorders in cancer cachexia. Using three mouse models of cancer cachexia (the C26, MC38 and HCT116 models), we evidenced a reduction in the hepatic levels of several secondary bile acids, mainly taurodeoxycholic (TDCA). This reduction in hepatic TDCA occurred before the appearance of cachexia. Longitudinal analysis of the gut microbiota pinpointed an ASV, identified as Xylanibacter rodentium, as a bacterium potentially involved in the reduced production of TDCA. Coherently, stable isotope-based experiments highlighted a robust decrease in the microbial 7α-dehydroxylation (7α-DH) activity with no changes in the bile salt hydrolase (BSH) activity in cachectic mice. This approach also highlighted a reduced microbial 7α-hydroxysteroid dehydrogenase (7α-HSDH) and 12α-hydroxysteroid dehydrogenase (12α-HSDH) activities in these mice. The contribution of the lower production of TDCA to cancer cachexia was explored in vitro and in vivo. In vitro, TDCA prevented myotube atrophy, whereas in vivo hepatic whole transcriptome analysis revealed that TDCA administration to cachectic mice improved the unfolded protein response and cholesterol homeostasis pathways. Coherently, TDCA administration reversed hepatic cholesterol accumulation in these mice. Altogether, this work highlights the contribution of the gut microbiota to bile acid dysmetabolism and the therapeutic interest of the secondary bile acid TDCA for hepatic cholesterol homeostasis in the context of cancer cachexia. Such discovery may prove instrumental in the understanding of other metabolic diseases characterized by microbial dysbiosis. More broadly, our work demonstrates the interest and relevance of microbial activity measurements using stable isotopes, an approach currently underused in the microbiome field.
Collapse
Affiliation(s)
- Morgane M. Thibaut
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Martin Roumain
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Edwige Piron
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Justine Gillard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Laboratory of Hepato-Gastroenterology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Axelle Loriot
- Computational Biology and Bioinformatics Unit (CBIO), de Duve Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Audrey M. Neyrinck
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Julie Rodriguez
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Isabelle Massart
- Endocrinology, Diabetology and Nutrition Department, Institut de Recherches Expérimentales et Cliniques, UCLouvain, Université catholique de Louvain, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Jean-Paul Thissen
- Endocrinology, Diabetology and Nutrition Department, Institut de Recherches Expérimentales et Cliniques, UCLouvain, Université catholique de Louvain, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Joshua R. Huot
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrea Bonetto
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Nathalie M. Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Giulio G. Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Laure B. Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université catholique de Louvain, Brussels, Belgium
- Welbio Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
2
|
Wolf PG, Welsh C, Binion B, Dai H, Oliveira ML, Hamm A, Goldberg S, Buobu PS, Schering T, Vergis S, Kessee N, Gomez SL, Yazici C, Maienschein-Cline M, Byrd DA, Gaskins HR, Ridlon JM, Mutlu E, Greening C, Tussing-Humphreys L. Secondary Bile Acid Derivatives Are Contributors to the Fecal Bile Acid Pool and Associated With Bile Acid-Modulating Nutrients. J Nutr 2025; 155:826-838. [PMID: 39805403 PMCID: PMC11934243 DOI: 10.1016/j.tjnut.2024.12.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Accumulation of hydrophobic bile acids (BAs) is linked with cancer development. However, derivatives of deoxycholic acid (DCA) and lithocholic acid (LCA) produced via bacterial metabolism may mitigate the proinflammatory and cytotoxic effects of hydrophobic BAs. The impact of diet on secondary BA derivative production has not been determined. OBJECTIVES This study aimed to study the associations between BA-modulating nutrients and the composition of secondary BAs and their derivatives. METHODS Stool and blood were collected from 138 participants aged 45-75 y that self-identified as Black or non-Hispanic White. BAs were extracted from stool and serum and quantified using LC/ESI-MS/MS. Energy, macronutrients, micronutrients, and specific dietary nutrients were estimated from two 24-h diet recalls. The abundance of genes for microbial BA metabolism was assessed from stool metagenomes. Kendall τ correlation and regression-based modeling were performed to determine associations between BA categories, microbial genes, and select energy-adjusted dietary variables (alcohol, calcium, coffee, fiber, fat, and protein). RESULTS Participants had a mean age of 60 y and a mean BMI of 31 kg/m2. BA derivatives were present in all participant stools, with lagodeoxycholic acid being the most abundant derivative quantified. Analysis of stool microbial metagenomes revealed the presence of genes for secondary BA derivative production in all participants. Protein is positively associated with the accumulation of secondary BAs. monounsaturated fatty acids (MUFA)s were negatively associated with high abundant derivatives of DCA in regression models. Total fiber and coffee intake were positively correlated with increased conversion of BAs to derivatives. Race and smoking status were significant predictors of associations between dietary variables and BA derivatives. CONCLUSION Protein, MUFAs, total fiber and coffee are significantly associated with concentrations of secondary BAs and their derivatives. Future work should account for social and structural influences on dietary intake and its relationship with BA-elicited cancer risk.
Collapse
Affiliation(s)
- Patricia G Wolf
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States; Institute for Cancer Research, Purdue University, West Lafayette, IN, United States.
| | - Caitlin Welsh
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Briawna Binion
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Champaign, IL, United States
| | - Hanchu Dai
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, United States
| | - Manoela Lima Oliveira
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, United States; University of Illinois Cancer Center, Chicago, Illinois, United States
| | - Alyshia Hamm
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, United States; University of Illinois Cancer Center, Chicago, Illinois, United States
| | - Sarah Goldberg
- Department of Clinical Nutrition, Rush University, Chicago, IL, United States
| | - Pius Sarfo Buobu
- Department of Nutrition Science, Purdue University, West Lafayette, IN, United States; Institute for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Teresa Schering
- University of Illinois Cancer Center, Chicago, Illinois, United States
| | - Sevasti Vergis
- University of Illinois Cancer Center, Chicago, Illinois, United States; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Nicollette Kessee
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, United States
| | - Sandra L Gomez
- Department of Clinical Nutrition, Rush University, Chicago, IL, United States
| | - Cemal Yazici
- Department of Medicine, University of Illinois Chicago, Chicago, IL
| | | | - Doratha A Byrd
- Department of Cancer Epidemiology, Division of Population Science, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - H Rex Gaskins
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Champaign, IL, United States; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, United States; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, United States; Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Jason M Ridlon
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Champaign, IL, United States; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, United States; Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, United States; Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, United States
| | - Ece Mutlu
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois Chicago, Chicago, IL, United States
| | - Chris Greening
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Lisa Tussing-Humphreys
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL, United States; University of Illinois Cancer Center, Chicago, Illinois, United States.
