1
|
Zhao M, Qiao K, Zhang L, Liang L, Chen S, Chen L, Zhang Y. Research Progress on Anti-Hyperlipidemia Peptides Derived from Foods. Nutrients 2025; 17:1181. [PMID: 40218939 PMCID: PMC11990363 DOI: 10.3390/nu17071181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/20/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Hyperlipidemia is a metabolic disorder in which cholesterol (TC) and triglycerides (TGs) in the blood exceed the normal physiological levels. The incidence of the condition has continued to rise in recent years, posing a serious threat to public health. Its clinical treatment mainly relies on drug interventions, such as statins, fibrate, and niacin. Although these drugs have shown some efficacy in the treatment of hyperlipidemia, their adverse effects cannot be ignored. In contrast, naturally derived peptides have gradually become potential candidates for the prevention and treatment of hyperlipidemia due to their strong anti-hyperlipidemic activity and safety; examples of such peptides include those from dairy products, grains, legumes, and seafood. This review systematically summarizes peptides with anti-hyperlipidemic activity and analyzes their mechanisms of action, providing a theoretical basis for further research. In addition, we also outline some challenges facing the application of peptides, hoping to prevent hyperlipidemia and reduce its incidence by encouraging the consumption of foods rich in anti-hyperlipidemia peptides.
Collapse
Affiliation(s)
- Mingxia Zhao
- Food Laboratory of Zhongyuan·Beijing Technology and Business University, Luohe 462300, China
- Food Laboratory of Zhongyuan, Luohe Food Engineering Vocational University, Luohe 462000, China
| | - Kaina Qiao
- Food Laboratory of Zhongyuan·Beijing Technology and Business University, Luohe 462300, China
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Flavor Science of China General Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
| | - Lili Zhang
- Food Laboratory of Zhongyuan·Beijing Technology and Business University, Luohe 462300, China
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Flavor Science of China General Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
| | - Li Liang
- Food Laboratory of Zhongyuan·Beijing Technology and Business University, Luohe 462300, China
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Flavor Science of China General Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
| | - Shuxing Chen
- Food Laboratory of Zhongyuan, Luohe 462300, China
| | - Lishui Chen
- Food Laboratory of Zhongyuan, Luohe 462300, China
| | - Yuyu Zhang
- Food Laboratory of Zhongyuan·Beijing Technology and Business University, Luohe 462300, China
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Flavor Science of China General Chamber of Commerce, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
2
|
Dai H, Shan Z, Shi L, Duan Y, An Y, He C, Lyu Y, Zhao Y, Wang M, Du Y, Xie J, Yang Y, Zhao B. Mulberry leaf polysaccharides ameliorate glucose and lipid metabolism disorders via the gut microbiota-bile acids metabolic pathway. Int J Biol Macromol 2024; 282:136876. [PMID: 39490871 DOI: 10.1016/j.ijbiomac.2024.136876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Mulberry leaf polysaccharides (MLP) are integral components of Mulberry leaves that confer hypoglycemic and hypolipidemic properties. This study investigated the efficacy of MLP in treating Type 2 Diabetes Mellitus (T2DM) and the underlying mechanisms related to gut microbiota-bile acids metabolism in T2DM rats. The findings revealed that MLP apparently reduced fasting blood glucose and lipid levels, ameliorated disorders in glucose and lipid metabolism, and mitigated insulin resistance. MLP enhanced the abundance of Prevotella, Ruminococcus, and Lactobacillus, thereby rectifying the gut microbiota dysbiosis in rats, which effectively restored gut microbiota homeostasis and composition. Furthermore, the data demonstrated that MLP modulated bile acid metabolism, as evidenced by reduced serum cholesterol levels, enhanced mRNA expression of hepatic cholesterol 7α- hydroxylase (Cyp7a1) and cholesterol 12α- hydroxylase (Cyp8b1), and ileal G protein-coupled bile acid receptor (Tgr5), while suppressing hepatic and ileal farnesoid X receptor (Fxr) mRNA expression in T2DM rats. Additionally, MLP upregulated the protein expression of hepatic CYP7A1 and CYP8B1, and ileal TGR5, while inhibiting FXR protein levels in the liver and ileum of T2DM rats. These results suggest that MLP can rectify disorders in glucose and lipid metabolism via the gut microbiota-bile acids metabolic pathway.
Collapse
Affiliation(s)
- Hongyu Dai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Operations and Management Department, Ya'an People's Hospital, Ya'an 625099, China
| | - Ziyi Shan
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Lu Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Central Laboratories, Qingdao Municipal Hospital, Qingdao 266011, China
| | - Yuhui Duan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yongcheng An
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Changhao He
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yinglan Lyu
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yige Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Menglu Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yuhang Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jiamei Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yang Yang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Baosheng Zhao
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
3
|
Alpízar M, de Jesús Reséndiz J, García Martínez E, Dwivedi S, Trejo MA. Pharmacokinetic Simulation and Area under the Curve Estimation of Drugs Subject to Enterohepatic Circulation. Pharmaceutics 2024; 16:1044. [PMID: 39204389 PMCID: PMC11360071 DOI: 10.3390/pharmaceutics16081044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/13/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Enterohepatic circulation (EHC) is a complex process where drugs undergo secretion and reabsorption from the intestinal lumen multiple times, resulting in pharmacokinetic profiles with multiple peaks. The impact of EHC on area under the curve (AUC) has been a topic of extensive debate, questioning the suitability of conventional AUC estimation methods. Moreover, a universal model for accurately estimating AUC in EHC scenarios is lacking. To address this gap, we conducted a simulation study evaluating five empirical models under various sampling strategies to assess their performance in AUC estimation. Our results identify the most suitable model for EHC scenarios and underscore the critical role of meal-based sampling strategies in accurate AUC estimation. Additionally, we demonstrate that while the trapezoidal method performs comparably to other models with a large number of samples, alternative models are essential when sample numbers are limited. These findings not only illuminate how EHC influences AUC but also pave the way for the application of empirical models in real-world drug studies.
