1
|
Zhang J, Zhang X, Liu H, Wang P, Li L, Bionaz M, Lin P, Yao J. Altered bile acid and correlations with gut microbiome in transition dairy cows with different glucose and lipid metabolism status. J Dairy Sci 2024; 107:9915-9933. [PMID: 38908707 DOI: 10.3168/jds.2024-24658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/22/2024] [Indexed: 06/24/2024]
Abstract
The transition from pregnancy to lactation is critical in dairy cows. Among others, dairy cows experience a metabolic stress due to a large change in glucose and lipid metabolism. Recent studies revealed that bile acids (BA), other than being involved in both the emulsification and solubilization of fats during intestinal absorption, can also affect the metabolism of glucose and lipids, both directly or indirectly by affecting the gut microbiota. Thus, we used untargeted and targeted metabolomics and 16S rRNA gene sequencing approaches to investigate the concentration of plasma metabolites and BA, the composition of the rectum microbial community, and assess their interaction in transition dairy cows. In Experiment 1, we investigated BA and other blood parameters and gut microbiota in dairy cows without clinical diseases during the transition period, which can be seen as well adapted to the challenge of changed glucose and lipid metabolism. As expected, we detected an increased plasma concentrations of BHB and nonesterified fatty acids (NEFA) but decreased concentrations of glucose, cholesterol, and triglycerides (TG). Untargeted metabolomic analysis of the plasma revealed primary BA biosynthesis was one of the affected pathways, and was consistent with the increased concentration of BA in the plasma. A correlation approach revealed a complex association between BA and microbiota with the host plasma concentration of glucose and lipid metabolites. Among BA, chenodeoxycholic acid derivates such as glycolithocholic acid, taurolithocholic acid, lithocholic acid, taurochenodeoxycholic acid, and taurodeoxycholic acid were the main hub nodes connecting microbe and blood metabolites (such as glucose, TG, and NEFA). In Experiment 2, we investigated early postpartum dairy cows with or without hyperketonemia (HPK). As expected, HPK cows had increased concentration of NEFA and decreased concentrations of glucose and triglycerides. The untargeted metabolomic analysis of the plasma revealed that primary BA biosynthesis was also one of the affected pathways. Even though the BA concentration was similar among the 2 groups, the profiles of taurine-conjugated BA changed significantly. A correlation analysis also revealed an association between BA and microbiota with the concentration in plasma of glucose and lipid metabolites (such as BHB). Among BA, cholic acid and its derivates such as taurocholic acid, tauro α-muricholic acid, and taurodeoxycholic acid were the main hub nodes connecting microbe and blood metabolites. Our results indicated an association between BA, intestinal microbe, and glucose and lipid metabolism in transition dairy cows. These findings provide new insight into the adaptation mechanisms of dairy cows during the transition period.
Collapse
Affiliation(s)
- Jun Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xia Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Huifeng Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Peiyue Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Lei Li
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97331
| | - Pengfei Lin
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
2
|
Fang Y, Qin M, Zheng Q, Wang K, Han X, Yang Q, Sang X, Cao G. Role of Bile Acid Receptors in the Development and Function of Diabetic Nephropathy. Kidney Int Rep 2024; 9:3116-3133. [PMID: 39534198 PMCID: PMC11551060 DOI: 10.1016/j.ekir.2024.08.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/25/2024] [Accepted: 08/04/2024] [Indexed: 11/16/2024] Open
Abstract
Diabetic nephropathy (DN) is a prevalent microvascular complication that occurs often in individuals with diabetes. It significantly raises the mortality rate of affected patients. Therefore, there is an urgent need to identify therapeutic targets for controlling and preventing the occurrence and development of DN. Bile acids (BAs) are now recognized as intricate metabolic integrators and signaling molecules. The activation of BAs has great promise as a therapeutic approach for preventing DN, renal damage caused by obesity, and nephrosclerosis. The nuclear receptors (NRs), farnesoid X receptor (FXR), pregnane X receptor (PXR), vitamin D receptor (VDR); and the G protein-coupled BA receptor, Takeda G-protein-coupled receptor 5 (TGR5) have important functions in controlling lipid, glucose, and energy metabolism, inflammation, as well as drug metabolism and detoxification. Over the past 10 years, there has been advancement in comprehending the biology and processes of BA receptors in the kidney, as well as in the creation of targeted BA receptor agonists. In this review, we discuss the role of BA receptors, FXR, PXR, VDR, and TGR5 in DN and their role in renal physiology, as well as the development and application of agonists that activate BA receptors for the treatment of kidney diseases.
Collapse
Affiliation(s)
- Yuanyuan Fang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Minjing Qin
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qitong Zheng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kuilong Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Han
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qiao Yang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xia'nan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
3
|
Kugler BA, Maurer A, Fu X, Franczak E, Ernst N, Schwartze K, Allen J, Li T, Crawford PA, Koch LG, Britton SL, Burgess SC, Thyfault JP. Aerobic capacity and exercise mediate protection against hepatic steatosis via enhanced bile acid metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.21.619494. [PMID: 39484384 PMCID: PMC11526936 DOI: 10.1101/2024.10.21.619494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
High cardiorespiratory fitness and exercise show evidence of altering bile acid (BA) metabolism and are known to protect or treat diet-induced hepatic steatosis, respectively. Here, we tested the hypothesis that high intrinsic aerobic capacity and exercise both increase hepatic BA synthesis measured by the incorporation of 2H2O. We also leveraged mice with inducible liver-specific deletion of Cyp7a1 (LCyp7a1KO), which encodes the rate-limiting enzyme for BA synthesis, to test if exercise-induced BA synthesis is critical for exercise to reduce hepatic steatosis. The synthesis of hepatic BA, cholesterol, and de novo lipogenesis was measured in rats bred for either high (HCR) vs. low (LCR) aerobic capacity consuming acute and chronic high-fat diets. HCR rats had increased synthesis of cholesterol and certain BA species in the liver compared to LCR rats. We also found that chronic exercise with voluntary wheel running (VWR) (4 weeks) increased newly synthesized BAs of specific species in male C57BL/6J mice compared to sedentary mice. Loss of Cyp7a1 resulted in fewer new BAs and increased liver triglycerides compared to controls after a 10-week high-fat diet. Additionally, exercise via VWR for 4 weeks effectively reduced hepatic triglycerides in the high-fat diet-fed control male and female mice as expected; however, exercise in LCyp7a1KO mice did not lower liver triglycerides in either sex. These results show that aerobic capacity and exercise increase hepatic BA metabolism, which may be critical for combatting hepatic steatosis.
Collapse
Affiliation(s)
- Benjamin A. Kugler
- Departments of Cell Biology and Physiology
- Internal Medicine, Division of Endocrinology and Clinical Pharmacology and KU Diabetes Institute
| | | | - Xiaorong Fu
- Center for Human Nutrition and Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Edziu Franczak
- Departments of Cell Biology and Physiology
- Internal Medicine, Division of Endocrinology and Clinical Pharmacology and KU Diabetes Institute
| | - Nick Ernst
- Departments of Cell Biology and Physiology
| | | | | | - Tiangang Li
- Department of Biochemistry and Physiology, and Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter A. Crawford
- Division of Molecular Medicine, Department of Medicine, and Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN
| | - Lauren G. Koch
- Dept of Physiology and Pharmacology, The University of Toledo, Toledo, OH, USA
| | | | - Shawn C. Burgess
- Center for Human Nutrition and Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John P. Thyfault
- Departments of Cell Biology and Physiology
- Internal Medicine, Division of Endocrinology and Clinical Pharmacology and KU Diabetes Institute
- Kansas Center for Metabolism and Obesity Research, Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
4
|
Jiang Z, Qian M, Zhen Z, Yang X, Xu C, Zuo L, Jiang J, Zhang W, Hu N. Gut microbiota and metabolomic profile changes play critical roles in tacrolimus-induced diabetes in rats. Front Cell Infect Microbiol 2024; 14:1436477. [PMID: 39355267 PMCID: PMC11442430 DOI: 10.3389/fcimb.2024.1436477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 08/14/2024] [Indexed: 10/03/2024] Open
Abstract
Aims Hyperglycemia is one of the adverse effects of tacrolimus (TAC), but the underlying mechanism is not fully identified. We used multi-omics analysis to evaluate the changes in the gut microbiota and metabolic profile of rats with TAC-induced diabetes. Methods To establish a diabetic animal model, Sprague Dawley rats were divided randomly into two groups. Those in the TAC group received intraperitoneal injections of TAC (3 mg/kg) for 8 weeks, and those in the CON group served as the control. 16S rRNA sequencing was used to analyze fecal microbiota. The metabolites of the two groups were detected and analyzed by nontargeted and targeted metabolomics, including amino acids (AAs), bile acids (BAs), and short-chain fatty acids (SCFAs). Results The rats treated with TAC exhibited hyperglycemia as well as changes in the gut microbiota and metabolites. Specifically, their gut microbiota had significantly higher abundances of Escherichia-Shigella, Enterococcus, and Allobaculum, and significantly lower abundances of Ruminococcus, Akkermansia, and Roseburia. In addition, they had significantly reduced serum levels of AAs including asparagine, aspartic acid, glutamic acid, and methionine. With respect to BAs, they had significantly higher serum levels of taurocholic acid (TCA), and glycochenodeoxycholic acid (GCDCA), but significantly lower levels of taurodeoxycholic acid (TDCA) and tauroursodeoxycholic acid (TUDCA). There were no differences in the levels of SCFAs between the two groups. Correlations existed among glucose metabolism indexes (fasting blood glucose and fasting insulin), gut microbiota (Ruminococcus and Akkermansia), and metabolites (glutamic acid, hydroxyproline, GCDCA, TDCA, and TUDCA). Conclusions Both AAs and BAs may play crucial roles as signaling molecules in the regulation of TAC-induced diabetes.
Collapse
Affiliation(s)
- Zhenwei Jiang
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, China
| | - Minyan Qian
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, China
| | - Zeng Zhen
- Changzhou Key Laboratory of Human Use Experience Research & Transformation of Menghe Medical School, Changzhou Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou, China
| | - Xuping Yang
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, China
| | - Caomei Xu
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, China
| | - Li'an Zuo
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wenting Zhang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Pediatric Central Laboratory, Affiliated Changzhou Children's Hospital of Nantong University, Changzhou, China
| | - Nan Hu
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University/The First People's Hospital of Changzhou, Changzhou, China
| |
Collapse
|
5
|
Wang L, Sun Y, Du L, Wang Q, Zhan M, Li S, Xiao X. Daily koumiss has positive regulatory effects on blood lipids and immune system: A metabolomics study. Heliyon 2024; 10:e36429. [PMID: 39253138 PMCID: PMC11382052 DOI: 10.1016/j.heliyon.2024.e36429] [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: 11/04/2023] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 09/11/2024] Open
Abstract
Koumiss, a traditional Mongolian beverage, is believed to possess high nutritional value and potential medical benefits. However, there is a lack of comprehensive research on its potential impact on the human body. Metabolomics, as a sensitive approach in systems biology, offers a new avenue for studying the overall effects of koumiss. In this work, metabolomics was utilized to identify potential biomarkers and pathways associated with koumiss using UPLC-MS detection, pattern recognition analysis, pathway enrichment, network pharmacology. The findings indicated that koumiss exerts a beneficial regulatory influence on lipid metabolism, neurotransmitters, hormones, phospholipids and arachidonic acid metabolism, besides up regulating the content of nutrients. It could reduce the risks of dyslipidemia and inflammatory responses. This study confirmed the benign regulatory effect of koumiss on normal organism from the perspective of endogenous metabolites, and provided objective support for the promotion and application of this ethnic food.
Collapse
Affiliation(s)
- Leqi Wang
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuanfang Sun
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Lijing Du
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Qian Wang
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Zhan
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shasha Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xue Xiao
- Institute of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
6
|
Gonzalez E, Lee MD, Tierney BT, Lipieta N, Flores P, Mishra M, Beckett L, Finkelstein A, Mo A, Walton P, Karouia F, Barker R, Jansen RJ, Green SJ, Weging S, Kelliher J, Singh NK, Bezdan D, Galazska J, Brereton NJB. Spaceflight alters host-gut microbiota interactions. NPJ Biofilms Microbiomes 2024; 10:71. [PMID: 39209868 PMCID: PMC11362537 DOI: 10.1038/s41522-024-00545-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
The ISS rodent habitat has provided crucial insights into the impact of spaceflight on mammals, inducing symptoms characteristic of liver disease, insulin resistance, osteopenia, and myopathy. Although these physiological responses can involve the microbiome on Earth, host-microbiota interactions during spaceflight are still being elucidated. We explore murine gut microbiota and host gene expression in the colon and liver after 29 and 56 days of spaceflight using multiomics. Metagenomics revealed significant changes in 44 microbiome species, including relative reductions in bile acid and butyrate metabolising bacteria like Extibacter muris and Dysosmobacter welbionis. Functional prediction indicate over-representation of fatty acid and bile acid metabolism, extracellular matrix interactions, and antibiotic resistance genes. Host gene expression described corresponding changes to bile acid and energy metabolism, and immune suppression. These changes imply that interactions at the host-gut microbiome interface contribute to spaceflight pathology and that these interactions might critically influence human health and long-duration spaceflight feasibility.