| |
Collapse
|
3
|
Abavisani M, Faraji S, Ebadpour N, Karav S, Sahebkar A. Beyond the Hayflick limit: How microbes influence cellular aging. Ageing Res Rev 2025; 104:102657. [PMID: 39788433 DOI: 10.1016/j.arr.2025.102657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/23/2024] [Accepted: 01/05/2025] [Indexed: 01/12/2025]
Abstract
Cellular senescence, a complex biological process resulting in permanent cell-cycle arrest, is central to aging and age-related diseases. A key concept in understanding cellular senescence is the Hayflick Limit, which refers to the limited capacity of normal human cells to divide, after which they become senescent. Senescent cells (SC) accumulate with age, releasing pro-inflammatory and tissue-remodeling factors collectively known as the senescence-associated secretory phenotype (SASP). The causes of senescence are multifaceted, including telomere attrition, oxidative stress, and genotoxic damage, and they extend to influences from microbial sources. Research increasingly emphasizes the role of the microbiome, especially gut microbiota (GM), in modulating host senescence processes. Beneficial microbial metabolites, such as short-chain fatty acids (SCFAs), support host health by maintaining antioxidant defenses and reducing inflammation, potentially mitigating senescence onset. Conversely, pathogenic bacteria like Pseudomonas aeruginosa and Helicobacter pylori introduce factors that damage host DNA or increase ROS, accelerating senescence via pathways such as NF-κB and p53-p21. This review explores the impact of bacterial factors on cellular senescence, highlighting the role of specific bacterial toxins in promoting senescence. Additionally, it discusses how dysbiosis and the loss of beneficial microbial species further contribute to age-related cellular deterioration. Modulating the gut microbiome to delay cellular senescence opens a path toward targeted anti-aging strategies. This work underscores the need for deeper investigation into microbial influence on aging, supporting innovative interventions to manage and potentially reverse cellular senescence.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saba Faraji
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Chen LB, Chen Q, Chao S, Yuan ZH, Jia L, Niu YL. Influence of gut flora on diabetes management after kidney transplantation. BMC Nephrol 2024; 25:468. [PMID: 39716100 DOI: 10.1186/s12882-024-03899-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/02/2024] [Indexed: 12/25/2024] Open
Abstract
Post-transplant diabetes mellitus (PTDM) is a common complication following renal transplantation, and its incidence has been gradually increasing in recent years, posing a significant public health challenge. Managing PTDM is complex, as studies suggest that it involves changes in the microbial flora across multiple organs. Recent research highlights the critical role of gut flora metabolism in the development of diabetes among post-renal transplant patients. This paper reviews the alterations in gut flora observed in PTDM patients and explores how gut flora influences PTDM. These findings may offer new perspectives on targeting gut flora metabolites for the prevention and treatment of PTDM.
Collapse
Affiliation(s)
- Luo-Bei Chen
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, No. 28 Guimedical Street, Yunyan District, Guiyang, Guizhou Province, 550000, China
| | - Qian Chen
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, No. 28 Guimedical Street, Yunyan District, Guiyang, Guizhou Province, 550000, China
| | - Sheng Chao
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, No. 28 Guimedical Street, Yunyan District, Guiyang, Guizhou Province, 550000, China
| | - Zhi-Hui Yuan
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, No. 28 Guimedical Street, Yunyan District, Guiyang, Guizhou Province, 550000, China
| | - Lei Jia
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, No. 28 Guimedical Street, Yunyan District, Guiyang, Guizhou Province, 550000, China
| | - Yu-Lin Niu
- Department of Organ Transplantation, Affiliated Hospital of Guizhou Medical University, No. 28 Guimedical Street, Yunyan District, Guiyang, Guizhou Province, 550000, China.
| |
Collapse
|
5
|
Jiang Z, Mei L, Li Y, Guo Y, Yang B, Huang Z, Li Y. Enzymatic Regulation of the Gut Microbiota: Mechanisms and Implications for Host Health. Biomolecules 2024; 14:1638. [PMID: 39766345 PMCID: PMC11727233 DOI: 10.3390/biom14121638] [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: 10/19/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
The gut microbiota, a complex ecosystem, is vital to host health as it aids digestion, modulates the immune system, influences metabolism, and interacts with the brain-gut axis. Various factors influence the composition of this microbiota. Enzymes, as essential catalysts, actively participate in biochemical reactions that have an impact on the gut microbial community, affecting both the microorganisms and the gut environment. Enzymes play an important role in the regulation of the intestinal microbiota, but the interactions between enzymes and microbial communities, as well as the precise mechanisms of enzymes, remain a challenge in scientific research. Enzymes serve both traditional nutritional functions, such as the breakdown of complex substrates into absorbable small molecules, and non-nutritional roles, which encompass antibacterial function, immunomodulation, intestinal health maintenance, and stress reduction, among others. This study categorizes enzymes according to their source and explores the mechanistic principles by which enzymes drive gut microbial activity, including the promotion of microbial proliferation, the direct elimination of harmful microbes, the modulation of bacterial interaction networks, and the reduction in immune stress. A systematic understanding of enzymes in regulating the gut microbiota and the study of their associated molecular mechanisms will facilitate the application of enzymes to precisely regulate the gut microbiota in the future and suggest new therapeutic strategies and dietary recommendations. In conclusion, this review provides a comprehensive overview of the role of enzymes in modulating the gut microbiota. It explores the underlying molecular and cellular mechanisms and discusses the potential applications of enzyme-mediated microbiota regulation for host gut health.
Collapse
Affiliation(s)
- Zipeng Jiang
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510641, China
| | - Liang Mei
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| | - Yuqi Li
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| | - Yuguang Guo
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| | - Bo Yang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510641, China
| | - Zhiyi Huang
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| | - Yangyuan Li
- Guangdong VTR Bio-Tech Co,. Ltd., Zhuhai 519060, China
| |
Collapse
|
6
|
Chu C, Behera TR, Huang Y, Qiu W, Chen J, Shen Q. Research progress of gut microbiome and diabetic nephropathy. Front Med (Lausanne) 2024; 11:1490314. [PMID: 39735707 PMCID: PMC11671260 DOI: 10.3389/fmed.2024.1490314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/19/2024] [Indexed: 12/31/2024] Open
Abstract
Diabetic nephropathy is an important complication of diabetic microvascular injury, and it is also an important cause of end-stage renal disease. Its high prevalence and disability rate significantly impacts patients' quality of life while imposing substantial social and economic burdens. Gut microbiota affects host metabolism, multiple organ functions, and regulates host health throughout the life cycle. With the rapid development of technology, researchers have found that gut microbiota is closely related to the progression of diabetic kidney disease. This review explores the role of gut microbiome in diabetic nephropathy summarizing proposed mechanisms of progression and focusing on microbial metabolites, intestinal barrier disruption, inflammation, filtration barrier damage and renal fibrosis. This review also examines the mechanism and limitations of current treatments, including drugs, fecal microbiota transplantation, and lifestyle changes, offering new perspectives on prevention and treatment.
Collapse
Affiliation(s)
- Chenling Chu
- Department of Clinical Medicine, Hangzhou Normal University, Hangzhou, China
| | - Tapas Ranjan Behera
- Department of Cancer Biology, Cleveland Clinic, Cleveland, OH, United States
| | - Ying Huang
- Department of Public Health and Preventive Medicine, Hangzhou Medical College, Hangzhou, China
| | - Wenhui Qiu
- Department of Basic Medicine and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Jiayi Chen
- Department of Clinical Medicine, Hangzhou Normal University, Hangzhou, China
| | - Quanquan Shen
- Department of Nephrology, Zhejiang Provincial People’s Hospital Bijie Hospital, Bijie, China
- Department of Nephrology, Urology & Nephrology Center, Zhejiang Provincial People’s Hospital (Affiliated People’s Hospital, Hangzhou Medical College), Hangzhou, China
| |
Collapse
|
7
|
Zhou Z, Li W, Wu Y, Wang T, Zhang J, You L, Li H, Zheng C, Gao Y, Sun X. Bidirectional Mendelian randomization links gut microbiota to primary biliary cholangitis. Sci Rep 2024; 14:28301. [PMID: 39550468 PMCID: PMC11569131 DOI: 10.1038/s41598-024-79227-z] [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: 12/12/2023] [Accepted: 11/07/2024] [Indexed: 11/18/2024] Open
Abstract
Primary biliary cholangitis (PBC) and gut microbiota (GM) are epidemiologically correlated but the causal inter-relationships remain poorly understood. We aim to explore the causal relationships between GM and PBC. Using the MiBioGen consortium, GWAS data for GM at the species level and the largest publicly available PBC GWAS data to date, we performed a bidirectional two-sample Mendelian randomization by the inverse variance weighted, MR-Egger, weighted median, weighted model and MR-PRESSO to elucidate the potential causal role of GM in PBC. To measure the heterogeneity of instrumental variables (IV), Cochran's Q statistic and MR-Egger intercept test were used. Genetically instrumented order Coriobacteriales (odds ratio [OR] = 2.18, 95% confidence interval [CI] 1.30-3.66, P = 0.004) significantly increased the risk for PBC, while genetically driven class Deltaproteobacteria (OR = 0.52, 95% CI 0.36-0.74, P = 0.002) causally decrease the NAFLD risk. Reverse MR analysis showed no significant association between PBC and the two specific GM. However, it indicated that PBC progression significantly increases the abundance of the class Bacteroidia, order Bacteroidales, and phylum Bacteroidetes (OR = 1.02, 95% CI 1.002-1.03, P = 0.026), while decreasing the abundance of the genus Lachnospiraceae UCG010 (OR = 0.98, 95% CI 0.96-0.995, P = 0.026). Our study demonstrated that genetically driven order Coriobacteriales and class Deltaproteobacteria were causally related to PBC risk. This causality provided a new perspective on ameliorating PBC by modulating GM. Our study demonstrated that genetically driven order Coriobacteriales and class Deltaproteobacteria were causally related to PBC risk. PBC was causally related to the abundance of four GM taxa(class Bacteroidia, order Bacteroidales, phylum Bacteroidetes and genus Lachnospiraceae UCG010). This causality provided a new perspective on ameliorating PBC by modulating GM.