Collapse
Affiliation(s)
- Melchor Alpízar
- Specialized Centre for Diabetes, Obesity and Prevention of Cardiovascular Diseases (CEDOPEC) & Bioequivalence Studies (E-BIOS-CEDOPEC), Mexico City 11650, Mexico; (J.d.J.R.); (E.G.M.); (S.D.)
| | | | | | | | - Miguel Alejandro Trejo
- Specialized Centre for Diabetes, Obesity and Prevention of Cardiovascular Diseases (CEDOPEC) & Bioequivalence Studies (E-BIOS-CEDOPEC), Mexico City 11650, Mexico; (J.d.J.R.); (E.G.M.); (S.D.)
| |
Collapse
|
4
|
Nguyen Y, Rudd Zhong Manis J, Ronczkowski NM, Bui T, Oxenrider A, Jadeja RN, Thounaojam MC. Unveiling the gut-eye axis: how microbial metabolites influence ocular health and disease. Front Med (Lausanne) 2024; 11:1377186. [PMID: 38799150 PMCID: PMC11122920 DOI: 10.3389/fmed.2024.1377186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/19/2024] [Indexed: 05/29/2024] Open
Abstract
The intricate interplay between the gut microbiota and ocular health has surpassed conventional medical beliefs, fundamentally reshaping our understanding of organ interconnectivity. This review investigates into the intricate relationship between gut microbiota-derived metabolites and their consequential impact on ocular health and disease pathogenesis. By examining the role of specific metabolites, such as short-chain fatty acids (SCFAs) like butyrate and bile acids (BAs), herein we elucidate their significant contributions to ocular pathologies, thought-provoking the traditional belief of organ sterility, particularly in the field of ophthalmology. Highlighting the dynamic nature of the gut microbiota and its profound influence on ocular health, this review underlines the necessity of comprehending the complex workings of the gut-eye axis, an emerging field of science ready for further exploration and scrutiny. While acknowledging the therapeutic promise in manipulating the gut microbiome and its metabolites, the available literature advocates for a targeted, precise approach. Instead of broad interventions, it emphasizes the potential of exploiting specific microbiome-related metabolites as a focused strategy. This targeted approach compared to a precision tool rather than a broad-spectrum solution, aims to explore the therapeutic applications of microbiome-related metabolites in the context of various retinal diseases. By proposing a nuanced strategy targeted at specific microbial metabolites, this review suggests that addressing specific deficiencies or imbalances through microbiome-related metabolites might yield expedited and pronounced outcomes in systemic health, extending to the eye. This focused strategy holds the potential in bypassing the irregularity associated with manipulating microbes themselves, paving a more efficient pathway toward desired outcomes in optimizing gut health and its implications for retinal diseases.
Collapse
Affiliation(s)
- Yvonne Nguyen
- Mercer University School of Medicine, Macon, GA, United States
| | | | | | - Tommy Bui
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Allston Oxenrider
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| | - Ravirajsinh N. Jadeja
- Biochemistry and Molecular Biology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Menaka C. Thounaojam
- Departments of Cellular Biology and Anatomy, Augusta University, Augusta, GA, United States
| |
Collapse
|
5
|
Li X, Lu W, Kharitonenkov A, Luo Y. Targeting the FGF19-FGFR4 pathway for cholestatic, metabolic, and cancerous diseases. J Intern Med 2024; 295:292-312. [PMID: 38212977 DOI: 10.1111/joim.13767] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Human fibroblast growth factor 19 (FGF19, or FGF15 in rodents) plays a central role in controlling bile acid (BA) synthesis through a negative feedback mechanism. This process involves a postprandial crosstalk between the BA-activated ileal farnesoid X receptor and the hepatic Klotho beta (KLB) coreceptor complexed with fibrobalst growth factor receptor 4 (FGFR4) kinase. Additionally, FGF19 regulates glucose, lipid, and energy metabolism by coordinating responses from functional KLB and FGFR1-3 receptor complexes on the periphery. Pharmacologically, native FGF19 or its analogs decrease elevated BA levels, fat content, and collateral tissue damage. This makes them effective in treating both cholestatic diseases such as primary biliary or sclerosing cholangitis (PBC or PSC) and metabolic abnormalities such as nonalcoholic steatohepatitis (NASH). However, chronic administration of FGF19 drives oncogenesis in mice by activating the FGFR4-dependent mitogenic or hepatic regenerative pathway, which could be a concern in humans. Agents that block FGF19 or FGFR4 signaling have shown great potency in preventing FGF19-responsive hepatocellular carcinoma (HCC) development in animal models. Recent phase 1/2 clinical trials have demonstrated promising results for several FGF19-based agents in selectively treating patients with PBC, PSC, NASH, or HCC. This review aims to provide an update on the clinical development of both analogs and antagonists targeting the FGF19-FGFR4 signaling pathway for patients with cholestatic, metabolic, and cancer diseases. We will also analyze potential safety and mechanistic concerns that should guide future research and advanced trials.
Collapse
Affiliation(s)
- Xiaokun Li
- School of Pharmacological Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiqin Lu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, El Paso, Texas, USA
| | | | - Yongde Luo
- School of Pharmacological Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
6
|
Wang J, de Bruijn V, Rietjens IM, Kramer NI, Bouwmeester H. Use of Physiologically Based Kinetic Modeling to Predict Deoxynivalenol Metabolism and Its Role in Intestinal Inflammation and Bile Acid Kinetics in Humans. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:761-772. [PMID: 38131302 PMCID: PMC10786035 DOI: 10.1021/acs.jafc.3c07137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/03/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Current points of departure used to derive health-based guidance values for deoxynivalenol (DON) are based on studies in laboratory animals. Here, an animal-free testing approach was adopted in which a reverse dosimetry physiologically based kinetic (PBK) modeling is used to predict in vivo dose response curves for DON's effects on intestinal pro-inflammatory cytokine secretion and intestinal bile acid reabsorption in humans from concentration-effect relationships for DON in vitro. The calculated doses for inducing a 5% added effect above the background level (ED5) of DON for increasing IL-1β secretion in intestinal tissue and for increasing the amounts in the colon lumen of glycochenodeoxycholic acid (GCDCA) were 246 and 36 μg/kg bw/day, respectively. These in vitro-in silico-derived ED5 values were compared to human dietary DON exposure levels, indicating that the risk of DON's effects on these end points occurring in various human populations cannot be excluded. This in vitro-in silico approach provides a novel testing strategy for hazard and risk assessment without using laboratory animals.