Collapse
Affiliation(s)
- E Gonzalez
- Microbiome Unit, Canadian Centre for Computational Genomics, Department of Human Genetics, McGill University, Montréal, Canada
- Centre for Microbiome Research, McGill University, Montréal, Canada
| | - M D Lee
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
| | - B T Tierney
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - N Lipieta
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, 02142, USA
| | - P Flores
- BioServe Space Technologies, University of Colorado Boulder, Boulder, CO, USA
| | - M Mishra
- Grossman School of Medicine, New York University, New York, USA
| | - L Beckett
- University of Nottingham, Nottingham, NG7 2RD, UK
| | - A Finkelstein
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - A Mo
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - P Walton
- NASA GeneLab for High Schools (GL4HS) program, NASA Ames Research Centre, Moffett Field, CA, USA
| | - F Karouia
- Exobiology Branch, NASA Ames Research Centre, Moffett Field, CA, USA
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Centre for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - R Barker
- Blue Marble Space Institute of Science, Seattle, WA, USA
- Yuri GmbH, Wiesentalstr. 40, 88074, Meckenbeuren, Germany
- University of Wisconsin-Madison, Madison, WI, USA
| | - R J Jansen
- Department of Public Health, North Dakota State University, Fargo, ND, USA
- Genomics, Phenomics, and Bioinformatics Program, North Dakota State University, Fargo, ND, USA
| | - S J Green
- Genomics and Microbiome Core Facility, Rush University Medical Centre, 1653 W. Congress Parkway, Chicago, IL, 60612, USA
| | - S Weging
- Institute of Computer Science, Martin-Luther University Halle-Wittenberg, Halle, Germany
| | - J Kelliher
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - N K Singh
- Department of Industrial Relations, Division of Occupational Safety and Health, Oakland, USA
| | - D Bezdan
- University of Wisconsin-Madison, Madison, WI, USA
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
- NGS Competence Centre Tübingen (NCCT), University of Tübingen, Tübingen, Germany
| | - J Galazska
- Space Biosciences Research Branch, NASA Ames Research Centre, Moffett Field, CA, USA
| | - N J B Brereton
- School of Biology and Environmental Science, University College Dublin, Dublin, Ireland.
| |
Collapse
|
7
|
Güven Gülhan Ü, Nikerel E, Çakır T, Erdoğan Sevilgen F, Durmuş S. Species-level identification of enterotype-specific microbial markers for colorectal cancer and adenoma. Mol Omics 2024; 20:397-416. [PMID: 38780313 DOI: 10.1039/d4mo00016a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Enterotypes have been shown to be an important factor for population stratification based on gut microbiota composition, leading to a better understanding of human health and disease states. Classifications based on compositional patterns will have implications for personalized microbiota-based solutions. There have been limited enterotype based studies on colorectal adenoma and cancer. Here, an enterotype-based meta-analysis of fecal shotgun metagenomic studies was performed, including 1579 samples of healthy controls (CTR), colorectal adenoma (ADN) and colorectal cancer (CRC) in total. Gut microbiota of healthy people were clustered into three enterotypes (Ruminococcus-, Bacteroides- and Prevotella-dominated enterotypes). Reference-based enterotype assignments were performed for CRC and ADN samples, using the supervised machine learning algorithm, K-nearest neighbors. Differential abundance analyses and random forest classification were conducted on each enterotype between healthy controls and CRC-ADN groups, revealing novel enterotype-specific microbial markers for non-invasive CRC screening strategies. Furthermore, we identified microbial species unique to each enterotype that play a role in the production of secondary bile acids and short-chain fatty acids, unveiling the correlation between cancer-associated gut microbes and dietary patterns. The enterotype-based approach in this study is promising in elucidating the mechanisms of differential gut microbiome profiles, thereby improving the efficacy of personalized microbiota-based solutions.
Collapse
Affiliation(s)
- Ünzile Güven Gülhan
- Department of Bioengineering, Gebze Technical University, Gebze, TR 41400, Turkey.
| | - Emrah Nikerel
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, TR 34755, Turkey
| | - Tunahan Çakır
- Department of Bioengineering, Gebze Technical University, Gebze, TR 41400, Turkey.
- PhiTech Bioinformatics, Gebze, TR 41470, Turkey
| | - Fatih Erdoğan Sevilgen
- The Institute for Data Science & Artificial Intelligence, Boğaziçi University, Istanbul, TR 34342, Turkey
- PhiTech Bioinformatics, Gebze, TR 41470, Turkey
| | - Saliha Durmuş
- Department of Bioengineering, Gebze Technical University, Gebze, TR 41400, Turkey.
- PhiTech Bioinformatics, Gebze, TR 41470, Turkey
| |
Collapse
|
8
|
Jin H, Xia P, Deng Z, Hou T, Li J, Li B. Effects of Konjac Glucomannan on Weight Management and Liver Health: Insights from Liver Lipidomics in Obese and Nonobese Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:7906-7918. [PMID: 38530902 DOI: 10.1021/acs.jafc.3c09540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Konjac glucomannan (KGM) is a water-soluble dietary fiber and is used for weight management. However, there is a lack of research on KGM for weight management in nonobese groups and the effects of high-dose KGM supplementation on liver function. This study investigated the metabolic responses to KGM intervention in obese and nonobese mice and explored the underlying mechanisms based on lipidomics. The findings demonstrated that KGM supplementation decreased body weight and mitigated lipid metabolism disorders at the mRNA and protein levels in obese mice. In contrast, no significant impact on these parameters was observed in nonobese mice. Interestingly, KGM had a more significant impact on remodeling hepatic lipid composition in obese mice compared to nonobese mice, leading to reducing harmful lipids and increasing beneficial lipids. However, high-dose KGM increased the risk of hepatocyte bile acid toxicity in obese mice and did not promote liver antioxidant status in nonobese mice. In summary, this study identified distinct metabolic responses to KGM intervention between obese and nonobese mice, providing insights for weight management using KGM.
Collapse
Affiliation(s)
- Hong Jin
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Pengkui Xia
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhichang Deng
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Tao Hou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Jing Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Key Laboratory of Environment Correlative Dietology, Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
9
|
Pausova Z, Sliz E. Large-Scale Population-Based Studies of Blood Metabolome and Brain Health. Curr Top Behav Neurosci 2024. [PMID: 38509405 DOI: 10.1007/7854_2024_463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Metabolomics technologies enable the quantification of multiple metabolomic measures simultaneously, which provides novel insights into molecular aspects of human health and disease. In large-scale, population-based studies, blood is often the preferred biospecimen. Circulating metabolome may relate to brain health either by affecting or reflecting brain metabolism. Peripheral metabolites may act at or cross the blood-brain barrier and, subsequently, influence brain metabolism, or they may reflect brain metabolism if similar pathways are engaged. Peripheral metabolites may also include those penetrating the circulation from the brain, indicating, for example, brain damage. Most brain health-related metabolomics studies have been conducted in the context of neurodegenerative disorders and cognition, but some studies have also focused on neuroimaging markers of these disorders. Moreover, several metabolomics studies of neurodevelopmental disorders have been performed. Here, we provide a brief background on the types of blood metabolites commonly assessed, and we review the literature describing the relationships between human blood metabolome (n > 50 metabolites) and brain health reported in large-scale studies (n > 500 individuals).
Collapse
Affiliation(s)
- Zdenka Pausova
- The Hospital for Sick Children, Toronto, ON, Canada
- Departments of Physiology and Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - Eeva Sliz
- Research Unit of Population Health, Faculty of Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
10
|
Correa Lopes B, Chen CC, Sung CH, Ishii PE, Medina LFDC, Gaschen FP, Suchodolski JS, Pilla R. Correlation between Peptacetobacter hiranonis, the baiCD Gene, and Secondary Bile Acids in Dogs. Animals (Basel) 2024; 14:216. [PMID: 38254385 PMCID: PMC10812727 DOI: 10.3390/ani14020216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/15/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Bile acid metabolism is a key pathway modulated by intestinal microbiota. Peptacetobacter (Clostridium) hiranonis has been described as the main species responsible for the conversion of primary into secondary fecal unconjugated bile acids (fUBA) in dogs. This multi-step biochemical pathway is encoded by the bile acid-inducible (bai) operon. We aimed to assess the correlation between P. hiranonis abundance, the abundance of one specific gene of the bai operon (baiCD), and secondary fUBA concentrations. In this retrospective study, 133 fecal samples were analyzed from 24 dogs. The abundances of P. hiranonis and baiCD were determined using qPCR. The concentration of fUBA was measured by gas chromatography-mass spectrometry. The baiCD abundance exhibited a strong positive correlation with secondary fUBA (ρ = 0.7377, 95% CI (0.6461, 0.8084), p < 0.0001). Similarly, there was a strong correlation between P. hiranonis and secondary fUBA (ρ = 0.6658, 95% CI (0.5555, 0.7532), p < 0.0001). Animals displaying conversion of fUBA and lacking P. hiranonis were not observed. These results suggest P. hiranonis is the main converter of primary to secondary bile acids in dogs.
Collapse
Affiliation(s)
- Bruna Correa Lopes
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77840, USA; (C.-C.C.); (C.-H.S.); (P.E.I.); (L.F.d.C.M.); (J.S.S.); (R.P.)
| | - Chih-Chun Chen
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77840, USA; (C.-C.C.); (C.-H.S.); (P.E.I.); (L.F.d.C.M.); (J.S.S.); (R.P.)
| | - Chi-Hsuan Sung
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77840, USA; (C.-C.C.); (C.-H.S.); (P.E.I.); (L.F.d.C.M.); (J.S.S.); (R.P.)
| | - Patricia Eri Ishii
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77840, USA; (C.-C.C.); (C.-H.S.); (P.E.I.); (L.F.d.C.M.); (J.S.S.); (R.P.)
| | - Luis Fernando da Costa Medina
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77840, USA; (C.-C.C.); (C.-H.S.); (P.E.I.); (L.F.d.C.M.); (J.S.S.); (R.P.)
| | - Frederic P. Gaschen
- Department of Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA;
| | - Jan S. Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77840, USA; (C.-C.C.); (C.-H.S.); (P.E.I.); (L.F.d.C.M.); (J.S.S.); (R.P.)
| | - Rachel Pilla
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77840, USA; (C.-C.C.); (C.-H.S.); (P.E.I.); (L.F.d.C.M.); (J.S.S.); (R.P.)
| |
Collapse
|
11
|
Lyu Y, Xu J, Verdoodt F, Vanhaecke L, Hemeryck LY, Hesta M. Faecal metabolome responses to an altered dietary protein:carbohydrate ratio in adult dogs. Vet Q 2023; 43:1-10. [PMID: 37869782 PMCID: PMC10614716 DOI: 10.1080/01652176.2023.2273891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023] Open
Abstract
High-protein diets may aid weight loss and weight maintenance programs in both humans and dogs, although the effect of dietary protein levels on gut metabolism and functionality has not been studied in depth. The current study aimed to investigate the effect of an altered dietary protein:carbohydrate ratio on gut function in adult dogs by means of faecal metabolomic fingerprinting. More specifically, functional metabolic differences in dogs fed a high-protein/low-carbohydrate (HPLC) vs. low-protein/high-carbohydrate (LPHC) diet were studied by equally allocating twelve clinically healthy (6 lean and 6 obese) Beagles into two groups in a cross-over design, with each group receiving two isocaloric diets for four weeks. The faecal metabolome revealed that different protein:carbohydrate ratio can influence host and/or gut microbiome metabolism and function, while no effect was observed on the body condition. Targeted analysis demonstrated that the HPLC diet significantly increased the concentration of indole, spermidine, and pipecolinic acid and decreased the concentration of azelaic acid, D-fructose, mannose, and galactose (p < 0.05). Multivariate modelling (OPLS-DA) of the untargeted faecal metabolome revealed distinctly different metabolomic profiles following the HPLC vs. LPHC diet, with 18 altered pathways. The HPLC diet influenced amino acid and lipid metabolism, potentially promoting weight loss and immune function, whereas the LPHC diet affected carbohydrate fermentation and may promote anti-oxidative function.
Collapse
Affiliation(s)
- Yang Lyu
- ECAN Equine and Companion Animal Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Jia Xu
- ECAN Equine and Companion Animal Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Fien Verdoodt
- ECAN Equine and Companion Animal Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Lynn Vanhaecke
- Laboratory of Integrative Metabolomics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Lieselot Y. Hemeryck
- Laboratory of Integrative Metabolomics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Myriam Hesta
- ECAN Equine and Companion Animal Nutrition, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
12
|
Zhang D, Wang H, Liu A, Wang S, Xu C, Lan K, Xiang W, Zhu K, Xiao Y, Fu J, Jiang R, Chen W, Ni Y. The chronic consumption of dietary fructose promotes the gut Clostridium species imbalance and bile acid alterations in developing nonalcoholic fatty liver disease. J Nutr Biochem 2023; 121:109434. [PMID: 37661068 DOI: 10.1016/j.jnutbio.2023.109434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 06/30/2023] [Accepted: 08/29/2023] [Indexed: 09/05/2023]
Abstract
Excessive fructose intake is associated with the rising prevalence of nonalcoholic fatty liver disease (NAFLD). The gut microbiome (GM) and bile acids (BAs) are involved in the pathogenesis of NAFLD, but the impact of fructose on their cross-talk is unclear. In this study, adult male C57BL/6J mice were fed a normal diet with tap water (ND) or with 4% fructose in the drinking water (Fru), 60% high-fat diet with tap water (HF) or with 4% fructose solution (HFF) for 12 weeks. Targeted BA analysis was performed in five anatomical sites including the liver, ileum contents, portal serum, cecum contents, and feces. Metagenomic sequencing was performed to explore gut dysbiosis. Within 12 weeks, the 4% fructose diet could initially stimulate gut dysbiosis and BA upregulation in the ileum, portal serum, and cecum when the intestinal and hepatic transport system remained stable without hepatic lipid accumulation. However, the chronic consumption of fructose promoted HF-induced NAFLD, with significantly increased body weight, impaired glucose tolerance, and advanced liver steatosis. BA transporters were inhibited in HFF, causing the block of internal BA circulation and increased BA secretion via cecum contents and feces. Notably, lithocholic acid (LCA) and its taurine conjugates were elevated within the enterohepatic circulation. Meanwhile, the Clostridium species were significantly altered in both Fru and HFF groups and were closely associated with fructose and BA metabolism. In summary, excessive fructose caused gut dysbiosis and BA alterations, promoting HF-induced NAFLD. The crosstalk between Clostridium sp. and LCA species were potential targets in fructose-mediated NAFLD.