Collapse
Affiliation(s)
- Zhijia Zhou
- Department of Hepatology, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenxuan Li
- Department of Hepatology, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuelan Wu
- Department of Hepatology, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Wang
- Department of Hepatology, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Central Laboratory, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinghao Zhang
- Department of Hepatology, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Liping You
- Department of Hepatology, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haoran Li
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chao Zheng
- Department of Hepatology, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Yueqiu Gao
- Department of Hepatology, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Xuehua Sun
- Department of Hepatology, ShuGuang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
8
|
Rowe JC, Summers SC, Quimby JM, Winston JA. Fecal bile acid dysmetabolism and reduced ursodeoxycholic acid correlate with novel microbial signatures in feline chronic kidney disease. Front Microbiol 2024; 15:1458090. [PMID: 39498133 PMCID: PMC11532117 DOI: 10.3389/fmicb.2024.1458090] [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: 07/01/2024] [Accepted: 09/16/2024] [Indexed: 11/07/2024] Open
Abstract
Background Microbial-derived secondary bile acids (SBAs) are reabsorbed and sensed via host receptors modulating cellular inflammation and fibrosis. Feline chronic kidney disease (CKD) occurs with progressive renal inflammation and fibrosis, mirroring the disease pathophysiology of human CKD patients. Methods Prospective cross-sectional study compared healthy cats (n = 6) with CKD (IRIS Stage 2 n = 17, Stage 3 or 4 n = 11). Single timepoint fecal samples from all cats underwent targeted bile acid metabolomics. 16S rRNA gene amplicon sequencing using DADA2 with SILVA taxonomy characterized the fecal microbiota. Results CKD cats had significantly reduced fecal concentrations (median 12.8 ng/mg, Mann-Whitney p = 0.0127) of the SBA ursodeoxycholic acid (UDCA) compared to healthy cats (median 39.4 ng/mg). Bile acid dysmetabolism characterized by <50% SBAs was present in 8/28 CKD and 0/6 healthy cats. Beta diversity significantly differed between cats with <50% SBAs and > 50% SBAs (PERMANOVA p < 0.0001). Twenty-six amplicon sequence variants (ASVs) with >97% nucleotide identity to Peptacetobacter hiranonis were identified. P. hiranonis combined relative abundance was significantly reduced (median 2.1%) in CKD cats with <50% SBAs compared to CKD cats with >50% SBAs (median 13.9%, adjusted p = 0.0002) and healthy cats with >50% SBAs (median 15.5%, adjusted p = 0.0112). P. hiranonis combined relative abundance was significantly positively correlated with the SBAs deoxycholic acid (Spearman r = 0.5218, adjusted p = 0.0407) and lithocholic acid (Spearman r = 0.5615, adjusted p = 0.0156). Three Oscillospirales ASVs and a Roseburia ASV were also identified as significantly correlated with fecal SBAs. Clinical and translational importance The gut-kidney axis mediated through microbial-derived SBAs appears relevant to the spontaneous animal CKD model of domestic cats. This includes reduced fecal concentrations of the microbial-derived SBA UDCA, known to regulate inflammation and fibrosis and be reno-protective. Microbes correlated with fecal SBAs include bai operon containing P. hiranonis, as well as members of Oscillospirales, which also harbor a functional bai operon. Ultimately, CKD cats represent a translational opportunity to study the role of SBAs in the gut-kidney axis, including the potential to identify novel microbial-directed therapeutics to mitigate CKD pathogenesis in veterinary patients and humans alike.
Collapse
Affiliation(s)
- John C. Rowe
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
| | - Stacie C. Summers
- Department of Clinical Sciences, Oregon State University Carlson College of Veterinary Medicine, Corvallis, OR, United States
| | - Jessica M. Quimby
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
| | - Jenessa A. Winston
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
- Comparative Hepatobiliary Intestinal Research Program (CHIRP), The Ohio State University College of Veterinary Medicine, Columbus, OH, United States
| |
Collapse
|
9
|
Daniel SL, Ridlon JM. Clostridium scindens : an endocrine keystone species in the mammalian gut. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.23.609444. [PMID: 39229245 PMCID: PMC11370556 DOI: 10.1101/2024.08.23.609444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Clostridium scindens is a keystone human gut microbial taxonomic group that, while low in abundance, has a disproportionate effect on bile acid and steroid metabolism in the mammalian gut. Numerous studies indicate that the two most studied strains of C. scindens (i.e., ATCC 35704 and VPI 12708) are important for a myriad of physiological processes in the host. We focus on both historical and current microbiological and molecular biology work on the Hylemon-Björkhem pathway and the steroid-17,20-desmolase pathway that were first discovered in C. scindens. Our most recent analysis now calls into question whether strains currently defined as C. scindens represent two separate taxonomic groups. Future directions include developing genetic tools to further explore the physiological role bile acid and steroid metabolism by strains of C. scindens , and the causal role of these pathways in host physiology and disease.
Collapse
|
10
|
Sun S, Zhang G, Lv S, Sun J. Potential mechanisms of traditional Chinese medicine in the treatment of liver cirrhosis: a focus on gut microbiota. Front Microbiol 2024; 15:1407991. [PMID: 39234554 PMCID: PMC11371771 DOI: 10.3389/fmicb.2024.1407991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Cirrhosis, a pathological stage that develops from various chronic liver diseases, is characterized by liver fibrosis, pseudolobular formation, and chronic inflammation. When it progresses to the decompensated phase, the mortality rate of cirrhosis can reach 80%. The role of gut microbiota in the progression of liver diseases has received significant attention. Numerous studies have shown that regulating gut microbiota has significant therapeutic effects on preventing and reversing liver cirrhosis. This article reviewed the mechanisms by which gut microbiota influence liver cirrhosis, explaining the effective therapeutic effects of traditional Chinese medicine. Through multi-directional regulation involving signaling pathways, gut microbiota diversity, and restoration of intestinal barrier function, traditional Chinese medicine has been promising in ameliorating liver cirrhosis, providing treatment options and pharmacological guidance for the occurrence and development of liver cirrhosis.