Collapse
Affiliation(s)
- Jingxuan Wang
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, Netherlands
| | - Veronique de Bruijn
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, Netherlands
| | - Ivonne M.C.M. Rietjens
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, Netherlands
| | - Nynke I. Kramer
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, Netherlands
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University and Research, Stippeneng 4, 6708 WE, Wageningen, Netherlands
| |
Collapse
|
7
|
Tecos ME, Steinberger AE, Guo J, Rubin DC, Davidson NO, Warner BW. Roles for Bile Acid Signaling and Nonsense-Mediated Ribonucleic Acid Decay in Small Bowel Resection-Associated Liver Injury. J Surg Res 2024; 293:433-442. [PMID: 37812877 DOI: 10.1016/j.jss.2023.09.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 08/19/2023] [Accepted: 09/13/2023] [Indexed: 10/11/2023]
Abstract
INTRODUCTION Massive intestinal loss resulting in short bowel syndrome has been linked to intestinal failure associated liver disease. Efforts to elucidate the driving force behind the observed hepatic injury have identified inflammatory mediators, alterations in the microbiome, extent of structural and functional intestinal adaptation, and toxic shifts in the bile acid pool. In the present study, we posit that ileocecal resection interrupts the delivery of these hepatotoxic substances to the liver by physically disrupting the enterohepatic circulation, thereby shielding the liver from exposure to the aforementioned noxious stimuli. METHODS Mice underwent sham, 50% proximal, or 50% distal small bowel resection (SBR), with or without tauroursodeoxycolic acid supplementation. Enterohepatic signaling and nonsense-mediated ribonucleic acid (RNA) decay were evaluated and correlated with hepatic injury. RESULTS When compared to 50% proximal SBR, mice that underwent ileocecal resection exhibited reduced hepatic oxidative stress and exhibited a more physiological bile acid profile with increased de novo bile acid synthesis, enhanced colonic bile acid signaling, and reduced hepatic proliferation. Distal intestinal resection promoted an adaptive response including via the nonsense-mediated RNA decay pathway to satisfactorily process injurious messenger RNA and successfully maintain homeostasis. By contrast, this adaptive response was not observed in the proximal SBR group and hepatic injury persisted. CONCLUSIONS In summary, interruption of enterohepatic circulation via ileocecal resection abrogates the liver's exposure to toxic and inflammatory mediators while promoting physiological adaptations in bile acid metabolism and maintaining existing homeostatic pathways.
Collapse
Affiliation(s)
- Maria E Tecos
- Division of General Surgery, Department of Surgery, University of Nebraska Medical Center, Omaha, Nebraska
| | - Allie E Steinberger
- Department of Surgery, Barnes Jewish Hospital, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Jun Guo
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children's Hospital, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Deborah C Rubin
- Division of Gastroenterology, Department of Medicine, Washington University in St. Louis School of Medicine, Washington University, St. Louis, Missouri
| | - Nicholas O Davidson
- Division of Gastroenterology, Department of Medicine, Washington University in St. Louis School of Medicine, Washington University, St. Louis, Missouri
| | - Brad W Warner
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children's Hospital, Washington University in St. Louis School of Medicine, St. Louis, Missouri.
| |
Collapse
|
8
|
Pan T, Li X, Guo X, Wang H, Zhou X, Shang R, Xie D, Qian X, Dai M, Fan E, Chen X, Chen C. Electroacupuncture Improves Insulin Resistance in Type 2 Diabetes Mice by Regulating Intestinal Flora and Bile Acid. Diabetes Metab Syndr Obes 2023; 16:4025-4042. [PMID: 38089431 PMCID: PMC10712683 DOI: 10.2147/dmso.s421134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 11/02/2023] [Indexed: 01/21/2025] Open
Abstract
INTRODUCTION Adjusting internal organs and dredging channel electroacupuncture has a definite effect on type 2 diabetes, but the specific mechanism still needs to be further clarified. This study aims to investigate the effects of electroacupuncture on the gut microbiota and bile acids in db/db mice after the intervention of "adjusting internal organs and dredging channel" and further explore its mechanism of action in treating T2DM. METHODS We used db/db mice as the animal model and db/m mice from the same litter as the blank control group, a total of 4 weeks of intervention were conducted. We evaluated the effectiveness of the "adjusting internal organs and dredging channel" treatment by detecting indicators related to glucose and lipid- metabolism. Detect changes in the gut microbiota of mice in each group using 16SrDNA sequencing technology. The content of bile acids in mouse feces was determined using liquid chromatography mass spectrometry, and the correlation analysis between different bile acids and differential bacterial communities was performed. The expression levels of TGR5 and GLP-1 proteins were measured using the Western blot method. RESULTS Adjusting internal organs and dredging channel electroacupuncture can improve blood glucose levels in db/db mice, increase the abundance of Firmicutes and Actinobacteria, and increase the content of fecal bile acid pool heavy CA and UDCA. At the same time, it also increased the content of TGR5/GLP1 in the small intestine. CONCLUSION Adjusting internal organs and dredging channel electroacupuncture can improve the disorder of glucose and lipid metabolism in db/db mice, regulate the abundance and colony composition of intestinal microbiota in mice, and regulate bile acid metabolism in mice. The interaction between bile acid and intestinal microbiota can also be observed; Mutual influence may play a role in regulating blood sugar together.
Collapse
Affiliation(s)
- Ting Pan
- College of Traditional Chinese Medicine, ChangChun University of Chinese Medicine, Changchun, Jilin, People’s Republic of China
| | - Xuefeng Li
- School of Acupuncture-Moxibustion and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, People’s Republic of China
| | - Xiaole Guo
- Prevention and Treatment Center, The Third Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, People’s Republic of China
| | - Haili Wang
- School of Acupuncture-Moxibustion and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, People’s Republic of China
| | - Xue Zhou
- School of Acupuncture-Moxibustion and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, People’s Republic of China
| | - Rui Shang
- Traditional Chinese Medicine Rehabilitation Center, Jilin Cancer Hospital, Changchun, Jilin, People’s Republic of China
| | - Donge Xie
- Acupuncture and moxibustion Clinical Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, People’s Republic of China
| | - Xin Qian
- Tuina Department, Shenzhen Traditional Chinese Medicine Hospital, ShenZhen, Guangdong, People’s Republic of China
| | - Mengyao Dai
- School of Acupuncture-Moxibustion and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, People’s Republic of China
| | - Enshuo Fan
- School of Acupuncture-Moxibustion and Tuina, Changchun University of Chinese Medicine, Changchun, Jilin, People’s Republic of China
| | - Xinhua Chen
- College of Traditional Chinese Medicine, ChangChun University of Chinese Medicine, Changchun, Jilin, People’s Republic of China
| | - Chunhai Chen
- Acupuncture and moxibustion Clinical Center, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, People’s Republic of China
| |
Collapse
|
9
|
Sweed D, Holah NS, Karman E, Asaad N, Mahmoud S. Farnesoid X receptor in chronic liver diseases: an immunohistochemical study. J Immunoassay Immunochem 2023; 44:381-395. [PMID: 37665366 DOI: 10.1080/15321819.2023.2242920] [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] [Indexed: 09/05/2023]
Abstract
Chronic hepatitis C virus (HCV) related liver diseases are still an ongoing cause of hepatic failure despite the effective role of direct-acting anti-viral agents. Farnesoid X receptor (FXR) agonists have a potential therapeutic effect on the management of chronic liver diseases (CLD). However, data regarding FXR protein expression in human CLDs are limited and conflicting. We aimed to assess the immunohistochemical expression of FXR in HCV-related chronic hepatitis and cirrhosis in comparison with metabolic-associated fatty liver disease (MAFLD) and normal liver tissue. The expression of FXR was low both in hepatocytes and bile ducts of HCV-related chronic hepatitis and cirrhosis (p = .001, respectively). In addition, a significantly low expression of FXR was observed in HCV-related hepatitis and cirrhosis groups compared to MAFLD in hepatocytes (p < .001, for both) and bile ducts (p = .004 and p = .018). FXR expression in HCV-related cirrhosis was significantly associated with compensated liver function (p = .032) and low inflammatory activity (p = .022). FXR expression decreases in HCV-related CLDs. There was some evidence that FXR expression could protect against post-hepatitis cirrhosis.