Collapse
Affiliation(s)
- Danni Zhang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China; Department of Epidemiology and Biostatistics, Zhejiang University School of Public Health, Hangzhou, China
| | - Huiying Wang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ana Liu
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Shan Wang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Cuifang Xu
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Ke Lan
- Key Laboratory of Drug Targeting and Drug Delivery System, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Wenqing Xiang
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Kun Zhu
- Department of Pathology, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Junfen Fu
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Runqiu Jiang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China; Medical School of Nanjing University, Nanjing, China
| | - Wenlian Chen
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yan Ni
- Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China; Department of Epidemiology and Biostatistics, Zhejiang University School of Public Health, Hangzhou, China.
| |
Collapse
|
13
|
Gao M, Li J, Zhang R, Li N, Li W, Zhang S, Wang P, Wang H, Fang Z, Yu Z, Hu G, Leng J, Yang X. Serum mannan-binding lectin-associated serine proteases in early pregnancy for gestational diabetes in Chinese pregnant women. Front Endocrinol (Lausanne) 2023; 14:1230244. [PMID: 37941903 PMCID: PMC10628726 DOI: 10.3389/fendo.2023.1230244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 09/27/2023] [Indexed: 11/10/2023] Open
Abstract
Aims This study aimed to explore associations of mannan-binding lectin-associated serine protease (MASP) levels in early pregnancy with gestational diabetes mellitus (GDM). We also examined interactions of MASPs and deoxycholic acid (DCA)/glycoursodeoxycholic acid (GUDCA) for the GDM risk and whether the interactive effects if any on the GDM risk were mediated via lysophosphatidylcholine (LPC) 18:0. Materials and methods A 1:1 case-control study (n = 414) nested in a prospective cohort of pregnant women was conducted in Tianjin, China. Binary conditional logistic regressions were performed to examine associations of MASPs with the GDM risk. Additive interaction measures were used to examine interactions between MASPs and DCA/GUDCA for the GDM risk. Mediation analyses and Sobel tests were used to examine mediation effects of LPC18:0 between the copresence of MASPs and DCA/GUDCA on the GDM risk. Results High MASP-2 was independently associated with GDM [odds ratio (OR): 2.62, 95% confidence interval (CI): 1.44-4.77], while the effect of high MASP-1 on GDM was attributable to high MASP-2 (P for Sobel test: 0.003). Low DCA markedly increased the OR of high MASP-2 alone from 2.53 (1.10-5.85) up to 10.6 (4.22-26.4), with a significant additive interaction. In addition, high LPC18:0 played a significant mediating role in the links from low DCA to GDM and from the copresence of high MASP-2 and low DCA to GDM (P for Sobel test <0.001) but not in the link from high MASP-2 to GDM. Conclusions High MASP-1 and MASP-2 in early pregnancy were associated with GDM in Chinese pregnant women. MASP-2 amplifies the risk of low DCA for GDM, which is mediated via LPC18:0.
Collapse
Affiliation(s)
- Ming Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University School of Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin Medical University School of Public Health, Tianjin, China
| | - Rui Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Ninghua Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Weiqin Li
- Project Office, Tianjin Women and Children’s Health Center, Tianjin, China
| | - Shuang Zhang
- Project Office, Tianjin Women and Children’s Health Center, Tianjin, China
| | - Peng Wang
- Project Office, Tianjin Women and Children’s Health Center, Tianjin, China
| | - Hui Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Zhongze Fang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Zhijie Yu
- Population Cancer Research Program and Department of Pediatrics, Dalhousie University, Halifax, NS, Canada
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Junhong Leng
- Project Office, Tianjin Women and Children’s Health Center, Tianjin, China
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University School of Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin Medical University School of Public Health, Tianjin, China
| |
Collapse
|
14
|
Chu Z, Hu Z, Luo Y, Zhou Y, Yang F, Luo F. Targeting gut-liver axis by dietary lignans ameliorate obesity: evidences and mechanisms. Crit Rev Food Sci Nutr 2023:1-22. [PMID: 37870876 DOI: 10.1080/10408398.2023.2272269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2023]
Abstract
An imbalance between energy consumption and energy expenditure causes obesity. It is characterized by increased adipose accumulation and accompanied by chronic low-grade inflammation. Many studies have suggested that the gut microbiota of the host mediates the relationship between high-fat diet consumption and the development of obesity. Diet and nutrition of the body are heavily influenced by gut microbiota. The alterations in the microbiota in the gut may have effects on the homeostasis of the host's energy levels, systemic inflammation, lipid metabolism, and insulin sensitivity. The liver is an important organ for fat metabolism and gut-liver axis play important role in the fat metabolism. Gut-liver axis is a bidirectional relationship between the gut and its microbiota and the liver. As essential plant components, lignans have been shown to have different biological functions. Accumulating evidences have suggested that lignans may have lipid-lowering properties. Lignans can regulate the level of the gut microbiota and their metabolites in the host, thereby affecting signaling pathways related to fat synthesis and metabolism. These signaling pathways can make a difference in inhibiting fat accumulation, accelerating energy metabolism, affecting appetite, and inhibiting chronic inflammation. It will provide the groundwork for future studies on the lipid-lowering impact of lignans and the creation of functional meals based on those findings.
Collapse
Affiliation(s)
- Zhongxing Chu
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| | - Zuomin Hu
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| | - Yi Luo
- Department of Clinic Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, P.R. China
| | - Yaping Zhou
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| | - Feiyan Yang
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| | - Feijun Luo
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, College of Food Science and Engineering, Central South University of Forestry and Technology, Changsha, Hunan, P.R. China
| |
Collapse
|
15
|
Zhao Z, Yang LL, Wang QL, Du JF, Zheng ZG, Jiang Y, Li P, Li HJ. Baohuoside I inhibits FXR signaling pathway to interfere with bile acid homeostasis via targeting ER α degradation. Cell Biol Toxicol 2023; 39:1215-1235. [PMID: 35802278 DOI: 10.1007/s10565-022-09737-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022]
Abstract
Epimedii folium (EF) is an effective herbal medicine in osteoporosis treatment, but the clinical utilization of EF has been limited due to potential hepatotoxicity. The previous studies identified that baohuoside I (BI), the main active component of EF, was relevant to EF-induced liver injury. However, the mechanisms of BI causing direct injury to hepatocytes remain unclear. Here, we reveal that BI inhibits FXR-mediated signaling pathway via targeting estrogen receptor α (ER α), leading to the accumulation of bile acids (BAs). Targeted bile acid analyses show BI alters the BA composition and distribution, resulting in impaired BA homeostasis. Mechanistically, BI induces FXR-dependent hepatotoxicity at transcriptional level. Additionally, ER α is predicted to bind to the FXR promoter region based on transcription factor binding sites databases and we further demonstrate that ER α positively regulates FXR promoter activity and affects the expression of target genes involved in BA metabolism. Importantly, we discover that ER α and its mediated FXR transcription regulation might be involved in BI-induced liver injury via ligand-dependent ER α degradation. Collectively, our findings indicate that FXR is a newly discovered target gene of ER α mediated BI-induced liver injury, and suggest BI may be responsible for EF-induced liver injury.
Collapse
Affiliation(s)
- Zhen Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Lu-Lu Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Qiao-Lei Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Jin-Fa Du
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Zu-Guo Zheng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Yan Jiang
- College of Chemical Engineering, Nanjing Forestry University, 159 Longpan Road, Nanjing, 210037, China.
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China.
| |
Collapse
|
16
|
Li N, Li J, Wang H, Liu J, Li W, Yang K, Huo X, Leng J, Yu Z, Hu G, Fang Z, Yang X. Aromatic Amino Acids and Their Interactions with Gut Microbiota-Related Metabolites for Risk of Gestational Diabetes: A Prospective Nested Case-Control Study in a Chinese Cohort. ANNALS OF NUTRITION & METABOLISM 2023; 79:291-300. [PMID: 37339616 DOI: 10.1159/000531481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 06/05/2023] [Indexed: 06/22/2023]
Abstract
INTRODUCTION The aim of this study was to explore associations of aromatic amino acids (AAA) in early pregnancy with gestational diabetes mellitus (GDM), and whether high AAA and gut microbiota-related metabolites had interactive effects on GDM risk. METHODS We conducted a 1:1 case-control study (n = 486) nested in a prospective cohort of pregnant women from 2010 to 2012. According to the International Association of Diabetes and Pregnancy Study Group's criteria, 243 women were diagnosed with GDM. Binary conditional logistic regression was performed to examine associations of AAA with GDM risk. Interactions between AAA and gut microbiota-related metabolites for GDM were examined using additive interaction measures. RESULTS High phenylalanine and tryptophan were associated with increased GDM risk (OR: 1.72, 95% CI: 1.07-2.78 and 1.66, 1.02-2.71). The presence of high trimethylamine (TMA) markedly increased the OR of high phenylalanine alone up to 7.95 (2.79-22.71), while the presence of low glycoursodeoxycholic acid (GUDCA) markedly increased the OR of high tryptophan alone up to 22.88 (5.28-99.26), both with significant additive interactions. Furthermore, high lysophosphatidylcholines (LPC18:0) mediated both interactive effects. CONCLUSIONS High phenylalanine may have an additive interaction with high TMA, while high tryptophan may have an additive interaction with low GUDCA toward increased risk of GDM, both being mediated via LPC18:0.
Collapse
Affiliation(s)
- Ninghua Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Hui Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jinnan Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Weiqin Li
- Project Office, Tianjin Women and Children's Health Center, Tianjin, China
| | - Kai Yang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiaoxu Huo
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| | - Junhong Leng
- Project Office, Tianjin Women and Children's Health Center, Tianjin, China
| | - Zhijie Yu
- Population Cancer Research Program and Department of Pediatrics, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| | - Zhongze Fang
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin, China
| |
Collapse
|
17
|
Fan Y, Ying J, Ma H, Cui H. Microbiota-related metabolites fueling the understanding of ischemic heart disease. IMETA 2023; 2:e94. [PMID: 38868424 PMCID: PMC10989774 DOI: 10.1002/imt2.94] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 01/04/2023] [Accepted: 01/21/2023] [Indexed: 06/14/2024]
Abstract
Up-to-date knowledge of gut microbial taxa associated with ischemic heart disease (IHD). Microbial metabolites for mechanistic dissection of IHD pathology. Microbiome-based therapies in IHD prevention and treatment.
Collapse
Affiliation(s)
- Yong Fan
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Jiajun Ying
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
- Department of Cardiology, Ningbo First HospitalNingbo UniversityNingboChina
| | - Hongchuang Ma
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
| | - Hanbin Cui
- Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang ProvinceNingboChina
- Department of Cardiology, Ningbo First HospitalNingbo UniversityNingboChina
- Ningbo Clinical Research Center for Cardiovascular DiseaseNingboChina
| |
Collapse
|
18
|
Aoi W, Inoue R, Mizushima K, Honda A, Björnholm M, Takagi T, Naito Y. Exercise-acclimated microbiota improves skeletal muscle metabolism via circulating bile acid deconjugation. iScience 2023; 26:106251. [PMID: 36915683 PMCID: PMC10005909 DOI: 10.1016/j.isci.2023.106251] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 10/31/2022] [Accepted: 02/16/2023] [Indexed: 02/23/2023] Open
Abstract
Habitual exercise alters the intestinal microbiota composition, which may mediate its systemic benefits. We examined whether transplanting fecal microbiota from trained mice improved skeletal muscle metabolism in high-fat diet (HFD)-fed mice. Fecal samples from sedentary and exercise-trained mice were gavage-fed to germ-free mice. After receiving fecal samples from trained donor mice for 1 week, recipient mice had elevated levels of AMP-activated protein kinase (AMPK) and insulin growth factor-1 in skeletal muscle. In plasma, bile acid (BA) deconjugation was found to be promoted in recipients transplanted with feces from trained donor mice; free-form BAs also induced more AMPK signaling and glucose uptake than tauro-conjugated BAs. The transplantation of exercise-acclimated fecal microbiota improved glucose tolerance after 8 weeks of HFD administration. Intestinal microbiota may mediate exercise-induced metabolic improvements in mice by modifying circulating BAs. Our findings provide insights into the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Wataru Aoi
- Division of Applied Life Sciences, Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 6068522, Japan
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, Osaka 5730101, Japan
| | - Katsura Mizushima
- Department of Human Immunology and Nutrition Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 6028566, Japan
| | - Akira Honda
- Gastroenterology, Tokyo Medical University Ibaraki Medical Center, Ibaraki 3000395, Japan
| | - Marie Björnholm
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 17176, Sweden
| | - Tomohisa Takagi
- Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 6028566, Japan.,Department for Medical Innovation and Translational Medical Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 6028566, Japan
| | - Yuji Naito
- Department of Human Immunology and Nutrition Science, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto 6028566, Japan
| |
Collapse
|
19
|
Zhou C, Peng K, Liu Y, Zhang R, Zheng X, Yue B, Du C, Wu Y. Comparative Analyses Reveal the Genetic Mechanism of Ambergris Production in the Sperm Whale Based on the Chromosome-Level Genome. Animals (Basel) 2023; 13:ani13030361. [PMID: 36766250 PMCID: PMC9913093 DOI: 10.3390/ani13030361] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/05/2023] [Accepted: 01/14/2023] [Indexed: 01/24/2023] Open
Abstract
Sperm whales are a marine mammal famous for the aromatic substance, the ambergris, produced from its colon. Little is known about the biological processes of ambergris production, and this study aims to investigate the genetic mechanism of ambergris production in the sperm whale based on its chromosome-level genome. Comparative genomics analyses found 1207 expanded gene families and 321 positive selected genes (PSGs) in the sperm whale, and functional enrichment analyses suggested revelatory pathways and terms related to the metabolism of steroids, terpenoids, and aldosterone, as well as microbiota interaction and immune network in the intestine. Furthermore, two sperm-whale-specific missense mutations (Tyr393His and Leu567Val) were detected in the PSG LIPE, which has been reported to play vital roles in lipid and cholesterol metabolism. In total, 46 CYP genes and 22 HSD genes were annotated, and then mapped to sperm whale chromosomes. Furthermore, phylogenetic analysis of CYP genes in six mammals found that CYP2E1, CYP51A and CYP8 subfamilies exhibited relative expansion in the sperm whale. Our results could help understand the genetic mechanism of ambergris production, and further reveal the convergent evolution pattern among animals that produce similar odorants.