Collapse
Affiliation(s)
- Siyuan Sun
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Guangheng Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shimeng Lv
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinhui Sun
- Gastroenterology Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
11
|
Vico-Oton E, Volet C, Jacquemin N, Dong Y, Hapfelmeier S, Meibom KL, Bernier-Latmani R. Strain-dependent induction of primary bile acid 7-dehydroxylation by cholic acid. BMC Microbiol 2024; 24:286. [PMID: 39090543 PMCID: PMC11293179 DOI: 10.1186/s12866-024-03433-y] [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: 04/02/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Bile acids (BAs) are steroid-derived molecules with important roles in digestion, the maintenance of host metabolism, and immunomodulation. Primary BAs are synthesized by the host, while secondary BAs are produced by the gut microbiome through transformation of the former. The regulation of microbial production of secondary BAs is not well understood, particularly the production of 7-dehydroxylated BAs, which are the most potent agonists for host BA receptors. The 7-dehydroxylation of cholic acid (CA) is well established and is linked to the expression of a bile acid-inducible (bai) operon responsible for this process. However, little to no 7-dehydroxylation has been reported for other host-derived BAs (e.g., chenodeoxycholic acid, CDCA or ursodeoxycholic acid, UDCA). RESULTS Here, we demonstrate that the 7-dehydroxylation of CDCA and UDCA by the human isolate Clostridium scindens is induced when CA is present, suggesting that CA-dependent transcriptional regulation is required for substantial 7-dehydroxylation of these primary BAs. This is supported by the finding that UDCA alone does not promote expression of bai genes. CDCA upregulates expression of the bai genes but the expression is greater when CA is present. In contrast, the murine isolate Extibacter muris exhibits a distinct response; CA did not induce significant 7-dehydroxylation of primary BAs, whereas BA 7-dehydroxylation was promoted upon addition of germ-free mouse cecal content in vitro. However, E. muris was found to 7-dehydroxylate in vivo. CONCLUSIONS The distinct expression responses amongst strains indicate that bai genes are regulated differently. CA promoted bai operon gene expression and the 7-dehydroxylating activity in C. scindens strains. Conversely, the in vitro activity of E. muris was promoted only after the addition of cecal content and the isolate did not alter bai gene expression in response to CA. The accessory gene baiJ was only upregulated in the C. scindens ATCC 35704 strain, implying mechanistic differences amongst isolates. Interestingly, the human-derived C. scindens strains were also capable of 7-dehydroxylating murine bile acids (muricholic acids) to a limited extent. This study shows novel 7-dehydroxylation activity in vitro resulting from the presence of CA and suggests distinct bai gene expression across bacterial species.
Collapse
Affiliation(s)
- Eduard Vico-Oton
- Environmental Microbiology Laboratory, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Colin Volet
- Environmental Microbiology Laboratory, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Nicolas Jacquemin
- Environmental Microbiology Laboratory, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Yuan Dong
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | | | - Karin Lederballe Meibom
- Environmental Microbiology Laboratory, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Rizlan Bernier-Latmani
- Environmental Microbiology Laboratory, School of Architecture, Civil and Environmental Engineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
- EPFL ENAC IIE EML CH A1 375 (Bâtiment CH), Station 6, CH-1015, Lausanne, Switzerland.
| |
Collapse
|
12
|
Luo Z, Zhou W, Xie T, Xu W, Shi C, Xiao Z, Si Y, Ma Y, Ren Q, Di L, Shan J. The role of botanical triterpenoids and steroids in bile acid metabolism, transport, and signaling: Pharmacological and toxicological implications. Acta Pharm Sin B 2024; 14:3385-3415. [PMID: 39220868 PMCID: PMC11365449 DOI: 10.1016/j.apsb.2024.04.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/28/2024] [Accepted: 04/22/2024] [Indexed: 09/04/2024] Open
Abstract
Bile acids (BAs) are synthesized by the host liver from cholesterol and are delivered to the intestine, where they undergo further metabolism by gut microbes and circulate between the liver and intestines through various transporters. They serve to emulsify dietary lipids and act as signaling molecules, regulating the host's metabolism and immune homeostasis through specific receptors. Therefore, disruptions in BA metabolism, transport, and signaling are closely associated with cholestasis, metabolic disorders, autoimmune diseases, and others. Botanical triterpenoids and steroids share structural similarities with BAs, and they have been found to modulate BA metabolism, transport, and signaling, potentially exerting pharmacological or toxicological effects. Here, we have updated the research progress on BA, with a particular emphasis on new-found microbial BAs. Additionally, the latest advancements in targeting BA metabolism and signaling for disease treatment are highlighted. Subsequently, the roles of botanical triterpenoids in BA metabolism, transport, and signaling are examined, analyzing their potential pharmacological, toxicological, or drug interaction effects through these mechanisms. Finally, a research paradigm is proposed that utilizes the gut microbiota as a link to interpret the role of these important natural products in BA signaling.
Collapse
Affiliation(s)
- Zichen Luo
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Children’s Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Wei Zhou
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tong Xie
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Children’s Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weichen Xu
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Children’s Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chen Shi
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Children’s Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zihan Xiao
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Children’s Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yu Si
- Jiangsu CM Clinical Medicine Innovation Center for Obstetrics, Gynecology, and Reproduction, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210001, China
| | - Yan Ma
- National Institute of Biological Sciences, Beijing 102206, China
| | - Qingling Ren
- Jiangsu CM Clinical Medicine Innovation Center for Obstetrics, Gynecology, and Reproduction, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210001, China
| | - Liuqing Di
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinjun Shan
- Medical Metabolomics Center, Institute of Pediatrics, Jiangsu Key Laboratory of Children’s Health and Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
13
|
Shu T, Zhang J, Hu R, Zhou F, Li H, Liu J, Fan Y, Li X, Ding P. Qi Huang Fang improves intestinal barrier function and intestinal microbes in septic mice through NLRP3 inflammasome-mediated cellular pyroptosis. Transpl Immunol 2024; 85:102072. [PMID: 38857634 DOI: 10.1016/j.trim.2024.102072] [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: 12/08/2023] [Revised: 06/03/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
OBJECTIVE Sepsis has a high incidence, morbidity, and mortality rate and is a great threat to human safety. Gut health plays an important role in sepsis development. Qi Huang Fang (QHF) contains astragalus, rhubarb, zhishi, and atractylodes. It is used to treat syndromes of obstructive qi and deficiency of righteousness. This study aimed to investigate whether QHF improves intestinal barrier function and microorganisms in mice through NLRP3 inflammatory vesicle-mediated cellular focal death. METHODS A mouse model of sepsis was constructed by cecal ligation and puncture (CLP) of specific pathogen-free (SPF)-grade C57BL/6 mice after continuous gavage of low, medium, and high doses of astragalus formula or probiotics for 4 weeks. Twenty-four hours postoperatively, the mechanism of action of QHF in alleviating septic intestinal dysfunction and restoring intestinal microecology, thereby alleviating intestinal injury, was evaluated by pathological observation, immunohistochemistry, western blotting, ELISA, and 16S rDNA high-throughput sequencing. RESULTS Different doses of QHF and probiotics ameliorated intestinal injury and reduced colonic apoptosis in mice to varying degrees (P < 0.05). Meanwhile, different doses of QHF and probiotics were able to reduce the serum levels of IL-6, IL-1β, and TNF-α (P < 0.05); down-regulate the protein expression of NLRP3, caspase-1, and caspase-11 (P < 0.05); and up-regulate the protein expression of zonula occluden-1 (ZO-1) and occludin (P < 0.05), which improved the intestinal barrier function in mice. In addition, QHF decreased the relative abundance of harmful bacteria (Firmicutes, Muribaculaceae, Campilobacterota, Helicobacter, and Alistipes) and increased the relative abundance of beneficial bacteria (Bacteroidetes and Actinobacteria) (P < 0.05). CONCLUSION QHF improves intestinal barrier function and gut microbiology in mice via NLRP3 inflammasome-mediated cellular pyroptosis.