Collapse
Affiliation(s)
- Dina Sweed
- Pathology Department, National Liver Institute, Menofia University, Shebin Elkom, Egypt
| | - Nanis Shawky Holah
- Pathology Department, Faculty of Medicine, Menofia University, Shebin Elkom, Egypt
| | - Esraa Karman
- Pathology Department, National Liver Institute, Menofia University, Shebin Elkom, Egypt
| | - Nancy Asaad
- Pathology Department, Faculty of Medicine, Menofia University, Shebin Elkom, Egypt
| | - Shereen Mahmoud
- Pathology Department, Faculty of Medicine, Menofia University, Shebin Elkom, Egypt
| |
Collapse
|
10
|
Olivença DV, Davis JD, Kumbale CM, Zhao CY, Brown SP, McCarty NA, Voit EO. Mathematical models of cystic fibrosis as a systemic disease. WIREs Mech Dis 2023; 15:e1625. [PMID: 37544654 PMCID: PMC10843793 DOI: 10.1002/wsbm.1625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 06/22/2023] [Accepted: 07/06/2023] [Indexed: 08/08/2023]
Abstract
Cystic fibrosis (CF) is widely known as a disease of the lung, even though it is in truth a systemic disease, whose symptoms typically manifest in gastrointestinal dysfunction first. CF ultimately impairs not only the pancreas and intestine but also the lungs, gonads, liver, kidneys, bones, and the cardiovascular system. It is caused by one of several mutations in the gene of the epithelial ion channel protein CFTR. Intense research and improved antimicrobial treatments during the past eight decades have steadily increased the predicted life expectancy of a person with CF (pwCF) from a few weeks to over 50 years. Moreover, several drugs ameliorating the sequelae of the disease have become available in recent years, and notable treatments of the root cause of the disease have recently generated substantial improvements in health for some but not all pwCF. Yet, numerous fundamental questions remain unanswered. Complicating CF, for instance in the lung, is the fact that the associated insufficient chloride secretion typically perturbs the electrochemical balance across epithelia and, in the airways, leads to the accumulation of thick, viscous mucus and mucus plaques that cannot be cleared effectively and provide a rich breeding ground for a spectrum of bacterial and fungal communities. The subsequent infections often become chronic and respond poorly to antibiotic treatments, with outcomes sometimes only weakly correlated with the drug susceptibility of the target pathogen. Furthermore, in contrast to rapidly resolved acute infections with a single target pathogen, chronic infections commonly involve multi-species bacterial communities, called "infection microbiomes," that develop their own ecological and evolutionary dynamics. It is presently impossible to devise mathematical models of CF in its entirety, but it is feasible to design models for many of the distinct drivers of the disease. Building upon these growing yet isolated modeling efforts, we discuss in the following the feasibility of a multi-scale modeling framework, known as template-and-anchor modeling, that allows the gradual integration of refined sub-models with different granularity. The article first reviews the most important biomedical aspects of CF and subsequently describes mathematical modeling approaches that already exist or have the potential to deepen our understanding of the multitude aspects of the disease and their interrelationships. The conceptual ideas behind the approaches proposed here do not only pertain to CF but are translatable to other systemic diseases. This article is categorized under: Congenital Diseases > Computational Models.
Collapse
Affiliation(s)
- Daniel V. Olivença
- Center for Engineering Innovation, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, Texas 75080, USA
| | - Jacob D. Davis
- Department of Biomedical Engineering, Georgia Tech and Emory University, Atlanta, Georgia
| | - Carla M. Kumbale
- Department of Biomedical Engineering, Georgia Tech and Emory University, Atlanta, Georgia
| | - Conan Y. Zhao
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Samuel P. Brown
- Department of Biological Sciences, Georgia Tech and Emory University, Atlanta, Georgia
| | - Nael A. McCarty
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Eberhard O. Voit
- Department of Biomedical Engineering, Georgia Tech and Emory University, Atlanta, Georgia
| |
Collapse
|
11
|
Tecos ME, Steinberger AE, Guo J, Rubin DC, Davidson NO, Warner BW. Disruption of Enterohepatic Circulation of Bile Acids Ameliorates Small Bowel Resection Associated Hepatic Injury. J Pediatr Surg 2023; 58:1074-1078. [PMID: 36914459 PMCID: PMC10355217 DOI: 10.1016/j.jpedsurg.2023.02.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
BACKGROUND Massive small bowel resection (SBR) is associated with liver injury and fibrosis. Efforts to elucidate the driving force behind hepatic injury have identified multiple factors, including the generation of toxic bile acid metabolites. METHODS Sham, 50% proximal, and 50% distal SBR were carried out in C57BL/6 mice to determine the effect of jejunal (proximal SBR) versus ileocecal resection (distal SBR) on bile acid metabolism and liver injury. Tissues were harvested at 2 and 10-week postoperative timepoints. RESULTS When compared with 50% proximal SBR, mice that underwent distal SBR exhibited less hepatic oxidative stress as verified by decreased mRNA expression of tumor necrosis factor-α (TNFα, p ≤ 0.0001), nicotinamide adenine dinucleotide phosphate oxidase (NOX, p ≤ 0.0001), and glutathione synthetase (GSS, p ≤ 0.05). Distal SBR mice also exhibited a more hydrophilic bile acid profile with reduced abundance of insoluble bile acids (cholic acid (CA), taurodeoxycholic acid (TCA), and taurolithocholic acid (TLCA)), and increased abundance of soluble bile acids (tauroursodeoxycholic acid (TUDCA)). In contrast with proximal SBR, ileocecal resection alters enterohepatic circulation leading to reduced oxidative stress and promotes physiological bile acid metabolism. CONCLUSION These findings challenge the notion that preservation of the ileocecal region is beneficial in patients with short bowel syndrome. Administration of selected bile acids may present potential therapy to mitigate resection-associated liver injury. LEVEL OF EVIDENCE III-Case-Control Study.