Collapse
Affiliation(s)
- Chuang Zhou
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Kexin Peng
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Yi Liu
- Key Laboratory of Sichuan Province for Fishes Conservation and Utilization in the Upper Reaches of the Yangtze River, Neijiang Normal University, Neijiang 641000, China
| | - Rusong Zhang
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Xiaofeng Zheng
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Bisong Yue
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610064, China
| | - Chao Du
- Baotou Teachers College, Baotou 014060, China
- Correspondence: (C.D.); (Y.W.)
| | - Yongjie Wu
- Key Laboratory of Bioresources and Ecoenvironment (Ministry of Education), College of Life Sciences, Sichuan University, Chengdu 610064, China
- Correspondence: (C.D.); (Y.W.)
| |
Collapse
|
20
|
Gillard J, Leclercq IA. Biological tuners to reshape the bile acid pool for therapeutic purposes in non-alcoholic fatty liver disease. Clin Sci (Lond) 2023; 137:65-85. [PMID: 36601783 PMCID: PMC9816373 DOI: 10.1042/cs20220697] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/08/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023]
Abstract
Bile acids synthesized within the hepatocytes are transformed by gut microorganisms and reabsorbed into the portal circulation. During their enterohepatic cycling, bile acids act as signaling molecules by interacting with receptors to regulate pathways involved in many physiological processes. The bile acid pool, composed of a variety of bile acid species, has been shown to be altered in diseases, hence contributing to disease pathogenesis. Thus, understanding the changes in bile acid pool size and composition in pathological processes will help to elaborate effective pharmacological treatments. Five crucial steps along the enterohepatic cycle shape the bile acid pool size and composition, offering five possible targets for therapeutic intervention. In this review, we provide an insight on the strategies to modulate the bile acid pool, and then we discuss the potential benefits in non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Justine Gillard
- Laboratory of Hepato‐Gastroenterology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Isabelle A. Leclercq
- Laboratory of Hepato‐Gastroenterology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
21
|
MacCannell AD, Roberts LD. Metabokines in the regulation of systemic energy metabolism. Curr Opin Pharmacol 2022; 67:102286. [PMID: 36137304 DOI: 10.1016/j.coph.2022.102286] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 07/05/2022] [Accepted: 08/01/2022] [Indexed: 01/25/2023]
Abstract
Metabolism consists of life-sustaining chemical reactions involving metabolites. Historically, metabolites were defined as the intermediates or end products of metabolism and considered to be passive participants changed by metabolic processes. However, recent research has redefined how we view metabolism. There is emerging evidence of metabolites which function to mediate cellular signalling and interorgan crosstalk, regulating local metabolism and systemic physiology. These bioactive metabolite signals have been termed metabokines. Metabokines regulate diverse energy metabolism pathways across multiple tissues, including fatty acid β-oxidation, mitochondrial oxidative phosphorylation, lipolysis, glycolysis and gluconeogenesis. There is increasing impetus to uncover novel metabokine signalling axes to better understand how these may be perturbed in metabolic diseases and determine their utility as therapeutic targets.
Collapse
Affiliation(s)
- Amanda Dv MacCannell
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Lee D Roberts
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK.
| |
Collapse
|
22
|
Moutinho TJ, Powers DA, Hanson GF, Levy S, Baveja R, Hefner I, Mohamed M, Abdelghani A, Baker RL, Papin JA, Moore SR, Hourigan SK. Fecal sphingolipids predict parenteral nutrition-associated cholestasis in the neonatal intensive care unit. JPEN J Parenter Enteral Nutr 2022; 46:1903-1913. [PMID: 35285019 PMCID: PMC9468188 DOI: 10.1002/jpen.2374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/23/2022] [Accepted: 03/06/2022] [Indexed: 01/26/2023]
Abstract
BACKGROUND Parenteral nutrition-associated cholestasis (PNAC) in the neonatal intensive care unit (NICU) causes significant morbidity and associated healthcare costs. Laboratory detection of PNAC currently relies on elevated serum conjugated bilirubin levels in the aftermath of impaired bile flow. Here, we sought to identify fecal biomarkers, which when integrated with clinical data, would better predict risk for developing PNAC. METHODS Using untargeted metabolomics in 200 serial stool samples from 60 infants, we applied statistical and machine learning approaches to identify clinical features and metabolic biomarkers with the greatest associative potential for risk of developing PNAC. Stools were collected prospectively from infants receiving PN with soybean oil-based lipid emulsion at a level IV NICU. RESULTS Low birth weight, extreme prematurity, longer duration of PN, and greater number of antibiotic courses were all risk factors for PNAC (P < 0.05). We identified 78 stool biomarkers with early predictive potential (P < 0.05). From these 78 biomarkers, we further identified 12 sphingomyelin lipids with high association for the development of PNAC in precholestasis stool samples when combined with birth anthropometry. CONCLUSION We demonstrate the potential for stool metabolomics to enhance early identification of PNAC risk. Earlier detection of high-risk infants would empower proactive mitigation with alterations to PN for at-risk infants and optimization of energy nutrition with PN for infants at lower risk.
Collapse
Affiliation(s)
- Thomas J. Moutinho
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Deborah A. Powers
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Gabriel F. Hanson
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Shira Levy
- Inova Children's HospitalFalls ChurchVirginiaUSA
| | - Rajiv Baveja
- Fairfax Neonatal AssociatesFalls ChurchVirginiaUSA
| | | | | | | | | | - Jason A. Papin
- Department of Biomedical EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Sean R. Moore
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of PediatricsUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Suchitra K. Hourigan
- Inova Children's HospitalFalls ChurchVirginiaUSA,Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of PediatricsUniversity of VirginiaCharlottesvilleVirginiaUSA,Division of Pediatric GastroenterologyPediatric Specialists of VirginiaFairfaxVirginiaUSA,Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious DiseasesNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
23
|
Wang Y, Ai Z, Xing X, Fan Y, Zhang Y, Nan B, Li X, Wang Y, Liu J. The ameliorative effect of probiotics on diet-induced lipid metabolism disorders: A review. Crit Rev Food Sci Nutr 2022; 64:3556-3572. [PMID: 36218373 DOI: 10.1080/10408398.2022.2132377] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
High-fat diet induces lipid metabolism disorders that has become one of the grievous public health problems and imposes a serious economic and social burden worldwide. Safety probiotics isolated from nature are regarded as a novel supplementary strategy for preventing and improving diet-induced lipid metabolism disorders and related chronic diseases. The present review summarized the latest researches of probiotics in high fat diet induced lipid metabolism disorders to provide a critical perspective on the regulatory function of probiotics for future research. Furthermore, the screening criteria and general sources of probiotics with lipid-lowering ability also outlined to enlarge microbial species resource bank instantly, which promoted the development of functional foods with lipid-lowering strains from nature. After critically reviewing the lipid-lowering potential of probiotics both in vitro and in vivo and even in clinical data of humans, we provided a perspective that probiotics activated AMPK signaling pathway to regulate fat synthesis and decomposition, as well as affected positively the gut microbiota structure, intestinal barrier function and systemic inflammatory response, then these beneficial effects are amplified along Gut-liver axis, which regulated intestinal flora metabolites such as SCFAs and BAs by HMGCR/FXR/SHP signaling pathway to improve high fat diet induced lipid metabolism disorders effectively.
Collapse
Affiliation(s)
- Yu Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Zhiyi Ai
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Xinyue Xing
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Yuling Fan
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Yue Zhang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Bo Nan
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Xia Li
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
| | - Yuhua Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- Jilin Province Innovation Center for Food Biological Manufacture, Jilin Agricultural University, Changchun, China
- National Processing Laboratory for Soybean Industry and Technology, Changchun, China
- National Engineering Research Center for Wheat and Cord Deep Processing, Changchun, China
| | - Jingsheng Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
- National Engineering Research Center for Wheat and Cord Deep Processing, Changchun, China
| |
Collapse
|
24
|
Jiang J, Ma Y, Liu Y, Lu D, Gao X, Krausz KW, Desai D, Amin SG, Patterson AD, Gonzalez FJ, Xie C. Glycine-β-muricholic acid antagonizes the intestinal farnesoid X receptor-ceramide axis and ameliorates NASH in mice. Hepatol Commun 2022; 6:3363-3378. [PMID: 36196594 PMCID: PMC9701488 DOI: 10.1002/hep4.2099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/10/2022] [Indexed: 01/21/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is a rapidly developing pathology around the world, with limited treatment options available. Some farnesoid X receptor (FXR) agonists have been applied in clinical trials for NASH, but side effects such as pruritus and low-density lipoprotein elevation have been reported. Intestinal FXR is recognized as a promising therapeutic target for metabolic diseases. Glycine-β-muricholic acid (Gly-MCA) is an intestine-specific FXR antagonist previously shown to have favorable metabolic effects on obesity and insulin resistance. Herein, we identify a role for Gly-MCA in the pathogenesis of NASH, and explore the underlying molecular mechanism. Gly-MCA improved lipid accumulation, inflammatory response, and collagen deposition in two different NASH models. Mechanistically, Gly-MCA decreased intestine-derived ceramides by suppressing ceramide synthesis-related genes via decreasing intestinal FXR signaling, leading to lower liver endoplasmic reticulum (ER) stress and proinflammatory cytokine production. The role of bile acid metabolism and adiposity was excluded in the suppression of NASH by Gly-MCA, and a correlation was found between intestine-derived ceramides and NASH severity. This study revealed that Gly-MCA, an intestine-specific FXR antagonist, has beneficial effects on NASH by reducing ceramide levels circulating to liver via lowering intestinal FXR signaling, and ceramide production, followed by decreased liver ER stress and NASH progression. Intestinal FXR is a promising drug target and Gly-MCA a novel agent for the prevention and treatment of NASH.
Collapse
Affiliation(s)
- Jie Jiang
- School of Chinese Materia MedicaNanjing University of Chinese MedicineNanjingChina,State Key Laboratory of Drug ResearchShanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
| | - Yuandi Ma
- State Key Laboratory of Drug ResearchShanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina,University of Chinese Academy of SciencesBeijingChina
| | - Yameng Liu
- State Key Laboratory of Drug ResearchShanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina
| | - Dasheng Lu
- Department of Pharmacology, College of MedicineThe Pennsylvania State UniversityHersheyPennsylvaniaUSA
| | - Xiaoxia Gao
- Department of Pharmacology, College of MedicineThe Pennsylvania State UniversityHersheyPennsylvaniaUSA
| | - Kristopher W. Krausz
- Department of Pharmacology, College of MedicineThe Pennsylvania State UniversityHersheyPennsylvaniaUSA
| | - Dhimant Desai
- Laboratory of Metabolism, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Shantu G. Amin
- Laboratory of Metabolism, Center for Cancer ResearchNational Cancer Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and CarcinogenesisThe Pennsylvania State UniversityUniversity ParkPennsylvaniaUSA
| | - Frank J. Gonzalez
- Department of Pharmacology, College of MedicineThe Pennsylvania State UniversityHersheyPennsylvaniaUSA
| | - Cen Xie
- School of Chinese Materia MedicaNanjing University of Chinese MedicineNanjingChina,State Key Laboratory of Drug ResearchShanghai Institute of Materia Medica, Chinese Academy of SciencesShanghaiChina,University of Chinese Academy of SciencesBeijingChina,Department of Pharmacology, College of MedicineThe Pennsylvania State UniversityHersheyPennsylvaniaUSA
| |
Collapse
|
25
|
An C, Chon H, Ku W, Eom S, Seok M, Kim S, Lee J, Kim D, Lee S, Koo H, Cho H, Han S, Moon J, Kang M, Ryu K. Bile Acids: Major Regulator of the Gut Microbiome. Microorganisms 2022; 10:microorganisms10091792. [PMID: 36144395 PMCID: PMC9502002 DOI: 10.3390/microorganisms10091792] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/16/2022] Open
Abstract
Bile acids are synthesized from cholesterol and play an important role in regulating intestinal microflora. The different degrees of hydrophobicity and acidity of individual bile acids may affect their antimicrobial properties. We examined the antimicrobial effects of different bile acids on various microorganisms in vitro and confirmed whether these remain consistent in vivo. Using human bile acids, including ursodeoxycholic acid, cholic acid, chenodeoxycholic acid, deoxycholic acid, and lithocholic acid, a disc diffusion test was performed, and a rodent model was created to determine the antimicrobial effects of each bile acid. The fecal bacterial population was analyzed using a real-time polymerase chain reaction. Each bile acid showed different microbial inhibitory properties. The inhibitory activity of bile acids against microbiota which normally resides in the gastrointestinal tract and biliary system, was low; however, normal flora of other organs was significantly inhibited. Changes in microbial counts after bile acid administration in a rodent model differed in the colon and cecum. The in vivo and in vitro results show that the antimicrobial effects of bile acids against intestinal microbiota were similar. In conclusion, bile acids could be a novel treatment strategy to regulate gut microbiota.