Collapse
Affiliation(s)
- Tingting Shu
- Department of Intensive Care Unit, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Jun Zhang
- Department of Intensive Care Unit, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Ruiying Hu
- Department of Emergency Medicine, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Fang Zhou
- Department of Emergency Medicine, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Hanyong Li
- Department of Intensive Care Unit, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Jing Liu
- Department of Medical, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Yanbo Fan
- Department of Science and Education Section, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Xucheng Li
- Department of Emergency Medicine, Wuhan Hospital of Traditional Chinese Medicine, China
| | - Peiwu Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
14
|
Leung HKM, Lo EKK, Zhang F, Felicianna, Ismaiah MJ, Chen C, El-Nezami H. Modulation of Gut Microbial Biomarkers and Metabolites in Cancer Management by Tea Compounds. Int J Mol Sci 2024; 25:6348. [PMID: 38928054 PMCID: PMC11203446 DOI: 10.3390/ijms25126348] [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: 04/29/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Cancers are causing millions of deaths and leaving a huge clinical and economic burden. High costs of cancer drugs are limiting their access to the growing number of cancer cases. The development of more affordable alternative therapy could reach more patients. As gut microbiota plays a significant role in the development and treatment of cancer, microbiome-targeted therapy has gained more attention in recent years. Dietary and natural compounds can modulate gut microbiota composition while providing broader and more accessible access to medicine. Tea compounds have been shown to have anti-cancer properties as well as modulate the gut microbiota and their related metabolites. However, there is no comprehensive review that focuses on the gut modulatory effects of tea compounds and their impact on reshaping the metabolic profiles, particularly in cancer models. In this review, the effects of different tea compounds on gut microbiota in cancer settings are discussed. Furthermore, the relationship between these modulated bacteria and their related metabolites, along with the mechanisms of how these changes led to cancer intervention are summarized.
Collapse
Affiliation(s)
- Hoi Kit Matthew Leung
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Emily Kwun Kwan Lo
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Fangfei Zhang
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Felicianna
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Marsena Jasiel Ismaiah
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Congjia Chen
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Hani El-Nezami
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| |
Collapse
|
15
|
Kandalgaonkar MR, Kumar V, Vijay‐Kumar M. Digestive dynamics: Unveiling interplay between the gut microbiota and the liver in macronutrient metabolism and hepatic metabolic health. Physiol Rep 2024; 12:e16114. [PMID: 38886098 PMCID: PMC11182692 DOI: 10.14814/phy2.16114] [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: 04/11/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Although the liver is the largest metabolic organ in the body, it is not alone in functionality and is assisted by "an organ inside an organ," the gut microbiota. This review attempts to shed light on the partnership between the liver and the gut microbiota in the metabolism of macronutrients (i.e., proteins, carbohydrates, and lipids). All nutrients absorbed by the small intestines are delivered to the liver for further metabolism. Undigested food that enters the colon is metabolized further by the gut microbiota that produces secondary metabolites, which are absorbed into portal circulation and reach the liver. These microbiota-derived metabolites and co-metabolites include ammonia, hydrogen sulfide, short-chain fatty acids, secondary bile acids, and trimethylamine N-oxide. Further, the liver produces several compounds, such as bile acids that can alter the gut microbial composition, which can in turn influence liver health. This review focuses on the metabolism of these microbiota metabolites and their influence on host physiology. Furthermore, the review briefly delineates the effect of the portosystemic shunt on the gut microbiota-liver axis, and current understanding of the treatments to target the gut microbiota-liver axis.
Collapse
Affiliation(s)
- Mrunmayee R. Kandalgaonkar
- Department of Physiology and PharmacologyUniversity of Toledo College of Medicine and Life SciencesToledoOhioUSA
| | - Virender Kumar
- College of Pharmacy and Pharmaceutical SciencesUniversity of ToledoToledoOhioUSA
| | - Matam Vijay‐Kumar
- Department of Physiology and PharmacologyUniversity of Toledo College of Medicine and Life SciencesToledoOhioUSA
| |
Collapse
|
16
|
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
|
17
|
Guzior DV, Okros M, Shivel M, Armwald B, Bridges C, Fu Y, Martin C, Schilmiller AL, Miller WM, Ziegler KM, Sims MD, Maddens ME, Graham SF, Hausinger RP, Quinn RA. Bile salt hydrolase acyltransferase activity expands bile acid diversity. Nature 2024; 626:852-858. [PMID: 38326608 DOI: 10.1038/s41586-024-07017-8] [Citation(s) in RCA: 49] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 01/02/2024] [Indexed: 02/09/2024]
Abstract
Bile acids (BAs) are steroid detergents in bile that contribute to the absorption of fats and fat-soluble vitamins while shaping the gut microbiome because of their antimicrobial properties1-4. Here we identify the enzyme responsible for a mechanism of BA metabolism by the gut microbiota involving amino acid conjugation to the acyl-site of BAs, thus producing a diverse suite of microbially conjugated bile acids (MCBAs). We show that this transformation is mediated by acyltransferase activity of bile salt hydrolase (bile salt hydrolase/transferase, BSH/T). Clostridium perfringens BSH/T rapidly performed acyl transfer when provided various amino acids and taurocholate, glycocholate or cholate, with an optimum at pH 5.3. Amino acid conjugation by C. perfringens BSH/T was diverse, including all proteinaceous amino acids except proline and aspartate. MCBA production was widespread among gut bacteria, with strain-specific amino acid use. Species with similar BSH/T amino acid sequences had similar conjugation profiles and several bsh/t alleles correlated with increased conjugation diversity. Tertiary structure mapping of BSH/T followed by mutagenesis experiments showed that active site structure affects amino acid selectivity. These MCBA products had antimicrobial properties, where greater amino acid hydrophobicity showed greater antimicrobial activity. Inhibitory concentrations of MCBAs reached those measured natively in the mammalian gut. MCBAs fed to mice entered enterohepatic circulation, in which liver and gallbladder concentrations varied depending on the conjugated amino acid. Quantifying MCBAs in human faecal samples showed that they reach concentrations equal to or greater than secondary and primary BAs and were reduced after bariatric surgery, thus supporting MCBAs as a significant component of the BA pool that can be altered by changes in gastrointestinal physiology. In conclusion, the inherent acyltransferase activity of BSH/T greatly diversifies BA chemistry, creating a set of previously underappreciated metabolites with the potential to affect the microbiome and human health.
Collapse
Affiliation(s)
- Douglas V Guzior
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Department of Microbiology, Genetics & Immunology, Michigan State University, East Lansing, MI, USA
| | - Maxwell Okros
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Madison Shivel
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Bruin Armwald
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Christopher Bridges
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Department of Microbiology, Genetics & Immunology, Michigan State University, East Lansing, MI, USA
| | - Yousi Fu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Christian Martin
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Anthony L Schilmiller
- Mass Spectrometry and Metabolomics Core, Michigan State University, East Lansing, MI, USA
| | - Wendy M Miller
- Corewell Health, William Beaumont University Hospital, Royal Oak, MI, USA
- Oakland University, William Beaumont School of Medicine, Rochester, MI, USA
| | - Kathryn M Ziegler
- Corewell Health, William Beaumont University Hospital, Royal Oak, MI, USA
- Oakland University, William Beaumont School of Medicine, Rochester, MI, USA
| | - Matthew D Sims
- Corewell Health, William Beaumont University Hospital, Royal Oak, MI, USA
- Oakland University, William Beaumont School of Medicine, Rochester, MI, USA
| | - Michael E Maddens
- Corewell Health, William Beaumont University Hospital, Royal Oak, MI, USA
- Oakland University, William Beaumont School of Medicine, Rochester, MI, USA
| | - Stewart F Graham
- Corewell Health, William Beaumont University Hospital, Royal Oak, MI, USA
- Oakland University, William Beaumont School of Medicine, Rochester, MI, USA
- Beaumont Research Institute, Royal Oak, MI, USA
| | - Robert P Hausinger
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Department of Microbiology, Genetics & Immunology, Michigan State University, East Lansing, MI, USA
| | - Robert A Quinn
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
18
|
Wang R, Wang M, Zeng Q, Wang L, Zhang Q, Pu S, Ma X, Wang J, Pan Y. Correlation between microbial characteristics and reproductive status of the yak uterus based on macrogenomic analysis. BMC Vet Res 2024; 20:4. [PMID: 38172906 PMCID: PMC10763020 DOI: 10.1186/s12917-023-03845-4] [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: 04/25/2023] [Accepted: 12/05/2023] [Indexed: 01/05/2024] Open
Abstract
INTRODUCTION This study aimed to investigate the microbial characteristics of yak uteri collected using intrauterine cotton swabs (CS) during different reproductive stages and the correlation of these microbial characteristics with reproductive status. METHODS We used a macrogenomic approach to analyze the functional aspects of different microorganisms in samples collected during the pre-estrus, estrus, late estrus, and diestrus stages. RESULTS The results revealed the presence of 1293 microbial genera and 3401 microbial species in the uteri of yaks at different reproductive stages. The dominant bacterial species varied across the different periods, with Micrococcus and Proteus being dominant during pre-estrus; Pseudomonas, Clostridium, Flavobacterium, Bacillus, and Staphylococcus during estrus; Acinetobacter, Bacillus and Proteus during late estrus; and Pseudomonas, Escherichia coli, and Proteus during diestrus. DISCUSSION The primary functions of these bacteria are enriched in various metabolic pathways, including carbohydrate and amino acid metabolism, intracellular transport and secretion, post-translational protein modification, and drug resistance. These findings suggest that the microbial diversity in the uterus of yaks plays a crucial role in reproductive regulation and can help prevent reproductive tract-related diseases.