Collapse
Affiliation(s)
- Maria E Tecos
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children's Hospital, Washington University in St. Louis School of Medicine, One Children's Place, Suite 6110 St. Louis, MO, 63110, USA
| | - Allie E Steinberger
- Department of Surgery, Barnes Jewish Hospital, Washington University in St. Louis School of Medicine, 9901 Wohl Hospital, Campus Box 8109, St. Louis, MO, 63110, USA
| | - Jun Guo
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children's Hospital, Washington University in St. Louis School of Medicine, One Children's Place, Suite 6110 St. Louis, MO, 63110, USA
| | - Deborah C Rubin
- Division of Gastroenterology, Department of Medicine, Washington University in St. Louis School of Medicine, Washington University, Campus Box 8124, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Nicholas O Davidson
- Division of Gastroenterology, Department of Medicine, Washington University in St. Louis School of Medicine, Washington University, Campus Box 8124, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Brad W Warner
- Division of Pediatric Surgery, Department of Surgery, St. Louis Children's Hospital, Washington University in St. Louis School of Medicine, One Children's Place, Suite 6110 St. Louis, MO, 63110, USA.
| |
Collapse
|
12
|
Bebelman MP, Bovyn MJ, Mayer CM, Delpierre J, Naumann R, Martins NP, Honigmann A, Kalaidzidis Y, Haas PA, Zerial M. Hepatocyte apical bulkheads provide a mechanical means to oppose bile pressure. J Cell Biol 2023; 222:213840. [PMID: 36716168 PMCID: PMC9930133 DOI: 10.1083/jcb.202208002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 12/15/2022] [Accepted: 01/13/2023] [Indexed: 01/31/2023] Open
Abstract
Hepatocytes grow their apical surfaces anisotropically to generate a 3D network of bile canaliculi (BC). BC elongation is ensured by apical bulkheads, membrane extensions that traverse the lumen and connect juxtaposed hepatocytes. We hypothesize that apical bulkheads are mechanical elements that shape the BC lumen in liver development but also counteract elevated biliary pressure. Here, by resolving their structure using STED microscopy, we found that they are sealed by tight junction loops, connected by adherens junctions, and contain contractile actomyosin, characteristics of mechanical function. Apical bulkheads persist at high pressure upon microinjection of fluid into the BC lumen, and laser ablation demonstrated that they are under tension. A mechanical model based on ablation results revealed that apical bulkheads double the pressure BC can hold. Apical bulkhead frequency anticorrelates with BC connectivity during mouse liver development, consistent with predicted changes in biliary pressure. Our findings demonstrate that apical bulkheads are load-bearing mechanical elements that could protect the BC network against elevated pressure.
Collapse
Affiliation(s)
- Maarten P. Bebelman
- https://ror.org/05b8d3w18Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Matthew J. Bovyn
- https://ror.org/05b8d3w18Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany,Center for Systems Biology Dresden, Dresden, Germany,Max Planck Institute for the Physics of Complex Systems, Dresden, Germany
| | - Carlotta M. Mayer
- https://ror.org/05b8d3w18Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Julien Delpierre
- https://ror.org/05b8d3w18Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ronald Naumann
- https://ror.org/05b8d3w18Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Nuno P. Martins
- https://ror.org/05b8d3w18Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Alf Honigmann
- https://ror.org/05b8d3w18Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Yannis Kalaidzidis
- https://ror.org/05b8d3w18Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Pierre A. Haas
- https://ror.org/05b8d3w18Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany,Center for Systems Biology Dresden, Dresden, Germany,Max Planck Institute for the Physics of Complex Systems, Dresden, Germany,Pierre A. Haas:
| | - Marino Zerial
- https://ror.org/05b8d3w18Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany,Center for Systems Biology Dresden, Dresden, Germany,Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany,Correspondence to Marino Zerial:
| |
Collapse
|
13
|
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: 1.0] [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
|
14
|
de Bruijn VMP, Rietjens IMCM, Bouwmeester H. Population pharmacokinetic model to generate mechanistic insights in bile acid homeostasis and drug-induced cholestasis. Arch Toxicol 2022; 96:2717-2730. [PMID: 35876888 PMCID: PMC9352636 DOI: 10.1007/s00204-022-03345-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/14/2022] [Indexed: 12/05/2022]
Abstract
Bile acids (BA) fulfill a wide range of physiological functions, but are also involved in pathologies, such as cholestasis. Cholestasis is characterized by an intrahepatic accumulation of BAs and subsequent spillage to the systemic circulation. The aim of the present study was to develop physiologically based kinetic (PBK) models that would provide a tool to predict dose-dependent BA accumulation in humans upon treatment with a Bile Salt Export Pump (BSEP) inhibitor. We developed a PBK model describing the BA homeostasis using glycochenodeoxycholic acid as an exemplary BA. Population wide distributions of BSEP abundances were incorporated in the PBK model using Markov Chain Monte Carlo simulations, and alternatively the total amount of BAs was scaled empirically to describe interindividual differences in plasma BA levels. Next, the effects of the BSEP inhibitor bosentan on the BA levels were simulated. The PBK model developed adequately predicted the in vivo BA dynamics. Both the Markov Chain Monte Carlo simulations based on a distribution of BSEP abundances and empirical scaling of the total BA pool readily described the variations within and between data in human volunteers. Bosentan treatment disproportionally increased the maximum BA concentration in individuals with a large total BA pool or low BSEP abundance. Especially individuals having a large total BA pool size and a low BSEP abundance were predicted to be at risk for rapid saturation of BSEP and subsequent intrahepatic BA accumulation. This model provides a first estimate of personalized safe therapeutic external dose levels of compounds with BSEP-inhibitory properties.
Collapse
Affiliation(s)
- Véronique M P de Bruijn
- Division of Toxicology, Wageningen University and Research, Wageningen, 6708 WE, The Netherlands.