Collapse
Affiliation(s)
- Chihyeok An
- Department of Gastroenterology, Konyang University Myunggok Medical Research Institute, Daejeon 35365, Korea
| | - Hyeyeon Chon
- Department of Gastroenterology, Konyang University Myunggok Medical Research Institute, Daejeon 35365, Korea
| | - Wanrim Ku
- Department of Gastroenterology, Konyang University Myunggok Medical Research Institute, Daejeon 35365, Korea
| | - Sunho Eom
- Department of Gastroenterology, Konyang University Myunggok Medical Research Institute, Daejeon 35365, Korea
| | - Mingyu Seok
- Department of Gastroenterology, Konyang University Myunggok Medical Research Institute, Daejeon 35365, Korea
| | - Sangha Kim
- Department of Laboratory Medicine, Konyang University College of Medicine, Daejeon 35365, Korea
| | - Jaesun Lee
- Department of Gastroenterology, Konyang University Myunggok Medical Research Institute, Daejeon 35365, Korea
| | - Daesung Kim
- Department of Gastroenterology, Konyang University Myunggok Medical Research Institute, Daejeon 35365, Korea
| | - Sanghyuk Lee
- Department of Gastroenterology, Konyang University Myunggok Medical Research Institute, Daejeon 35365, Korea
| | - Hoonsup Koo
- Department of Gastroenterology, Konyang University Myunggok Medical Research Institute, Daejeon 35365, Korea
| | - Hyunjung Cho
- Department of Laboratory Medicine, Konyang University College of Medicine, Daejeon 35365, Korea
| | - Seungyun Han
- Department of Anatomy, Konyang University College of Medicine, Daejeon 35365, Korea
| | - Juik Moon
- Department of Surgery, Konyang University College of Medicine, Daejeon 35365, Korea
| | - Miil Kang
- Department of Rheumatology and Clinical Immunology, Dankook University Hospital, Cheonan 31116, Korea
- Correspondence: (M.K.); (K.R.); Tel.: +82-10-3440-7428 (M.K.); +82-10-7464-0620 (K.R.); Fax: +82-41-550-7006 (M.K.); +82-42-600-9090 (K.R.)
| | - Kihyun Ryu
- Department of Gastroenterology, Konyang University Myunggok Medical Research Institute, Daejeon 35365, Korea
- Correspondence: (M.K.); (K.R.); Tel.: +82-10-3440-7428 (M.K.); +82-10-7464-0620 (K.R.); Fax: +82-41-550-7006 (M.K.); +82-42-600-9090 (K.R.)
| |
Collapse
|
26
|
Cai J, Rimal B, Jiang C, Chiang JYL, Patterson AD. Bile acid metabolism and signaling, the microbiota, and metabolic disease. Pharmacol Ther 2022; 237:108238. [PMID: 35792223 DOI: 10.1016/j.pharmthera.2022.108238] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/13/2022] [Accepted: 06/27/2022] [Indexed: 11/24/2022]
Abstract
The diversity, composition, and function of the bacterial community inhabiting the human gastrointestinal tract contributes to host health through its role in producing energy or signaling molecules that regulate metabolic and immunologic functions. Bile acids are potent metabolic and immune signaling molecules synthesized from cholesterol in the liver and then transported to the intestine where they can undergo metabolism by gut bacteria. The combination of host- and microbiota-derived enzymatic activities contribute to the composition of the bile acid pool and thus there can be great diversity in bile acid composition that depends in part on the differences in the gut bacteria species. Bile acids can profoundly impact host metabolic and immunological functions by activating different bile acid receptors to regulate signaling pathways that control a broad range of complex symbiotic metabolic networks, including glucose, lipid, steroid and xenobiotic metabolism, and modulation of energy homeostasis. Disruption of bile acid signaling due to perturbation of the gut microbiota or dysregulation of the gut microbiota-host interaction is associated with the pathogenesis and progression of metabolic disorders. The metabolic and immunological roles of bile acids in human health have led to novel therapeutic approaches to manipulate the bile acid pool size, composition, and function by targeting one or multiple components of the microbiota-bile acid-bile acid receptor axis.
Collapse
Affiliation(s)
- Jingwei Cai
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Bipin Rimal
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Changtao Jiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, and the Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, PR China
| | - John Y L Chiang
- Department of Integrative Medical Sciences, College of Medicine, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
27
|
Study on the mechanism of anti-hepatic fibrosis of Glycyrrhiza Uralensis-Salvia miltiorrhiza prescription based on serum and urine metabolomics and network pharmacology. J Chromatogr B Analyt Technol Biomed Life Sci 2022; 1209:123416. [PMID: 36027704 DOI: 10.1016/j.jchromb.2022.123416] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/08/2022] [Accepted: 08/09/2022] [Indexed: 11/22/2022]
Abstract
Hepatic fibrosis (HF) is a kind of chronic epidemic liver disease. Glycyrrhiza Uralensis and Salvia Miltiorrhiza (GUSM), traditional Chinese medicine, has the obvious clinical treatment of liver fibrosis. This study aimed to investigate the mechanisms of GUSM against HF by an integrated strategy combining untargeted metabolomics with network pharmacology. The results showed that GUSM prescription can improve the morphology and structure of liver tissue, inhibit the proliferation of collagen fibers and reducing the inflammatory response of the liver and so on. Endogenous metabolites and HF-related potential biomarkers in serum and urine were detected by ultra-high-performance liquid chromatography coupled with quadrupole-time-of-flight mass spectrometry (UPLC-Q-TOF/MS). The metabolic pathways were enriched by MetaboAnalyst. GUSM prescription showed an antifibrotic effect on rats by regulating metabolic pathways, mainly pentose and glucuronate interconversions and arachidonic acid metabolism. Network pharmacology was then applied to find 42 overlapping targets of GUSM-HF. Quercetin was found to be the main active component and STAT3 was the main active target in GUSM prescription. Molecular docking showed high affinities between quercetin and STAT3. Therefore, GUSM has protective effects on HF by regulating the metabolism and different signaling pathways. The work also shows that the metabolomic and network pharmacology methods are promising tools to gain insight into the efficacy and mechanism research of traditional Chinese medicines.
Collapse
|
28
|
Chew NWS, Ng CH, Truong E, Noureddin M, Kowdley KV. Nonalcoholic Steatohepatitis Drug Development Pipeline: An Update. Semin Liver Dis 2022; 42:379-400. [PMID: 35709720 DOI: 10.1055/a-1877-9656] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a burgeoning global health crisis that mirrors the obesity pandemic. This global health crisis has stimulated active research to develop novel NASH pharmacotherapies targeting dysregulated inflammatory, cellular stress, and fibrogenetic processes that include (1) metabolic pathways to improve insulin sensitivity, de novo lipogenesis, and mitochondrial utilization of fatty acids; (2) cellular injury or inflammatory targets that reduce inflammatory cell recruitment and signaling; (3) liver-gut axis targets that influence bile acid enterohepatic circulation and signaling; and (4) antifibrotic targets. In this review, we summarize several of the therapeutic agents that have been studied in phase 2 and 3 randomized trials. In addition to reviewing novel therapeutic drugs targeting nuclear receptor pathways, liver chemokine receptors, liver lipid metabolism, lipotoxicity or cell death, and glucagon-like peptide-1 receptors, we also discuss the rationale behind the use of combination therapy and the lessons learned from unsuccessful or negative clinical trials.
Collapse
Affiliation(s)
- Nicholas W S Chew
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, Singapore
| | - Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Emily Truong
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| | - Mazen Noureddin
- Division of Digestive and Liver Diseases, Department of Medicine, Cedars-Sinai Fatty Liver Program, Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Kris V Kowdley
- Liver Institute Northwest and Elson S. Floyd College of Medicine, Washington State University, Seattle, Washington
| |
Collapse
|
29
|
Zhang Q, Zhou X, Zhang J, Li Q, Qian Z. Selenium and vitamin B6 co-supplementation improve dyslipidemia and fatty liver syndrome by SIRT1/SREBP-1c pathway in hyperlipidemic Sprague-Dawley rats induced by high-fat diet. Nutr Res 2022; 106:101-118. [DOI: 10.1016/j.nutres.2022.06.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022]
|
30
|
Yu M, Teitelbaum SL, Dolios G, Dang LHT, Tu P, Wolff MS, Petrick LM. Molecular Gatekeeper Discovery: Workflow for Linking Multiple Exposure Biomarkers to Metabolomics. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:6162-6171. [PMID: 35129943 PMCID: PMC9164279 DOI: 10.1021/acs.est.1c04039] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The exposome reflects multiple exposures across the life-course that can affect health. Metabolomics can reveal the underlying molecular basis linking exposures to health conditions. Here, we explore the concept and general data analysis framework of "molecular gatekeepers"─key metabolites that link single or multiple exposure biomarkers with correlated clusters of endogenous metabolites─to inform health-relevant biological targets. We performed untargeted metabolomics on plasma from 152 adolescent girls participating in the Growing Up Healthy Study in New York City. We then performed network analysis to link metabolites to exposure biomarkers including five trace elements (Cd, Mn, Pb, Se, and Hg) and five perfluorinated chemicals (PFCs; n-PFOS, Sm-PFOS, n-PFOA, PFHxS, and PFNA). We found 144 molecular gatekeepers and annotated 22 of them. Lysophosphatidylcholine (16:0) and taurodeoxycholate were correlated with both n-PFOA and n-PFOS, suggesting a shared dysregulation from multiple xenobiotic exposures. Sphingomyelin (d18:2/14:0) was significantly associated with age at menarche; yet, no direct association was detected between any exposure biomarkers and age at menarche. Thus, molecular gatekeepers can also discover molecular linkages between exposure biomarkers and health outcomes that may otherwise be obscured by complex interactions in direct measurements.
Collapse
Affiliation(s)
- Miao Yu
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Susan L Teitelbaum
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Georgia Dolios
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Lam-Ha T Dang
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Peijun Tu
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Mary S Wolff
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Lauren M Petrick
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
31
|
Wang M, Xu W, Yu J, Liu Y, Ma H, Ji C, Zhang C, Xue J, Li R, Cui H. Astaxanthin From Haematococcus pluvialis Prevents High-Fat Diet-Induced Hepatic Steatosis and Oxidative Stress in Mice by Gut-Liver Axis Modulating Properties. Front Nutr 2022; 9:840648. [PMID: 35495929 PMCID: PMC9039660 DOI: 10.3389/fnut.2022.840648] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Scope Evidence is mounting that astaxanthin (ATX), a xanthophyll carotenoid, used as a nutritional supplement to prevent chronic metabolic diseases. The present study aims to identify the potential function of ATX supplementation in preventing steatohepatitis and hepatic oxidative stress in diet-induced obese mice. Methods and Results In this study, ATX as dose of 0.25, 0.5, and 0.75% have orally administered to mice along with a high-fat diet (HFD) to investigate the role of ATX in regulating liver lipid metabolism and gut microbiota. The study showed that ATX dose-dependently reduces body weight, lipid droplet formation, hepatic triglycerides and ameliorated hepatic steatosis and oxidative stress. 0.75% ATX altered the levels of 34 lipid metabolites related to hepatic cholesterol and fatty acid metabolism which might be associated with downregulation of lipogenesis-related genes and upregulation of bile acid biosynthesis-related genes. The result also revealed that ATX alleviates HFD-induced gut microbiota dysbiosis by significantly inhibiting the growth of obesity-related Parabacteroides and Desulfovibrio while promoting the growth of Allobaculum and Akkermansia. Conclusion The study results suggested that dietary ATX may prevent the development of hepatic steatosis and oxidative stress with the risk of metabolic disease by gut-liver axis modulating properties.
Collapse
Affiliation(s)
- Meng Wang
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Wenxin Xu
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Jie Yu
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Yingying Liu
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Haotian Ma
- Health Science Center, College of Forensic Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Chunli Ji
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Chunhui Zhang
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Jinai Xue
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China
| | - Runzhi Li
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China.,State Key Laboratory of Integrative Sustainable Dryland Agriculture, Shanxi Agricultural University, Taiyuan, China
| | - Hongli Cui
- College of Agriculture, Institute of Molecular Agriculture and Bioenergy, Shanxi Agricultural University, Jinzhong, China.,State Key Laboratory of Integrative Sustainable Dryland Agriculture, Shanxi Agricultural University, Taiyuan, China
| |
Collapse
|
32
|
Ban Q, Sun X, Jiang Y, Cheng J, Guo M. Effect of synbiotic yogurt fortified with monk fruit extract on hepatic lipid biomarkers and metabolism in rats with type 2 diabetes. J Dairy Sci 2022; 105:3758-3769. [PMID: 35248379 DOI: 10.3168/jds.2021-21204] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/14/2022] [Indexed: 01/03/2024]
Abstract
Monk fruit extract (MFE) is widely used as a sweetener in foods. In this study, the effects of the consumption of MFE-sweetened synbiotic yogurt on the lipid biomarkers and metabolism in the livers of type 2 diabetic rats were evaluated. The results revealed that the MFE-sweetened symbiotic yogurt affected the phosphatidylcholines, phosphatidylethanolamines, phosphatidylglycerol, lysophosphatidic acids, lysophosphatidylcholines, lysophosphatidylethanolamines, lysophosphatidylglycerols, lysophosphatidylinositols, lysophosphatidylserines, and fatty acid-hydroxy fatty acids biomarkers in the livers of type 2 diabetic rats. In addition, the consumption of the MFE-sweetened synbiotic yogurt significantly altered 12 hepatic metabolites, which are involved in phenylalanine metabolism, sphingolipid metabolism, bile secretion, and glyoxylate and dicarboxylate metabolism in the liver. Furthermore, a multiomics (metabolomic and transcriptomic) association study revealed that there was a significant correlation between the MFE-sweetened synbiotic yogurt and the metabolites and genes involved in fatty acid biosynthesis, bile secretion, and glyoxylate and dicarboxylate metabolism. The findings of this study will provide new insights on exploring the function of sweeteners for improving type 2 diabetes mellitus liver lipid biomarkers.