Collapse
Affiliation(s)
- Rui Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Meng Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, Gansu, China
| | - Qiaoying Zeng
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Libin Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, Gansu, China
| | - Qian Zhang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, Gansu, China
| | - Sisi Pu
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Xin Ma
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, Gansu, China
| | - Jinglei Wang
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, Gansu, China
| | - Yangyang Pan
- College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, Gansu, China.
- Technology and Research Center of Gansu Province for Embryonic Engineering of Bovine and Sheep & Goat, Lanzhou, Gansu, China.
| |
Collapse
|
19
|
Pan Y, Zhang H, Li M, He T, Guo S, Zhu L, Tan J, Wang B. Novel approaches in IBD therapy: targeting the gut microbiota-bile acid axis. Gut Microbes 2024; 16:2356284. [PMID: 38769683 PMCID: PMC11110704 DOI: 10.1080/19490976.2024.2356284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/13/2024] [Indexed: 05/22/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic and recurrent condition affecting the gastrointestinal tract. Disturbed gut microbiota and abnormal bile acid (BA) metabolism are notable in IBD, suggesting a bidirectional relationship. Specifically, the diversity of the gut microbiota influences BA composition, whereas altered BA profiles can disrupt the microbiota. IBD patients often exhibit increased primary bile acid and reduced secondary bile acid concentrations due to a diminished bacteria population essential for BA metabolism. This imbalance activates BA receptors, undermining intestinal integrity and immune function. Consequently, targeting the microbiota-BA axis may rectify these disturbances, offering symptomatic relief in IBD. Here, the interplay between gut microbiota and bile acids (BAs) is reviewed, with a particular focus on the role of gut microbiota in mediating bile acid biotransformation, and contributions of the gut microbiota-BA axis to IBD pathology to unveil potential novel therapeutic avenues for IBD.
Collapse
Affiliation(s)
- Yinping Pan
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Haojie Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Minghui Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Tingjing He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Sihao Guo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Liancai Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| | - Jun Tan
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological & Chemical engineering, Chongqing University of Education, Chongqing, PR China
| | - Bochu Wang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, PR China
| |
Collapse
|
20
|
Reuter MA, Tucker M, Marfori Z, Shishani R, Bustamante JM, Moreno R, Goodson ML, Ehrlich A, Taha AY, Lein PJ, Joshi N, Brito I, Durbin-Johnson B, Nandakumar R, Cummings BP. Dietary resistant starch supplementation increases gut luminal deoxycholic acid abundance in mice. Gut Microbes 2024; 16:2315632. [PMID: 38375831 PMCID: PMC10880513 DOI: 10.1080/19490976.2024.2315632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 02/02/2024] [Indexed: 02/21/2024] Open
Abstract
Bile acids (BA) are among the most abundant metabolites produced by the gut microbiome. Primary BAs produced in the liver are converted by gut bacterial 7-α-dehydroxylation into secondary BAs, which can differentially regulate host health via signaling based on their varying affinity for BA receptors. Despite the importance of secondary BAs in host health, the regulation of 7-α-dehydroxylation and the role of diet in modulating this process is incompletely defined. Understanding this process could lead to dietary guidelines that beneficially shift BA metabolism. Dietary fiber regulates gut microbial composition and metabolite production. We tested the hypothesis that feeding mice a diet rich in a fermentable dietary fiber, resistant starch (RS), would alter gut bacterial BA metabolism. Male and female wild-type mice were fed a diet supplemented with RS or an isocaloric control diet (IC). Metabolic parameters were similar between groups. RS supplementation increased gut luminal deoxycholic acid (DCA) abundance. However, gut luminal cholic acid (CA) abundance, the substrate for 7-α-dehydroxylation in DCA production, was unaltered by RS. Further, RS supplementation did not change the mRNA expression of hepatic BA producing enzymes or ileal BA transporters. Metagenomic assessment of gut bacterial composition revealed no change in the relative abundance of bacteria known to perform 7-α-dehydroxylation. P. ginsenosidimutans and P. multiformis were positively correlated with gut luminal DCA abundance and increased in response to RS supplementation. These data demonstrate that RS supplementation enriches gut luminal DCA abundance without increasing the relative abundance of bacteria known to perform 7-α-dehydroxylation.
Collapse
Affiliation(s)
- Melanie A. Reuter
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California – Davis, Sacramento, CA, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California – Davis, Davis, CA, USA
| | - Madelynn Tucker
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California – Davis, Sacramento, CA, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California – Davis, Davis, CA, USA
| | - Zara Marfori
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California – Davis, Sacramento, CA, USA
| | - Rahaf Shishani
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California – Davis, Sacramento, CA, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California – Davis, Davis, CA, USA
| | - Jessica Miranda Bustamante
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California – Davis, Sacramento, CA, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California – Davis, Davis, CA, USA
| | - Rosalinda Moreno
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California – Davis, Sacramento, CA, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California – Davis, Davis, CA, USA
| | - Michael L. Goodson
- Department of Environmental Toxicology, College of Agricultural and Environmental Sciences, University of California – Davis, Davis, CA, USA
| | - Allison Ehrlich
- Department of Environmental Toxicology, College of Agricultural and Environmental Sciences, University of California – Davis, Davis, CA, USA
| | - Ameer Y. Taha
- Department of Food Science and Technology, University of California - Davis, Davis, CA, USA
| | - Pamela J. Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California – Davis, Davis, CA, USA
| | - Nikhil Joshi
- Bioinformatics Core, UC Davis Genome Center, University of California – Davis, Davis, CA, USA
| | - Ilana Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Blythe Durbin-Johnson
- Bioinformatics Core, UC Davis Genome Center, University of California – Davis, Davis, CA, USA
| | - Renu Nandakumar
- Biomarkers Core Laboratory, Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, NY, USA
| | - Bethany P. Cummings
- Department of Surgery, Center for Alimentary and Metabolic Sciences, School of Medicine, University of California – Davis, Sacramento, CA, USA
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California – Davis, Davis, CA, USA
| |
Collapse
|
21
|
McMillan AS, Theriot CM. Bile acids impact the microbiota, host, and C. difficile dynamics providing insight into mechanisms of efficacy of FMTs and microbiota-focused therapeutics. Gut Microbes 2024; 16:2393766. [PMID: 39224076 PMCID: PMC11376424 DOI: 10.1080/19490976.2024.2393766] [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: 03/27/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
Clostridioides difficile is a major nosocomial pathogen, causing significant morbidity and mortality worldwide. Antibiotic usage, a major risk factor for Clostridioides difficile infection (CDI), disrupts the gut microbiota, allowing C. difficile to proliferate and cause infection, and can often lead to recurrent CDI (rCDI). Fecal microbiota transplantation (FMT) and live biotherapeutic products (LBPs) have emerged as effective treatments for rCDI and aim to restore colonization resistance provided by a healthy gut microbiota. However, much is still unknown about the mechanisms mediating their success. Bile acids, extensively modified by gut microbes, affect C. difficile's germination, growth, and toxin production while also shaping the gut microbiota and influencing host immune responses. Additionally, microbial interactions, such as nutrient competition and cross-feeding, contribute to colonization resistance against C. difficile and may contribute to the success of microbiota-focused therapeutics. Bile acids as well as other microbial mediated interactions could have implications for other diseases being treated with microbiota-focused therapeutics. This review focuses on the intricate interplay between bile acid modifications, microbial ecology, and host responses with a focus on C. difficile, hoping to shed light on how to move forward with the development of new microbiota mediated therapeutic strategies to combat rCDI and other intestinal diseases.