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, Wageningen, 6708 WE, The Netherlands
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University and Research, Wageningen, 6708 WE, The Netherlands
| |
Collapse
|
15
|
Gu Y, Li L, Yang M, Liu T, Song X, Qin X, Xu X, Liu J, Wang B, Cao H. Bile acid-gut microbiota crosstalk in irritable bowel syndrome. Crit Rev Microbiol 2022; 49:350-369. [PMID: 35389754 DOI: 10.1080/1040841x.2022.2058353] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Irritable bowel syndrome (IBS) is a common disorder of gut-brain interaction with an increasing prevalence, and its precise aetiology remains unclear. Gut microbiota dysbiosis has been found to be associated with IBS pathogenesis. In addition, a high incidence of bile acid diarrhoea and disturbed bile acid metabolism has been observed in IBS patients. The abundant microorganisms inhabited in human gut have essential functions in bile acid biotransformation, and can immensely affect the size and constitution of bile acid pool. Meanwhile, the alterations of bile acid profile can inversely interfere with the gut microbiota. This review discussed the role of intricate correlations between bile acids and gut microbiota in IBS pathogenesis and delineated the possible molecular mechanisms, mainly the signalling induced by farnesoid X receptor and transmembrane G protein-coupled receptor 5. Besides, some biomarkers for identifying bile acid diarrhoea in IBS population were listed, assisting the diagnosis and classification of IBS. Moreover, it also assessed some therapeutic strategies for IBS that regulate the bile acid-gut microbiota axis, such as dietary modulation, probiotics/prebiotics, faecal microbiota transplantation, and antibiotics. Collectively, this article illustrated the relationship between bile acids and gut microbiota in IBS pathophysiology and might offer some novel therapeutic options for IBS.
Collapse
Affiliation(s)
- Yu Gu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lingfeng Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Min Yang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xueli Song
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiali Qin
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Xu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinghua Liu
- Department of Gastroenterology, Tianjin TEDA hospital, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
16
|
Ju Z, Shen L, Zhou M, Luo J, Yu Z, Qu C, Lei R, Lei M, Huang R. Helicobacter pylori and Alzheimer's Disease-Related Metabolic Dysfunction: Activation of TLR4/Myd88 Inflammation Pathway from p53 Perspective and a Case Study of Low-Dose Radiation Intervention. ACS Chem Neurosci 2022; 13:1065-1081. [PMID: 35312296 DOI: 10.1021/acschemneuro.2c00082] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Gut dysbiosis is observed in Alzheimer's disease (AD) and is frequently associated with AD-induced metabolic dysfunction. However, the extent and specific underlying molecular mechanisms triggered by alterations of gut microbiota composition and function mediating AD-induced metabolic dysfunction in AD remain incompletely uncovered. Here, we indicate that Helicobacter pylori (H. pylori) is abundant in AD patients with relative metabolic dysfunction. Fecal microbiota transplantation from the AD patients promoted metabolic dysfunction in mice and increased gut permeability. H. pylori increased gut permeability through activation of the TLR4/Myd88 inflammation pathway in a p53-dependent manner, leading to metabolic dysfunction. Moreover, p53 deficiency reduced bile acid concentration, leading to an increased abundance of H. pylori colonization. Overall, these data identify H. pylori as a key promoter of AD-induced metabolic dysfunction.
Collapse
Affiliation(s)
- Zhao Ju
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Meiling Zhou
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Jinhua Luo
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Zijian Yu
- The First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, Hunan 421001, People’s Republic of China
| | - Can Qu
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Ridan Lei
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| | - Mingjun Lei
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Ruixue Huang
- Department of Occupational and Environmental Health, Xiangya School of Public Health, Central South University, Changsha, Hunan 410078, China
| |
Collapse
|
17
|
Gillard J, Clerbaux LA, Nachit M, Sempoux C, Staels B, Bindels LB, Tailleux A, Leclercq IA. Bile acids contribute to the development of non-alcoholic steatohepatitis in mice. JHEP REPORTS : INNOVATION IN HEPATOLOGY 2021; 4:100387. [PMID: 34825156 PMCID: PMC8604813 DOI: 10.1016/j.jhepr.2021.100387] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 09/20/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023]
Abstract
Background & Aims Through FXR and TGR5 signaling, bile acids (BAs) modulate lipid and glucose metabolism, inflammation and fibrosis. Hence, BAs returning to the liver after enteric secretion, modification and reabsorption may contribute to the pathogenesis of non-alcoholic steatohepatitis (NASH). Herein, we characterized the enterohepatic profile and signaling of BAs in preclinical models of NASH, and explored the consequences of experimental manipulation of BA composition. Methods We used high-fat diet (HFD)-fed foz/foz and high-fructose western diet-fed C57BL/6J mice, and compared them to their respective controls. Mice received a diet supplemented with deoxycholic acid (DCA) to modulate BA composition. Results Compared to controls, mice with NASH had lower concentrations of BAs in their portal blood and bile, while systemic BA concentrations were not significantly altered. Notably, the concentrations of secondary BAs, and especially of DCA, and the ratio of secondary to primary BAs were strikingly lower in bile and portal blood of mice with NASH. Hence, portal blood was poor in FXR and TGR5 ligands, and conferred poor anti-inflammatory protection in mice with NASH. Enhanced primary BAs synthesis and conversion of secondary to primary BAs in NASH livers contributed to the depletion in secondary BAs. Dietary DCA supplementation in HFD-fed foz/foz mice restored the BA concentrations in portal blood, increased TGR5 and FXR signaling, improved the dysmetabolic status, protected from steatosis and hepatocellular ballooning, and reduced macrophage infiltration. Conclusions BA composition in the enterohepatic cycle, but not in systemic circulation, is profoundly altered in preclinical models of NASH, with specific depletion in secondary BAs. Dietary correction of the BA profile protected from NASH, supporting a role for enterohepatic BAs in the pathogenesis of NASH. Lay summary This study clearly demonstrates that the alterations of enterohepatic bile acids significantly contribute to the development of non-alcoholic steatohepatitis in relevant preclinical models. Indeed, experimental modulation of bile acid composition restored perturbed FXR and TGR5 signaling and prevented non-alcoholic steatohepatitis and associated metabolic disorders.