Collapse
Affiliation(s)
- Qingfeng Ban
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China; Key Laboratory of Dairy Science of Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Xiaomeng Sun
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China
| | - Yunqing Jiang
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China
| | - Jianjun Cheng
- College of Food Science, Northeast Agricultural University, Harbin, 150030, China.
| | - Mingruo Guo
- Department of Nutrition and Food Sciences, College of Agriculture and Life Sciences, University of Vermont, Burlington 05405.
| |
Collapse
|
33
|
Tie D, Fan Z, Chen D, Chen X, Chen Q, Chen J, Bo H. Mechanisms of Danggui Buxue Tang on Hematopoiesis via Multiple Targets and Multiple Components: Metabonomics Combined with Database Mining Technology. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:1155-1171. [PMID: 35475977 DOI: 10.1142/s0192415x22500471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
This study aimed to explore the mechanism of action of Danggui Buxue Tang (DBT) with its multiple components and targets in the synergistic regulation of hematopoiesis. Mouse models of hematopoiesis were established using antibiotics. Metabolomics was used to detect body metabolites and enriched pathways. The active ingredients, targets, and pathways of DBT were analyzed using system pharmacology. The results of metabolomics and system pharmacology were integrated to identify the key pathways and targets. A total of 515 metabolites were identified using metabolomics. After the action of antibiotics, 49 metabolites were markedly changed: 23 were increased, 26 were decreased, and 11 were significantly reversed after DBT administration. Pathway enrichment analysis showed that these 11 metabolites were related to bile secretion, cofactor biosynthesis, and fatty acid biosynthesis. The results of the pharmacological analysis showed that 616 targets were related to DBT-induced anemia, which were mainly enriched in biological processes, such as bile secretion, biosynthesis of cofactors, and cholesterol metabolism. Combined with the results of metabolomics and system pharmacology, we found that bile acid metabolism and biotin synthesis were the key pathways for DBT. Forty-two targets of DBT were related to these two metabolic pathways. PPI analysis revealed that the top 10 targets were CYP3A4, ABCG2, and UGT1A8. Twenty-one components interacted with these 10 targets. In one case, a target corresponds to multiple components, and a component corresponds to multiple targets. DBT acts on multiple targets of ABCG2, UGT1A8, and CYP3A4 through multiple components, affecting the biosynthesis of cofactors and bile secretion pathways to regulate hematopoiesis.
Collapse
Affiliation(s)
- Defu Tie
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, P. R. China
| | - Zhaohui Fan
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, P. R. China
| | - Dan Chen
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, P. R. China
| | - Xiao Chen
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, P. R. China
| | - Qizhu Chen
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, P. R. China
| | - Jun Chen
- College of Pharmacy, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong, P. R. China
| | - Huaben Bo
- School of Bioscience and Biopharmaceutics, Guangdong Province Key Laboratory for Biotechnology Drug Candidates, P. R. China
| |
Collapse
|
34
|
Zhang R, Sun X, Huang Z, Pan Y, Westbrook A, Li S, Bazzano L, Chen W, He J, Kelly T, Li C. Examination of serum metabolome altered by cigarette smoking identifies novel metabolites mediating smoking-BMI association. Obesity (Silver Spring) 2022; 30:943-952. [PMID: 35258150 PMCID: PMC8957487 DOI: 10.1002/oby.23386] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/25/2021] [Accepted: 01/03/2022] [Indexed: 11/07/2022]
Abstract
OBJECTIVE The authors hypothesize that an untargeted metabolomics study will identify novel mechanisms underlying smoking-associated weight loss. METHODS This study performed cross-sectional analyses among 1,252 participants in the Bogalusa Heart Study and assessed 1,202 plasma metabolites for mediation effects on smoking-BMI associations. Significant metabolites were tested for associations with smoking genetic risk scores among a subset of participants (n = 654) with available genomic data, followed by direction dependence analysis to investigate causal relationships between the metabolites and smoking and BMI. All analyses controlled for age, sex, race, education, alcohol drinking, and physical activity. RESULTS Compared with never smokers, current and former smokers had a 3.31-kg/m2 and 1.77-kg/m2 lower BMI after adjusting for all covariables, respectively. A total of 22 xenobiotics and 94 endogenous metabolites were significantly associated with current smoking. Eight xenobiotics were also associated with former smoking. Forty metabolites mediated the smoking-BMI associations, and five showed causal relationships with both smoking and BMI. These metabolites, including 1-oleoyl-GPE (18:1), 1-linoleoyl-GPE (18:2), 1-stearoyl-2-arachidonoyl-GPE (18:0/20:4), α-ketobutyrate, and 1-palmitoyl-GPE (16:0), mediated 26.0% of the association between current smoking and BMI. CONCLUSIONS This study cataloged plasma metabolites altered by cigarette smoking and identified five metabolites that partially mediated the association between current smoking and BMI.
Collapse
Affiliation(s)
- Ruiyuan Zhang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, New Orleans, LA 70112, US
| | - Xiao Sun
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, New Orleans, LA 70112, US
| | - Zhijie Huang
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, New Orleans, LA 70112, US
| | - Yang Pan
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, New Orleans, LA 70112, US
| | - Adrianna Westbrook
- Pediatric Biostatistics Core, Department of Pediatrics, Emory University
| | - Shengxu Li
- Children’s Minnesota Research Institute, Children’s Hospitals and Clinics of Minnesota, Minneapolis, MN, US
| | - Lydia Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, New Orleans, LA 70112, US
| | - Wei Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, New Orleans, LA 70112, US
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, New Orleans, LA 70112, US
| | - Tanika Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, New Orleans, LA 70112, US
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, New Orleans, LA 70112, US
| |
Collapse
|
35
|
Sun S, Wang J, Yao J, Guo H, Dai J. Transcriptome analysis of 3D primary mouse liver spheroids shows that long-term exposure to hexafluoropropylene oxide trimer acid disrupts hepatic bile acid metabolism. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 812:151509. [PMID: 34762948 DOI: 10.1016/j.scitotenv.2021.151509] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/27/2021] [Accepted: 11/03/2021] [Indexed: 06/13/2023]
Abstract
Hexafluoropropylene oxide trimer acid (HFPO-TA), an alternative to perfluorooctanoic acid (PFOA), has been detected in various environmental and human matrices. However, information regarding its toxicity remains limited. Here, we established a three-dimensional (3D) primary mouse liver spheroid model to compare the hepatotoxicity of HFPO-TA and PFOA. The 3D spheroids were repeatedly exposed to 25-, 50-, or 100-μM HFPO-TA and PFOA for 28 d. Compared with the PFOA groups, the HFPO-TA groups showed higher bioaccumulation potential, higher lactate dehydrogenase (LDH) leakage, and lower adenosine triphosphate (ATP), albumin, and urea secretion. Transcriptome analysis identified 1603 and 772 differentially expressed genes in the 100-μM HFPO-TA- and PFOA-treated groups, respectively. Bioinformatics analysis indicated that cholesterol metabolism, bile acid metabolism, and inflammatory response were significantly altered. Exposure to 100-μM HFPO-TA increased triglyceride content but decreased total cholesterol content, while no changes were observed in the 100-μM PFOA-treated group. Total bile acids in the re-polarized 3D spheroids increased significantly after 100-μM HFPO-TA and PFOA treatment, which did not affect bile acid synthesis but inhibited the expression levels of Bsep and Mrp2 related to bile acid transport. Thus, HFPO-TA exhibited more serious hepatotoxicity than PFOA in 3D primary liver spheroids and may not be a safe alternative.
Collapse
Affiliation(s)
- Sujie Sun
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Sciences and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China; Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianshe Wang
- School of Pharmacy, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Yantai University, Yantai 264005, Shandong Province, China
| | - Jingzhi Yao
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Sciences and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China; Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hua Guo
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Sciences and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China; Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiayin Dai
- State Environmental Protection Key Laboratory of Environmental Health Impact Assessment of Emerging Contaminants, School of Environmental Sciences and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China; Key Laboratory of Animal Ecology and Conservation Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
36
|
Kovacevic B, Jones M, Ionescu C, Walker D, Wagle S, Chester J, Foster T, Brown D, Mikov M, Mooranian A, Al-Salami H. The emerging role of bile acids as critical components in nanotechnology and bioengineering: Pharmacology, formulation optimizers and hydrogel-biomaterial applications. Biomaterials 2022; 283:121459. [DOI: 10.1016/j.biomaterials.2022.121459] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 02/27/2022] [Accepted: 03/04/2022] [Indexed: 12/16/2022]
|
37
|
Interplay between Dysbiosis of Gut Microbiome, Lipid Metabolism, and Tumorigenesis: Can Gut Dysbiosis Stand as a Prognostic Marker in Cancer? DISEASE MARKERS 2022; 2022:2941248. [PMID: 35178126 PMCID: PMC8847007 DOI: 10.1155/2022/2941248] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 01/11/2022] [Indexed: 02/07/2023]
Abstract
The gut bacterial community is involved in the metabolism of bile acids and short-chain fatty acids (SCFAs). Bile acids are involved in the absorption of fat and the regulation of lipid homeostasis through emulsification and are transformed into unconjugated bile acids by the gut microbiota. The gut microbiota is actively involved in the production of bile acid metabolites, such as deoxycholic acid, lithocholic acid, choline, and SCFAs such as acetate, butyrate, and propionate. Metabolites derived from the gut microbiota or modified gut microbiota metabolites contribute significantly to host pathophysiology. Gut bacterial metabolites, such as deoxycholic acid, contribute to the development of hepatocellular carcinoma and colon cancer by factors such as inflammation and oxidative DNA damage. Butyrate, which is derived from gut bacteria such as Megasphaera, Roseburia, Faecalibacterium, and Clostridium, is associated with the activation of Treg cell differentiation in the intestine through histone acetylation. Butyrate averts the action of class I histone deacetylases (HDAC), such as HDAC1 and HDAC3, which are responsible for the transcription of genes such as p21/Cip1, and cyclin D3 through hyperacetylation of histones, which orchestrates G1 cell cycle arrest. It is essential to identify the interaction between the gut microbiota and bile acid and SCFA metabolism to understand their role in gastrointestinal carcinogenesis including colon, gastric, and liver cancer. Metagenomic approaches with bioinformatic analyses are used to identify the bacterial species in the metabolism of bile acids and SCFAs. This review provides an overview of the current knowledge of gut microbiota-derived bile acid metabolism in tumor development and whether it can stand as a marker for carcinogenesis. Additionally, this review assesses the evidence of gut microbiota-derived short-chain fatty acids including butyric acid in antitumor activity. Future research is required to identify the beneficial commensal gut bacteria and their metabolites which will be considered to be therapeutic targets in inflammation-mediated gastrointestinal cancers.
Collapse
|
38
|
Dong Z, He F, Yan X, Xing Y, Lei Y, Gao J, He M, Li D, Bai L, Yuan Z, Y-J. Shyy J. Hepatic Reduction in Cholesterol 25-Hydroxylase Aggravates Diet-induced Steatosis. Cell Mol Gastroenterol Hepatol 2022; 13:1161-1179. [PMID: 34990887 PMCID: PMC8873960 DOI: 10.1016/j.jcmgh.2021.12.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/28/2021] [Accepted: 12/28/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND & AIMS Cholesterol 25-hydroxylase (Ch25h), converting cholesterol to 25-hydroxycholesterol (25-HC), is critical in modulating cellular lipid metabolism and anti-inflammatory and antiviral activities. However, its role in nonalcoholic fatty liver disease remains unclear. METHODS Ch25h expression was detected in livers of ob/ob mice and E3 rats fed a high-fat diet (HFD). Gain- or loss-of-function of Ch25h was performed using Ch25h+/+ (wild type [WT]) mice receiving AAV8-Ch25h or Ch25h knockout (Ch25h-/-) mice. WT mice fed an HFD were administered with 25-HC. The Ch25h-LXRα-CYP axis was measured in primary hepatocytes isolated from WT and Ch25h-/- mice. RESULTS We found that Ch25h level was decreased in livers of ob/ob mice and E3 rats fed an HFD. Ch25h-/- mice fed an HFD showed aggravated fatty liver and decreased level of cytochrome P450 7A1 (CYP7A1), in comparison with their WT littermates. RNA-seq analysis revealed that the differentially expressed genes in livers of HFD-fed Ch25h-/- mice were involved in pathways of positive regulation of lipid metabolic process, steroid metabolic process, cholesterol metabolic process, and bile acid biosynthetic process. As gain-of-function experiments, WT mice receiving AAV8-Ch25h or 25-HC showed alleviated NAFLD, when compared with the control group receiving AAV8-control or vehicle control. Consistently, Ch25h overexpression significantly elevated the levels of primary and secondary bile acids and CYP7A1 but decreased those of small heterodimer partner and FGFR4. CONCLUSIONS Elevated levels of Ch25h and its enzymatic product 25-HC alleviate HFD-induced hepatic steatosis via regulating enterohepatic circulation of bile acids. The underlying mechanism involves 25-HC activation of CYP7A1 via liver X receptor. These data suggest that targeting Ch25h or 25-HC may have therapeutic advantages against nonalcoholic fatty liver disease.