Collapse
Affiliation(s)
- Arthur S. McMillan
- Genetics Program, Department of Biological Sciences, College of Science, North Carolina State University, Raleigh, NC, USA
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Casey M. Theriot
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
22
|
Meibom KL, Marion S, Volet C, Nass T, Vico-Oton E, Menin L, Bernier-Latmani R. BaiJ and BaiB are key enzymes in the chenodeoxycholic acid 7α-dehydroxylation pathway in the gut microbe Clostridium scindens ATCC 35704. Gut Microbes 2024; 16:2323233. [PMID: 38465624 PMCID: PMC10936602 DOI: 10.1080/19490976.2024.2323233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 02/21/2024] [Indexed: 03/12/2024] Open
Abstract
Bile acid transformation is a common gut microbiome activity that produces secondary bile acids, some of which are important for human health. One such process, 7α-dehydroxylation, converts the primary bile acids, cholic acid and chenodeoxycholic acid, to deoxycholic acid and lithocholic acid, respectively. This transformation requires a number of enzymes, generally encoded in a bile acid-inducible (bai) operon and consists of multiple steps. Some 7α-dehydroxylating bacteria also harbor additional genes that encode enzymes with potential roles in this pathway, but little is known about their functions. Here, we purified 11 enzymes originating either from the bai operon or encoded at other locations in the genome of Clostridium scindens strain ATCC 35704. Enzyme activity was probed in vitro under anoxic conditions to characterize the biochemical pathway of chenodeoxycholic acid 7α-dehydroxylation. We found that more than one combination of enzymes can support the process and that a set of five enzymes, including BaiJ that is encoded outside the bai operon, is sufficient to achieve the transformation. We found that BaiJ, an oxidoreductase, exhibits an activity that is not harbored by the homologous enzyme from another C. scindens strain. Furthermore, ligation of bile acids to coenzyme A (CoA) was shown to impact the product of the transformation. These results point to differences in the 7α-dehydroxylation pathway among microorganisms and the crucial role of CoA ligation in the process.
Collapse
Affiliation(s)
- Karin Lederballe Meibom
- Environmental Microbiology Laboratory, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Solenne Marion
- Environmental Microbiology Laboratory, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Colin Volet
- Environmental Microbiology Laboratory, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Théo Nass
- Environmental Microbiology Laboratory, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Eduard Vico-Oton
- Environmental Microbiology Laboratory, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Laure Menin
- Institute of Chemical Sciences and Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Rizlan Bernier-Latmani
- Environmental Microbiology Laboratory, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
23
|
Cifuentes-Silva E, Cabello-Verrugio C. Bile Acids as Signaling Molecules: Role of Ursodeoxycholic Acid in Cholestatic Liver Disease. Curr Protein Pept Sci 2024; 25:206-214. [PMID: 37594109 DOI: 10.2174/1389203724666230818092800] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/05/2023] [Accepted: 07/18/2023] [Indexed: 08/19/2023]
Abstract
Ursodeoxycholic acid (UDCA) is a natural substance physiologically produced in the liver. Initially used to dissolve gallstones, it is now successfully used in treating primary biliary cirrhosis and as adjuvant therapy for various hepatobiliary cholestatic diseases. However, the mechanisms underlying its beneficial effects still need to be clarified. Evidence suggests three mechanisms of action for UDCA that could benefit humans with cholestatic liver disease (CLD): protection of cholangiocytes against hydrophobic bile acid (BA) cytotoxicity, stimulation of hepatobiliary excretion, and protection of hepatocytes against BA-induced apoptosis. These mechanisms may act individually or together to potentiate them. At the molecular level, it has been observed that UDCA can generate modifications in the transcription and translation of proteins essential in the transport of BA, correcting the deficit in BA secretion in CLD, in addition to activating signaling pathways to translocate these transporters to the sites where they should fulfill their function. Inhibition of BA-induced hepatocyte apoptosis may play a role in CLD, characterized by BA retention in the hepatocyte. Thus, different mechanisms of action contribute to the improvement after UDCA administration in CLD. On the other hand, the effects of UDCA on tissues that possess receptors that may interact with BAs in pathological contexts, such as skeletal muscle, are still unclear. This work aims to describe the main molecular mechanisms by which UDCA acts in the human body, emphasizing the interaction in tissues other than the liver.
Collapse
Affiliation(s)
- Eduardo Cifuentes-Silva
- Laboratory of Muscle Pathology, Fragility, and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Laboratory of Muscle Pathology, Fragility, and Aging, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
24
|
Kiriyama Y, Nochi H. The Role of Gut Microbiota-Derived Lithocholic Acid, Deoxycholic Acid and Their Derivatives on the Function and Differentiation of Immune Cells. Microorganisms 2023; 11:2730. [PMID: 38004742 PMCID: PMC10672800 DOI: 10.3390/microorganisms11112730] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/24/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
A wide variety and large number of bacterial species live in the gut, forming the gut microbiota. Gut microbiota not only coexist harmoniously with their hosts, but they also induce significant effects on each other. The composition of the gut microbiota can be changed due to environmental factors such as diet and antibiotic intake. In contrast, alterations in the composition of the gut microbiota have been reported in a variety of diseases, including intestinal, allergic, and autoimmune diseases and cancer. The gut microbiota metabolize exogenous dietary components ingested from outside the body to produce short-chain fatty acids (SCFAs) and amino acid metabolites. Unlike SCFAs and amino acid metabolites, the source of bile acids (BAs) produced by the gut microbiota is endogenous BAs from the liver. The gut microbiota metabolize BAs to generate secondary bile acids, such as lithocholic acid (LCA), deoxycholic acid (DCA), and their derivatives, which have recently been shown to play important roles in immune cells. This review focuses on current knowledge of the role of LCA, DCA, and their derivatives on immune cells.
Collapse
Affiliation(s)
- Yoshimitsu Kiriyama
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki 769-2193, Japan;
- Institute of Neuroscience, Tokushima Bunri University, Sanuki 769-2193, Japan
| | - Hiromi Nochi
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University, Sanuki 769-2193, Japan;
| |
Collapse
|
25
|
Olivos Caicedo KY, Fernandez-Materan FV, Hernandez AG, Daniel SL, Alves JMP, Ridlon JM. Complete genome sequence of the archetype bile acid 7α-dehydroxylating bacterium, Clostridium scindens VPI12708, isolated from human feces, circa 1980. Microbiol Resour Announc 2023; 12:e0002923. [PMID: 37555681 PMCID: PMC10508169 DOI: 10.1128/mra.00029-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 06/17/2023] [Indexed: 08/10/2023] Open
Abstract
Clostridium scindens strain VPI12708 serves as model organism to study bile acid 7α-dehydroxylating pathways. The closed circular genome of C. scindens VPI12708 was obtained by PacBio sequencing. The genome is composed of 3,983,052 bp, with 47.59% G + C, and 3,707 coding DNA sequences are predicted.