Collapse
Key Words
- ASBT, apical sodium-dependent BA transporter
- BA, bile acid
- CA, cholic acid
- CDCA, chenodeoxycholic acid
- CYP27A1, sterol 27-hydroxylase
- CYP2A12, bile acid 7α-hydroxylase
- CYP7A1, cholesterol 7α-hydroxylase
- CYP7B1, oxysterol 7α-hydroxylase
- CYP8B1, sterol 12α-hydroxylase
- DCA, deoxycholic acid
- FABP6, fatty acid binding protein 6
- FGF15, fibroblast growth factor 15
- FGFR4, fibroblast growth factor receptor 4
- FXR
- FXR, Farnesoid X receptor
- GLP-1, glucagon-like peptide-1
- HFD, high-fat diet
- LCA, lithocholic acid
- LPS, lipopolysaccharide
- NAFLD
- NAFLD, non-alcoholic fatty liver disease
- NAS, NAFLD activity score
- NASH
- NASH, non-alcoholic steatohepatitis
- ND, normal diet
- OGTT, oral glucose tolerance test
- OST, organic solute transporter
- SHP, small heterodimer protein
- TGR5
- TGR5, Takeda G-protein coupled receptor 5
- TLCA, tauro-lithocholic acid
- TNFα, tumor necrosis factor α
- WDF, western and high-fructose diet
- WT, wild-type
- metabolic syndrome
- αMCA, α-muricholic acid
- βMCA, β-muricholic acid
- ωMCA, ω-muricholic acid
Collapse
Affiliation(s)
- Justine Gillard
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium.,Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Laure-Alix Clerbaux
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Maxime Nachit
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| | - Christine Sempoux
- Institute of Pathology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Laure B Bindels
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Anne Tailleux
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Isabelle A Leclercq
- Laboratory of Hepato-Gastroenterology, Institute of Experimental and Clinical Research, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
18
|
Chong Nguyen C, Duboc D, Rainteau D, Sokol H, Humbert L, Seksik P, Bellino A, Abdoul H, Bouazza N, Treluyer JM, Saadi M, Wahbi K, Soliman H, Coffin B, Bado A, Le Gall M, Varenne O, Duboc H. Circulating bile acids concentration is predictive of coronary artery disease in human. Sci Rep 2021; 11:22661. [PMID: 34811445 PMCID: PMC8608912 DOI: 10.1038/s41598-021-02144-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 11/09/2021] [Indexed: 12/14/2022] Open
Abstract
Synthetized by the liver and metabolized by the gut microbiota, BA are involved in metabolic liver diseases that are associated with cardiovascular disorders. Animal models of atheroma documented a powerful anti-atherosclerotic effect of bile acids (BA). This prospective study examined whether variations in circulating BA are predictive of coronary artery disease (CAD) in human. Consecutive patients undergoing coronary angiography were enrolled. Circulating and fecal BA were measured by high pressure liquid chromatography and tandem mass spectrometry. Of 406 screened patients, 80 were prospectively included and divided in two groups with (n = 45) and without (n = 35) CAD. The mean serum concentration of total BA was twice lower in patients with, versus without CAD (P = 0.005). Adjusted for gender and age, this decrease was an independent predictor of CAD. In a subgroup of 17 patients, statin therapy doubled the serum BA concentration. Decreased serum concentrations of BA were predictors of CAD in humans. A subgroup analysis showed a possible correction by statins. With respect to the anti-atherosclerotic effect of BA in animal models, and their role in human lipid metabolism, this study describe a new metabolic disturbance associated to CAD in human.
Collapse
Affiliation(s)
- Caroline Chong Nguyen
- Centre de Recherche Sur I'inflammation, Inserm, UMR 1149, Université de Paris, 75018, Paris, France.,Cardiology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, 75014, Paris, France.,Department of Hepato Gastro Enterology and University of Paris, Louis Mourier Hospital, APHP, 92700, Colombes, France.,Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, 75012, Paris, France
| | - Denis Duboc
- Cardiology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, 75014, Paris, France
| | - Dominique Rainteau
- Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, 75012, Paris, France
| | - Harry Sokol
- Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, 75012, Paris, France
| | - Lydie Humbert
- Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, 75012, Paris, France
| | - Philippe Seksik
- Inserm, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Hôpital Saint Antoine, Service de Gastroentérologie, Sorbonne Université, 75012, Paris, France
| | - Adèle Bellino
- Unite de Recherche Clinique-Centre Dinvestigation Clinique Necker/Cochin, Hôpital Tarnier, Université de Paris, 75006, Paris, France
| | - Hendy Abdoul
- Unite de Recherche Clinique-Centre Dinvestigation Clinique Necker/Cochin, Hôpital Tarnier, Université de Paris, 75006, Paris, France
| | - Naïm Bouazza
- Unite de Recherche Clinique-Centre Dinvestigation Clinique Necker/Cochin, Hôpital Tarnier, Université de Paris, 75006, Paris, France
| | - Jean-Marc Treluyer
- Unite de Recherche Clinique-Centre Dinvestigation Clinique Necker/Cochin, Hôpital Tarnier, Université de Paris, 75006, Paris, France
| | - Malika Saadi
- Cardiology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, 75014, Paris, France
| | - Karim Wahbi
- Cardiology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, 75014, Paris, France
| | - Heithem Soliman
- Department of Hepato Gastro Enterology and University of Paris, Louis Mourier Hospital, APHP, 92700, Colombes, France
| | - Benoit Coffin
- Centre de Recherche Sur I'inflammation, Inserm, UMR 1149, Université de Paris, 75018, Paris, France.,Department of Hepato Gastro Enterology and University of Paris, Louis Mourier Hospital, APHP, 92700, Colombes, France
| | - André Bado
- Centre de Recherche Sur I'inflammation, Inserm, UMR 1149, Université de Paris, 75018, Paris, France
| | - Maude Le Gall
- Centre de Recherche Sur I'inflammation, Inserm, UMR 1149, Université de Paris, 75018, Paris, France
| | - Olivier Varenne
- Cardiology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, 75014, Paris, France
| | - Henri Duboc
- Centre de Recherche Sur I'inflammation, Inserm, UMR 1149, Université de Paris, 75018, Paris, France. .,Cardiology Department, Cochin Hospital, Assistance Publique-Hôpitaux de Paris, Université de Paris, 75014, Paris, France. .,Department of Hepato Gastro Enterology and University of Paris, Louis Mourier Hospital, APHP, 92700, Colombes, France. .,INSERM UMRS 1149, Université de Paris, 16 rue Henri Huchard, 75890, Paris Cedex 18, France.
| |
Collapse
|
19
|
Abstract
Phenotypic heterogeneity among single cells in a genetically identical population leads to diverse environmental adaptation. The human and animal pathogen Salmonella enterica serovar Typhimurium exhibits heterogeneous expression of virulence genes, including flagellar and Salmonella pathogenicity island (SPI) genes. Little is known about how the differential expression of flagellar genes among single cells affects bacterial adaptation to stresses. Here, we have developed a triple-fluorescence reporter to simultaneously monitor the expression of flagellar and SPI-1 pathways. We show that the two pathways cross talk at the single-cell level. Intriguingly, cells expressing flagella (fliC-ON) exhibit decreased tolerance to antibiotics compared to fliC-OFF cells. Such variation depends on TolC-dependent efflux pumps. We further show that fliC-ON cells contain higher intracellular proton concentrations. This suggests that the assembly and rotation of flagella consume the proton motive force and decrease the efflux activity, resulting in antibiotic sensitivity. Such a trade-off between motility and efflux highlights a novel mechanism of antibiotic tolerance.