Collapse
Affiliation(s)
- Zeyu Dong
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Fangzhou He
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Xiaosong Yan
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Yuanming Xing
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China,Department of Cardiology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yuyang Lei
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China,Department of Cardiology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Jie Gao
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China,Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi’an, Shaanxi, China
| | - Ming He
- Department of Medicine/Division of Cardiology, University of California, San Diego, La Jolla, California
| | - Dongmin Li
- Department of Genetics and Molecular Biology, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Liang Bai
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China,Department of Cardiology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China,Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi’an, Shaanxi, China,Correspondence Address correspondence to: Liang Bai, PhD, Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China. tel: 86 298 265 5363; fax: 86 298 265 5362.
| | - Zuyi Yuan
- Department of Cardiology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - John Y-J. Shyy
- Institute of Cardiovascular Science, Translational Medicine Institute, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China,Department of Medicine/Division of Cardiology, University of California, San Diego, La Jolla, California,John Y-J. Shyy, PhD, Department of Medicine/Division of Cardiology, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093. tel: (858) 534-3737.
| |
Collapse
|
39
|
Wang H, Li J, Leng J, Li W, Liu J, Yan X, Yu Z, Hu G, Ma RCW, Fang Z, Wang Y, Yang X. The CDKAL1 rs7747752-Bile Acids Interaction Increased Risk of Gestational Diabetes Mellitus: A Nested Case-Control Study. Front Endocrinol (Lausanne) 2022; 13:808956. [PMID: 35360068 PMCID: PMC8960111 DOI: 10.3389/fendo.2022.808956] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/16/2022] [Indexed: 11/19/2022] Open
Abstract
AIMS The study aimed to explore additive interactions of CDKAL1 rs7747752 and GUDCA/DCA for GDM risk and whether the interactive effects on the risk of GDM was mediated via increasing lysophosphatidylcholines (LPC) 18:0 and/or saturated fatty acid (SFA) 16:0. METHODS A 1:1 age-matched study nested in a prospective cohort of pregnant women (207 pairs) was organized in Tianjin, China. Additive interactions were used to test interaction effects while mediation analyses and Sobel tests were used to test mediation effects of LPC18:0 and SFA16:0 between copresence of rs7747752 and low GUDCA/DCA, and GDM risk. RESULTS The CDKAL1 rs7747752 was associated with GDM (P<0.05). The rs7747752 C polymorphism markedly enhanced ORs of low GUDCA from 4.04 (0.72-22.8) to 9.02 (1.63-49.7) and low DCA from 1.67 (0.68-4.11) to 4.24 (1.84-9.76), both with significant additive interactions. Further adjustment for LPC18:0 attenuated the interactive effects of rs7747752 and low DCA, with a significant mediation effect (P=0.003). High SFA16:0 did not mediate the interactive effects of rs7747752 and low DCA/GUDCA on GDM risk. CONCLUSIONS The CDKAL1 rs7747752 C carrier status and low GUDCA/DCA had significant additive interactions on the risk of GDM with the effect from interaction with DCA being partially mediated via increasing LPC18:0.
Collapse
Affiliation(s)
- Hui Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Jing Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin, China
| | - Junhong Leng
- Project Office, Tianjin Women and Children’s Health Center, Tianjin, China
| | - Weiqin Li
- Project Office, Tianjin Women and Children’s Health Center, Tianjin, China
| | - Jinnan Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Xiaoyan Yan
- School of Public Health, Shanxi Medical University, Shanxi, China
| | - Zhijie Yu
- Population Cancer Research Program and Department of Pediatrics, Dalhousie University, Halifax, NS, Canada
| | - Gang Hu
- Chronic Disease Epidemiology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Ronald C. W. Ma
- Department of Medicine and Therapeutics and Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zhongze Fang
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin, China
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
- *Correspondence: Xilin Yang, ; ; Ying Wang, ; Zhongze Fang,
| | - Ying Wang
- Scientific Research Platform of the Second School of Clinical Medicine & Key Laboratory of 3D Printing Technology in Stomatology, Guangdong Medical University, Dongguan, China
- *Correspondence: Xilin Yang, ; ; Ying Wang, ; Zhongze Fang,
| | - Xilin Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Key Laboratory of Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin, China
- Tianjin Center for International Collaborative Research on Environment, Nutrition and Public Health, Tianjin Medical University, Tianjin, China
- *Correspondence: Xilin Yang, ; ; Ying Wang, ; Zhongze Fang,
| |
Collapse
|
40
|
Zhang S, Zhou J, Wu W, Zhu Y, Liu X. The Role of Bile Acids in Cardiovascular Diseases: from Mechanisms to Clinical Implications. Aging Dis 2022; 14:261-282. [PMID: 37008052 PMCID: PMC10017164 DOI: 10.14336/ad.2022.0817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022] Open
Abstract
Bile acids (BAs), key regulators in the metabolic network, are not only involved in lipid digestion and absorption but also serve as potential therapeutic targets for metabolic disorders. Studies have shown that cardiac dysfunction is associated with abnormal BA metabolic pathways. As ligands for several nuclear receptors and membrane receptors, BAs systematically regulate the homeostasis of metabolism and participate in cardiovascular diseases (CVDs), such as myocardial infarction, diabetic cardiomyopathy, atherosclerosis, arrhythmia, and heart failure. However, the molecular mechanism by which BAs trigger CVDs remains controversial. Therefore, the regulation of BA signal transduction by modulating the synthesis and composition of BAs is an interesting and novel direction for potential therapies for CVDs. Here, we mainly summarized the metabolism of BAs and their role in cardiomyocytes and noncardiomyocytes in CVDs. Moreover, we comprehensively discussed the clinical prospects of BAs in CVDs and analyzed the clinical diagnostic and application value of BAs. The latest development prospects of BAs in the field of new drug development are also prospected. We aimed to elucidate the underlying mechanism of BAs treatment in CVDs, and the relationship between BAs and CVDs may provide new avenues for the prevention and treatment of these diseases.
Collapse
Affiliation(s)
- Shuwen Zhang
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Junteng Zhou
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
- Health Management Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Wenchao Wu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Ye Zhu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China.
- Correspondence should be addressed to: Prof. Xiaojing Liu (), and Prof. Ye Zhu (), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaojing Liu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China.
- Correspondence should be addressed to: Prof. Xiaojing Liu (), and Prof. Ye Zhu (), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
41
|
Jeong DW, Lee S, Chun YS. How cancer cells remodel lipid metabolism: strategies targeting transcription factors. Lipids Health Dis 2021; 20:163. [PMID: 34775964 PMCID: PMC8590761 DOI: 10.1186/s12944-021-01593-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/02/2021] [Indexed: 12/30/2022] Open
Abstract
Reprogramming of lipid metabolism has received increasing recognition as a hallmark of cancer cells because lipid dysregulation and the alteration of related enzyme profiles are closely correlated with oncogenic signals and malignant phenotypes, such as metastasis and therapeutic resistance. In this review, we describe recent findings that support the importance of lipids, as well as the transcription factors involved in cancer lipid metabolism. With recent advances in transcription factor analysis, including computer-modeling techniques, transcription factors are emerging as central players in cancer biology. Considering the limited number and the crucial role of transcription factors associated with lipid rewiring in cancers, transcription factor targeting is a promising potential strategy for cancer therapy.
Collapse
Affiliation(s)
- Do-Won Jeong
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea.,Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Seulbee Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea.,Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Yang-Sook Chun
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea. .,Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea. .,Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul, 03080, South Korea.
| |
Collapse
|
42
|
Tang J, Qin M, Tang L, Shan D, Zhang C, Zhang Y, Wei H, Qiu L, Yu J. Enterobacter aerogenes ZDY01 inhibits choline-induced atherosclerosis through CDCA-FXR-FGF15 axis. Food Funct 2021; 12:9932-9946. [PMID: 34492674 DOI: 10.1039/d1fo02021h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Atherosclerosis is the leading cause of cardiovascular diseases worldwide. Trimethylamine N-oxide (TMAO), a metabolite of intestinal flora from dietary quaternary amines, has been shown to be closely related to the development of atherosclerosis. Previous studies have shown that Enterobacter aerogenes ZDY01 significantly reduces the serum levels of TMAO and cecal trimethylamine (TMA) in Balb/c mice; however, its role in the inhibition of choline-induced atherosclerosis in ApoE-/- mice remains unclear. Here, we demonstrated that E. aerogenes ZDY01 inhibited choline-induced atherosclerosis in ApoE-/- mice fed with 1.3% choline by reducing cecal TMA and modulating CDCA-FXR/FGF15 axis. We observed that E. aerogenes ZDY01 decreased the cecal TMA and serum TMAO levels by utilizing cecal TMA as a nutrient, not by changing the expression of hepatic FMO3 and the composition of gut microbiota. Furthermore, E. aerogenes ZDY01 enhanced the expression of bile acid transporters and reduced the cecal CDCA levels, thereby attenuating the FXR/FGF15 pathway, upregulating the expression of Cyp7a1, promoting reverse cholesterol transport. Taken together, E. aerogenes ZDY01 attenuated choline-induced atherosclerosis in ApoE-/- mice by decreasing cecal TMA and promoting reverse cholesterol transport, implying that E. aerogenes ZDY01 treatment might have therapeutic potential in atherosclerosis.
Collapse
Affiliation(s)
- Jinghui Tang
- Key Laboratory for Pharmacology and Translational Research of Traditional Chinese Medicine of Nanchang, Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China. .,Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodelling Diseases, China
| | - Manman Qin
- Key Laboratory for Pharmacology and Translational Research of Traditional Chinese Medicine of Nanchang, Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China. .,Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodelling Diseases, China
| | - Le Tang
- Key Laboratory for Pharmacology and Translational Research of Traditional Chinese Medicine of Nanchang, Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China. .,Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodelling Diseases, China
| | - Dan Shan
- Key Laboratory for Pharmacology and Translational Research of Traditional Chinese Medicine of Nanchang, Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China. .,Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodelling Diseases, China
| | - Cheng Zhang
- Department of Physiology and Centre for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| | - Yifeng Zhang
- Key Laboratory for Pharmacology and Translational Research of Traditional Chinese Medicine of Nanchang, Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China. .,Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodelling Diseases, China
| | - Hua Wei
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, 330047, P. R. China
| | - Liang Qiu
- Key Laboratory for Pharmacology and Translational Research of Traditional Chinese Medicine of Nanchang, Centre for Translational Medicine, Jiangxi University of Chinese Medicine, Nanchang 330006, China. .,Jiangxi Key Laboratory of Traditional Chinese Medicine for Prevention and Treatment of Vascular Remodelling Diseases, China
| | - Jun Yu
- Department of Physiology and Centre for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
43
|
Liu N, Chen X, Song J, Chen M, Gong P, Jia W, Li G. Hypoglycemic effects of Auricularia auricula polysaccharides on high fat diet and streptozotocin-induced diabetic mice using metabolomics analysis. Food Funct 2021; 12:9994-10007. [PMID: 34505615 DOI: 10.1039/d1fo02022f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This study evaluated the hypoglycemic effect of Auricularia auricula polysaccharides (AAPs) on streptozotocin-induced type 2 diabetes mellitus (T2DM) mice using metabolomic analysis. The results of fasting blood glucose, oral glucose tolerance test, fasting serum insulin level, Homeostatic Model Assessment of Insulin Resistance index, TC, TG, HDL-C, LDL-C, and histopathological observation demonstrated that 200 mg per kg body weight per day AAP led to significant hypoglycemic activities. The metabolic profile of the mice was significantly changed after AAP intervention. 45 differential metabolites were screened as biomarkers for AAP adjuvant treatment, and AAPs' effects on the metabolism of amino acids, unsaturated fatty acids, bile acids, and glycerophospholipids were analyzed. Thus, the current results elucidated the metabolic pathway of AAPs for T2DM alleviation and provided guidance for functional food adjuvant development for T2DM treatment.
Collapse
Affiliation(s)
- Nannan Liu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China. .,College of Chemistry and Materials Science, Weinan Normal University, Weinan 714000, China
| | - Xuefeng Chen
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Juanna Song
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Mengyin Chen
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Pin Gong
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Wei Jia
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Guoliang Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| |
Collapse
|
44
|
Lin Y, Chen H, Cao Y, Zhang Y, Li W, Guo W, Lv X, Rao P, Ni L, Liu P. Auricularia auricula Melanin Protects against Alcoholic Liver Injury and Modulates Intestinal Microbiota Composition in Mice Exposed to Alcohol Intake. Foods 2021; 10:foods10102436. [PMID: 34681485 PMCID: PMC8535349 DOI: 10.3390/foods10102436] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 09/26/2021] [Accepted: 09/28/2021] [Indexed: 12/12/2022] Open
Abstract
The potential effects of Auricularia auricula melanin (AAM) on the intestinal flora and liver metabolome in mice exposed to alcohol intake were investigated for the first time. The results showed that oral administration of AAM significantly reduced the abnormal elevation of serum total triglyceride (TG), cholesterol (TC), low density lipoprotein cholesterol (LDL-C), aspartate aminotransferase (AST) and alanine aminotransferase (ALT), and significantly inhibited hepatic lipid accumulation and steatosis in mice exposed to alcohol intake. Besides, the abnormally high levels of bile acids (BAs) and lactate dehydrogenase (LDH) in the liver of mice with alcohol intake were significantly decreased by AAM intervention, while the hepatic levels of glutathione (GSH) and superoxide dismutase (SOD) were appreciably increased. Compared with the model group, AAM supplementation significantly changed the composition of intestinal flora and up-regulated the levels of Akkermansia, Bifidobacterium, Romboutsia, Muribaculaceae, Lachnospiraceae_NK4A136_group, etc. Furthermore, liver metabolomics demonstrated that AAM had a significant regulatory effect on the composition of liver metabolites in mice with alcohol intake, especially the metabolites involved in phosphatidylinositol signaling system, ascorbate and aldarate metabolism, starch and sucrose metabolism, galactose metabolism, alpha-linolenic acid metabolism, glycolysis/gluconeogenesis, and biosynthesis of unsaturated fatty acids. At the gene level, AAM treatment regulated the mRNA levels of lipid metabolism and inflammatory response related genes in liver, including ACC-1, FASn, CPT-1, CD36, IFN-γ, LDLr and TNF-α. Conclusively, these findings suggest that AAM has potential beneficial effects on alleviating alcohol-induced liver injury and is expected to become a new functional food ingredient.
Collapse
Affiliation(s)
- Yichen Lin
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.L.); (Y.C.); (W.G.)