Collapse
Affiliation(s)
| | - Francelys V. Fernandez-Materan
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Alvaro G. Hernandez
- Keck Center for Biotechnology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Steven L. Daniel
- Department of Biological Sciences, Eastern Illinois University, Charleston, Illinois, USA
| | - Joao M. P. Alves
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Jason M. Ridlon
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Cancer Center at Illinois, Urbana, Illinois, USA
- Center for Advanced Study, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
26
|
Ridlon JM, Daniel SL, Gaskins HR. The Hylemon-Björkhem pathway of bile acid 7-dehydroxylation: history, biochemistry, and microbiology. J Lipid Res 2023; 64:100392. [PMID: 37211250 PMCID: PMC10382948 DOI: 10.1016/j.jlr.2023.100392] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/07/2023] [Accepted: 05/11/2023] [Indexed: 05/23/2023] Open
Abstract
Bile acids are detergents derived from cholesterol that function to solubilize dietary lipids, remove cholesterol from the body, and act as nutrient signaling molecules in numerous tissues with functions in the liver and gut being the best understood. Studies in the early 20th century established the structures of bile acids, and by mid-century, the application of gnotobiology to bile acids allowed differentiation of host-derived "primary" bile acids from "secondary" bile acids generated by host-associated microbiota. In 1960, radiolabeling studies in rodent models led to determination of the stereochemistry of the bile acid 7-dehydration reaction. A two-step mechanism was proposed, which we have termed the Samuelsson-Bergström model, to explain the formation of deoxycholic acid. Subsequent studies with humans, rodents, and cell extracts of Clostridium scindens VPI 12708 led to the realization that bile acid 7-dehydroxylation is a result of a multi-step, bifurcating pathway that we have named the Hylemon-Björkhem pathway. Due to the importance of hydrophobic secondary bile acids and the increasing measurement of microbial bai genes encoding the enzymes that produce them in stool metagenome studies, it is important to understand their origin.
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 and Immunology, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.
| | - Steven L Daniel
- Department of Biological Sciences, Eastern Illinois University, Charleston, IL, 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
|
27
|
Zhao H, Yang CE, Liu T, Zhang MX, Niu Y, Wang M, Yu J. The roles of gut microbiota and its metabolites in diabetic nephropathy. Front Microbiol 2023; 14:1207132. [PMID: 37577423 PMCID: PMC10413983 DOI: 10.3389/fmicb.2023.1207132] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 07/13/2023] [Indexed: 08/15/2023] Open
Abstract
Diabetic nephropathy (DN) is a severe microvascular complication of diabetes, which increases the risk of renal failure and causes a high global disease burden. Due to the lack of sustainable treatment, DN has become the primary cause of end-stage renal disease worldwide. Gut microbiota and its metabolites exert critical regulatory functions in maintaining host health and are associated with many pathogenesis of aging-related chronic diseases. Currently, the theory gut-kidney axis has opened a novel angle to understand the relationship between gut microbiota and multiple kidney diseases. In recent years, accumulating evidence has revealed that the gut microbiota and their metabolites play an essential role in the pathophysiologic processes of DN through the gut-kidney axis. In this review, we summarize the current investigations of gut microbiota and microbial metabolites involvement in the progression of DN, and further discuss the potential gut microbiota-targeted therapeutic approaches for DN.
Collapse
Affiliation(s)
- Hui Zhao
- Clinical Experimental Center, Xi’an Engineering Technology Research Center for Cardiovascular Active Peptides, the Affiliated Xi’an International Medical Center Hospital, Northwest University, Xi’an, Shaanxi, China
- Faculty of Life Science and Medicine, Northwest University, Xi’an, Shaanxi, China
| | - Cheng-E Yang
- Department of Cardiology, Xi'an International Medical Center Hospital, Xi’an, Shaanxi, China
| | - Tian Liu
- Clinical Experimental Center, Xi’an Engineering Technology Research Center for Cardiovascular Active Peptides, the Affiliated Xi’an International Medical Center Hospital, Northwest University, Xi’an, Shaanxi, China
| | - Ming-Xia Zhang
- Clinical Experimental Center, Xi’an Engineering Technology Research Center for Cardiovascular Active Peptides, the Affiliated Xi’an International Medical Center Hospital, Northwest University, Xi’an, Shaanxi, China
| | - Yan Niu
- Clinical Experimental Center, Xi’an Engineering Technology Research Center for Cardiovascular Active Peptides, the Affiliated Xi’an International Medical Center Hospital, Northwest University, Xi’an, Shaanxi, China
| | - Ming Wang
- College of Food Science and Engineering, Northwest University, Xi’an, Shaanxi, China
| | - Jun Yu
- Clinical Experimental Center, Xi’an Engineering Technology Research Center for Cardiovascular Active Peptides, the Affiliated Xi’an International Medical Center Hospital, Northwest University, Xi’an, Shaanxi, China
| |
Collapse
|
28
|
Jing N, Wang L, Zhuang H, Jiang G, Liu Z. Enhancing therapeutic effects of murine cancer vaccine by reshaping gut microbiota with Lactobacillus rhamnosus GG and jujube powder. Front Immunol 2023; 14:1195075. [PMID: 37435064 PMCID: PMC10332846 DOI: 10.3389/fimmu.2023.1195075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/09/2023] [Indexed: 07/13/2023] Open
Abstract
Cancer vaccines have gained widespread attention in recent years as an emerging treatment for tumors. However, most therapeutic cancer vaccines have failed in phase III clinical trials due to faint clinical benefits. In this study, we funded that a specific synbiotic composing Lactobacillus rhamnosus GG (LGG) and jujube powder significantly enhanced the therapeutic effects of whole cells cancer vaccine in MC38 cancer cells bearing-mouse. The utilization of LGG increased the abundance of Muribaculaceae, which is conductive to an enhanced anti-tumor effect, but reduced microbial α-diversity. The use of jujube nursed probiotic microorganisms in Lachnospiaceae and enriched microbial diversity, as indicated by increased Shannon and Chao index. The reshaped gut microbiota by this synbiotic improved lipid metabolism conductive to intensified infiltration of CD8+ T cells in the tumor microenvironment and enhanced the potency of above-mentioned cancer vaccine. These encouraging findings are helpful for further efforts towards enhancing the therapeutic effects of cancer vaccines through nutritional intervention.
Collapse
Affiliation(s)
- Nan Jing
- Department of Chemical Engineering, Tsinghua University, Beijing, China
- Key Lab of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, China
| | - Luoyang Wang
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Huiren Zhuang
- Department of Chemical Engineering, Tsinghua University, Beijing, China
- Key Lab of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, China
| | - Guoqiang Jiang
- Department of Chemical Engineering, Tsinghua University, Beijing, China
- Key Lab of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, China
| | - Zheng Liu
- Department of Chemical Engineering, Tsinghua University, Beijing, China
- Key Lab of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, China
| |
Collapse
|
29
|
Abstract
Cardiometabolic disease comprises cardiovascular and metabolic dysfunction and underlies the leading causes of morbidity and mortality, both within the United States and worldwide. Commensal microbiota are implicated in the development of cardiometabolic disease. Evidence suggests that the microbiome is relatively variable during infancy and early childhood, becoming more fixed in later childhood and adulthood. Effects of microbiota, both during early development, and in later life, may induce changes in host metabolism that modulate risk mechanisms and predispose toward the development of cardiometabolic disease. In this review, we summarize the factors that influence gut microbiome composition and function during early life and explore how changes in microbiota and microbial metabolism influence host metabolism and cardiometabolic risk throughout life. We highlight limitations in current methodology and approaches and outline state-of-the-art advances, which are improving research and building toward refined diagnosis and treatment options in microbiome-targeted therapies.
Collapse
Affiliation(s)
- Curtis L Gabriel
- Division of Gastroenterology, Hepatology and Nutrition (C.L.G.), Vanderbilt University Medical Center, Nashville
- Tennessee Center for AIDS Research (C.L.G.), Vanderbilt University Medical Center, Nashville
| | - Jane F Ferguson
- Division of Cardiovascular Medicine (J.F.F.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Microbiome Innovation Center (J.F.F.), Vanderbilt University Medical Center, Nashville
- Vanderbilt Institute for Infection, Immunology, and Inflammation (J.F.F.), Vanderbilt University Medical Center, Nashville
| |
Collapse
|