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China; (Y.Z.); (P.R.); (L.N.)
| | - Hua Chen
- Fujian Province Key Laboratory of Agro-Ecological Processes in Hilly Red Soil, Agricultural Ecology Institute, Fujian Academy of Agriculture Sciences, Fuzhou 350003, China;
| | - Yingjia Cao
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.L.); (Y.C.); (W.G.)
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China; (Y.Z.); (P.R.); (L.N.)
| | - Yuanhui Zhang
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China; (Y.Z.); (P.R.); (L.N.)
| | - Wenfeng Li
- Department of Nutrition and Food Safety, School of Public Health, Fujian Medical University, Fuzhou 350122, China;
| | - Weiling Guo
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.L.); (Y.C.); (W.G.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Xucong Lv
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China; (Y.Z.); (P.R.); (L.N.)
- Correspondence: (X.L.); (P.L.)
| | - Pingfan Rao
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China; (Y.Z.); (P.R.); (L.N.)
| | - Li Ni
- Institute of Food Science and Technology, College of Biological Science and Technology, Fuzhou University, Fuzhou 350108, China; (Y.Z.); (P.R.); (L.N.)
| | - Penghu Liu
- National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (Y.L.); (Y.C.); (W.G.)
- Correspondence: (X.L.); (P.L.)
| |
Collapse
|
45
|
Kanu AB. Recent developments in sample preparation techniques combined with high-performance liquid chromatography: A critical review. J Chromatogr A 2021; 1654:462444. [PMID: 34380070 DOI: 10.1016/j.chroma.2021.462444] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 12/29/2022]
Abstract
This review article compares and contrasts sample preparation techniques coupled with high-performance liquid chromatography (HPLC) and describes applications developed in biomedical, forensics, and environmental/industrial hygiene in the last two decades. The proper sample preparation technique can offer valued data for a targeted application when coupled to HPLC and a suitable detector. Improvements in sample preparation techniques in the last two decades have resulted in efficient extraction, cleanup, and preconcentration in a single step, thus providing a pathway to tackle complex matrix applications. Applications such as biological therapeutics, proteomics, lipidomics, metabolomics, environmental/industrial hygiene, forensics, glycan cleanup, etc., have been significantly enhanced due to improved sample preparation techniques. This review looks at the early sample preparation techniques. Further, it describes eight sample preparation technique coupled to HPLC that has gained prominence in the last two decades. They are (1) solid-phase extraction (SPE), (2) liquid-liquid extraction (LLE), (3) gel permeation chromatography (GPC), (4) Quick Easy Cheap Effective Rugged, Safe (QuEChERS), (5) solid-phase microextraction (SPME), (6) ultrasonic-assisted solvent extraction (UASE), and (7) microwave-assisted solvent extraction (MWASE). SPE, LLE, GPC, QuEChERS, and SPME can be used offline and online with HPLC. UASE and MWASE can be used offline with HPLC but have also been combined with the online automated techniques of SPE, LLE, GPC, or QuEChERS for targeted analysis. Three application areas of biomedical, forensics, and environmental/industrial hygiene are reviewed for the eight sample preparation techniques. Three hundred and twenty references on the eight sample preparation techniques published over the last two decades (2001-2021) are provided. Other older references were included to illustrate the historical development of sample preparation techniques.
Collapse
Affiliation(s)
- A Bakarr Kanu
- Department of Chemistry, Winston-Salem State University, Winston-Salem, NC 27110, United States.
| |
Collapse
|
46
|
Zhang X, Li Y, Wei X, Hou Y, Jia S, Li S, Zhao X. Metabolomics analysis of the effects of quercetin on hepatotoxicity induced by acrylamide exposure in rats. Free Radic Res 2021; 55:831-841. [PMID: 34238086 DOI: 10.1080/10715762.2021.1950705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Acrylamide (AA) widely exists in human diet, which makes the public inevitably exposed to AA in daily life. Thisstudy aimed to investigatethe effects of quercetin on AA-induced hepatotoxicity utilizing metabolomics technology. Sixty male Wistar Rats were randomly divided into six groups: control, two dosages of quercetinintervention[10 and 50 mg/kgbody weight (bw)], AA-treated [5 mg/kgbw], and two dosages of quercetin combined with AA intervention. AA and quercetin were given to rats via drinking water and gavage respectively. After 16 weeksoftreatment, liver samples were collected for metabolomics analysis. 16metabolites were finally identified, the intensities of glutathione and NADP were decreased (p < 0.01), whereas the intensities of taurodeoxycholic acid, glycocholic acid, cholic acid, sphingosine, sphingosine1-phosphate, stearidonyl carnitine, N-undecanoylglycine, cholesterol, 13,14-Dihydro-15-keto-PGE2, LysoPE (20:5), LysoPE (18:3), LysoPC (20:4), and PC (22:5) were increased (p < 0.01) in the AA-treated groupthan those in the control group. After high-dose quercetin (50 mg/kgbw) plus AA treated concurrently to rats, the contents of the above 16 metabolites were significantly restored. This research showed that 50 mg/kg quercetin can alleviate AA-induced hepatotoxicity by reducing oxidative stress and inflammatory injury and regulating lipid metabolism.
Collapse
Affiliation(s)
- Xia Zhang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yaru Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinchen Wei
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yali Hou
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Siqi Jia
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Siqi Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiujuan Zhao
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
47
|
Effects of porcine bile acids on growth performance, antioxidant capacity, blood metabolites and nutrient digestibility of weaned pigs. Anim Feed Sci Technol 2021. [DOI: 10.1016/j.anifeedsci.2021.114931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
48
|
Faradonbeh FA, Sa II, Lastuvkova H, Cermanova J, Hroch M, Faistova H, Mokry J, Nova Z, Uher M, Nachtigal P, Pavek P, Micuda S. Metformin impairs bile acid homeostasis in ethinylestradiol-induced cholestasis in mice. Chem Biol Interact 2021; 345:109525. [PMID: 34058177 DOI: 10.1016/j.cbi.2021.109525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/28/2021] [Accepted: 05/16/2021] [Indexed: 12/12/2022]
Abstract
Metformin, an oral antidiabetic drug, recently demonstrated a reducing effect on bile acids (BA) plasma concentrations in one patient with intrahepatic cholestasis of pregnancy (ICP) by unknown mechanism. Therefore, the aim of the present study was to examine the effect of metformin on BA homeostasis and related molecular pathways in the liver and intestine using a mouse model of ICP. The cholestasis was induced in female C57BL/6 mice by repeated administration of ethinylestradiol (10 mg/kg BW s.c.) and/or metformin (150 mg/kg BW orally) over 5 consecutive days with subsequent bile collection and molecular analysis of samples. We demonstrated that metformin significantly increased the rate of bile secretion in control mice. This increase was BA dependent and was produced both by increased liver BA synthesis via induced cholesterol 7α-hydroxylase (Cyp7a1) and by increased BA reabsorption in the ileum via induction of the apical sodium-dependent BA transporter (Asbt). In contrast, metformin further worsened ethinylestradiol-induced impairment of bile secretion. This reduction was also BA dependent and corresponded with significant downregulation of Bsep, and Ntcp, major excretory and uptake transporters for BA in hepatocytes, respectively. The plasma concentrations of BA were consequently significantly increased in the metformin-treated mice. Altogether, our data indicate positive stimulation of bile secretion by metformin in the intact liver, but this drug also induces serious impairment of BA biliary secretion, with a marked increase in plasma concentrations in estrogen-induced cholestasis. Our results imply that metformin should be used with caution in situations with hormone-dependent cholestasis, such as ICP.
Collapse
Affiliation(s)
- Fatemeh Alaei Faradonbeh
- Department of Pharmacology, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Ivone Igreja Sa
- Department of Biological and Medical Sciences, Charles University, Faculty of Pharmacy in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Hana Lastuvkova
- Department of Pharmacology, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jolana Cermanova
- Department of Pharmacology, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Milos Hroch
- Department of Medical Biochemistry, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Hana Faistova
- Department of Pathology, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Jaroslav Mokry
- Department of Histology and Embryology, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Zuzana Nova
- Department of Pharmacology, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Martin Uher
- Department of Medical Biochemistry, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Petr Nachtigal
- Department of Biological and Medical Sciences, Charles University, Faculty of Pharmacy in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Petr Pavek
- Department of Pharmacology and Toxicology, Charles University, Faculty of Pharmacy in Hradec Kralove, Hradec Kralove, Czech Republic
| | - Stanislav Micuda
- Department of Pharmacology, Charles University, Faculty of Medicine in Hradec Kralove, Hradec Kralove, Czech Republic.
| |
Collapse
|
49
|
Mu J, Tan F, Zhou X, Zhao X. Lactobacillus fermentum CQPC06 in naturally fermented pickles prevents non-alcoholic fatty liver disease by stabilizing the gut-liver axis in mice. Food Funct 2021; 11:8707-8723. [PMID: 32945305 DOI: 10.1039/d0fo01823f] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Herein, we used a HFD/F to induce NAFLD in mice and intervened with CQPC06 to determine the preventive effect of CQPC06 on NAFLD and its potential regulatory mechanism. C57BL/6J mice were fed with LFD, HFD/F, HFD/F supplemented with CQPC06, and HFD/F supplemented with LDBS for 8 weeks to test the properties of the probiotic. Biochemical and molecular biology methods were used to determine the levels of related indexes in mouse serum, liver tissue, epididymal fat, small intestine tissue, and feces. The results showed that CQPC06 exhibited satisfactory probiotic properties, significantly inhibited mouse weight gain, and decreased the liver index and serum lipid levels, including ALT, AKP, AST, TC, TG, LDL-C, LPS, and HDL-C levels. The HOMA-IR index calculated based on the blood glucose levels and serum insulin levels showed that the HOMA-IR index of NAFLD mice treated with CQPC06 significantly decreased. From the molecular biology level, CQPC06 significantly increased the mRNA and protein expression of PPAR-α, CYP7A1, CPT1, and LPL in NAFLD mouse livers, and decreased the expression of PPAR-γ and C/EBP-α. Furthermore, CQPC06 enhanced the expression of ZO-1, occludin, and claudin-1 in the small intestine of NAFLD mice, and decreased the expression of CD36. CQPC06 decreased the level of Firmicutes and increased the levels of Bacteroides and Akkermansia in the feces of NAFLD mice, and the ratio of Firmicutes/Bacteroides was significantly decreased. CQPC06 is highly resistant in vitro and survived in the gastrointestinal tract and exerted its probiotic effect, altered the intestinal microecology of NAFLD mice, and played an important role in NAFLD prevention through the unique anatomical advantages of the gut-liver axis. There was a clear preventive effect with high concentrations of CQPC06 and it was stronger than that of l-carnitine.
Collapse
Affiliation(s)
- Jianfei Mu
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, China and College of Food Science, Southwest University, Chongqing 400715, China
| | - Fang Tan
- Department of Public Health, Our Lady of Fatima University, Valenzuela 838, Philippines
| | - Xianrong Zhou
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, China
| | - Xin Zhao
- Chongqing Collaborative Innovation Center for Functional Food, Chongqing University of Education, Chongqing 400067, China. and Chongqing Engineering Research Center of Functional Food, Chongqing University of Education, Chongqing 400067, China and Chongqing Engineering Laboratory for Research and Development of Functional Food, Chongqing University of Education, Chongqing 400067, China
| |
Collapse
|
50
|
Reiter S, Dunkel A, Metwaly A, Panes J, Salas A, Haller D, Hofmann T. Development of a Highly Sensitive Ultra-High-Performance Liquid Chromatography Coupled to Electrospray Ionization Tandem Mass Spectrometry Quantitation Method for Fecal Bile Acids and Application on Crohn's Disease Studies. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:5238-5251. [PMID: 33890469 DOI: 10.1021/acs.jafc.1c00769] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
In addition to their important role in fat digestion, bile acids are increasingly being used as markers for various diseases. The large diversity of bile acids results from the conversion of primary and conjugated bile acids into secondary bile acids by deconjugation and dehydroxylation reactions mediated by the intestinal microbiota. Here, we describe a fast and sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for absolute quantitation of 45 bile acids in human or mouse feces in combination with a simple workup and extraction procedure. Method validation outlined excellent limits of detection and quantitation, linearity, selectivity, recovery, extraction loss, and precision. To investigate the connection between microbiome alterations and bile acid metabolism, the method was applied on a Crohn's disease study including patients with histologically documented active disease or remission as well as on a model using humanized mice. As the complex mechanism including genetic and environmental factors leading to the development of Crohn's disease is so far not completely understood, the study investigates the microbial metabolism of bile acids and the potential use of bile acid profiles to predict disease state.
Collapse
Affiliation(s)
- Sinah Reiter
- Chair for Food Chemistry and Molecular Sensory Science, Technical University of Munich, Lise-Meitner-Straße 34, D-85354 Freising, Germany
- ZIEL-Institute for Food and Health, Technical University of Munich, Weihenstephaner Berg 1, 85354 Freising, Germany
| | - Andreas Dunkel
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich, Lise-Meitner-Straße 34, 85354 Freising, Germany
| | - Amira Metwaly
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Julian Panes
- Inflammatory Bowel Disease Unit, Hospital Clínic de Barcelona, IDIBAPS, CIBERehd, c/ Rosselló 149-153, 08036 Barcelona, Spain
| | - Azucena Salas
- Inflammatory Bowel Disease Unit, Hospital Clínic de Barcelona, IDIBAPS, CIBERehd, c/ Rosselló 149-153, 08036 Barcelona, Spain
| | - Dirk Haller
- ZIEL-Institute for Food and Health, Technical University of Munich, Weihenstephaner Berg 1, 85354 Freising, Germany
- Chair of Nutrition and Immunology, Technical University of Munich, Gregor-Mendel-Str. 2, 85354 Freising, Germany
| | - Thomas Hofmann
- Chair for Food Chemistry and Molecular Sensory Science, Technical University of Munich, Lise-Meitner-Straße 34, D-85354 Freising, Germany
| |
Collapse
|