Collapse
|
20
|
Sauerbruch T, Hennenberg M, Trebicka J, Beuers U. Bile Acids, Liver Cirrhosis, and Extrahepatic Vascular Dysfunction. Front Physiol 2021; 12:718783. [PMID: 34393832 PMCID: PMC8358446 DOI: 10.3389/fphys.2021.718783] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022] Open
Abstract
The bile acid pool with its individual bile acids (BA) is modulated in the enterohepatic circulation by the liver as the primary site of synthesis, the motility of the gallbladder and of the intestinal tract, as well as by bacterial enzymes in the intestine. The nuclear receptor farnesoid X receptor (FXR) and Gpbar1 (TGR5) are important set screws in this process. Bile acids have a vasodilatory effect, at least according to in vitro studies. The present review examines the question of the extent to which the increase in bile acids in plasma could be responsible for the hyperdynamic circulatory disturbance of liver cirrhosis and whether modulation of the bile acid pool, for example, via administration of ursodeoxycholic acid (UDCA) or via modulation of the dysbiosis present in liver cirrhosis could influence the hemodynamic disorder of liver cirrhosis. According to our analysis, the evidence for this is limited. Long-term studies on this question are lacking.
Collapse
Affiliation(s)
- Tilman Sauerbruch
- Department of Internal Medicine I, University of Bonn, Bonn, Germany
| | - Martin Hennenberg
- Department of Urology I, University Hospital, LMU Munich, Munich, Germany
| | - Jonel Trebicka
- Translational Hepatology, Medical Department, University of Frankfurt, Frankfurt, Germany
| | - Ulrich Beuers
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, location AMC, Amsterdam, Netherlands
| |
Collapse
|
21
|
Shahidi N, Pan M, Safaei S, Tran K, Crampin EJ, Nickerson DP. Hierarchical semantic composition of biosimulation models using bond graphs. PLoS Comput Biol 2021; 17:e1008859. [PMID: 33983945 PMCID: PMC8148364 DOI: 10.1371/journal.pcbi.1008859] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/25/2021] [Accepted: 04/27/2021] [Indexed: 11/19/2022] Open
Abstract
Simulating complex biological and physiological systems and predicting their behaviours under different conditions remains challenging. Breaking systems into smaller and more manageable modules can address this challenge, assisting both model development and simulation. Nevertheless, existing computational models in biology and physiology are often not modular and therefore difficult to assemble into larger models. Even when this is possible, the resulting model may not be useful due to inconsistencies either with the laws of physics or the physiological behaviour of the system. Here, we propose a general methodology for composing models, combining the energy-based bond graph approach with semantics-based annotations. This approach improves model composition and ensures that a composite model is physically plausible. As an example, we demonstrate this approach to automated model composition using a model of human arterial circulation. The major benefit is that modellers can spend more time on understanding the behaviour of complex biological and physiological systems and less time wrangling with model composition.
Collapse
Affiliation(s)
- Niloofar Shahidi
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Michael Pan
- Systems Biology Laboratory, School of Mathematics and Statistics, and Department of Biomedical Engineering, University of Melbourne, Melbourne, Victoria, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Faculty of Engineering and Information Technology, University of Melbourne, Melbourne, Victoria, Australia
| | - Soroush Safaei
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Kenneth Tran
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Edmund J. Crampin
- Systems Biology Laboratory, School of Mathematics and Statistics, and Department of Biomedical Engineering, University of Melbourne, Melbourne, Victoria, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Faculty of Engineering and Information Technology, University of Melbourne, Melbourne, Victoria, Australia
| | - David P. Nickerson
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
22
|
FXR in liver physiology: Multiple faces to regulate liver metabolism. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166133. [PMID: 33771667 DOI: 10.1016/j.bbadis.2021.166133] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/11/2021] [Accepted: 03/17/2021] [Indexed: 12/15/2022]
Abstract
The liver is the central metabolic hub which coordinates nutritional inputs and metabolic outputs. Food intake releases bile acids which can be sensed by the bile acid receptor FXR in the liver and the intestine. Hepatic and intestinal FXR coordinately regulate postprandial nutrient disposal in a network of interacting metabolic nuclear receptors. In this review we summarize and update the "classical roles" of FXR as a central integrator of the feeding state response, which orchestrates the metabolic processing of carbohydrates, lipids, proteins and bile acids. We also discuss more recent and less well studied FXR effects on amino acid, protein metabolism, autophagic turnover and inflammation. In addition, we summarize the recent understanding of how FXR signaling is affected by posttranslational modifications and by different FXR isoforms. These modifications and variations in FXR signaling might be considered when FXR is targeted pharmaceutically in clinical applications.
Collapse
|
23
|
Fiorucci S, Distrutti E, Carino A, Zampella A, Biagioli M. Bile acids and their receptors in metabolic disorders. Prog Lipid Res 2021; 82:101094. [PMID: 33636214 DOI: 10.1016/j.plipres.2021.101094] [Citation(s) in RCA: 153] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/03/2021] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
Bile acids are a large family of atypical steroids which exert their functions by binding to a family of ubiquitous cell membrane and nuclear receptors. There are two main bile acid activated receptors, FXR and GPBAR1, that are exclusively activated by bile acids, while other receptors CAR, LXRs, PXR, RORγT, S1PR2and VDR are activated by bile acids in addition to other more selective endogenous ligands. In the intestine, activation of FXR and GPBAR1 promotes the release of FGF15/19 and GLP1 which integrate their signaling with direct effects exerted by theother receptors in target tissues. This network is tuned in a time ordered manner by circadian rhythm and is critical for the regulation of metabolic process including autophagy, fast-to-feed transition, lipid and glucose metabolism, energy balance and immune responses. In the last decade FXR ligands have entered clinical trials but development of systemic FXR agonists has been proven challenging because their side effects including increased levels of cholesterol and Low Density Lipoproteins cholesterol (LDL-c) and reduced High-Density Lipoprotein cholesterol (HDL-c). In addition, pruritus has emerged as a common, dose related, side effect of FXR ligands. Intestinal-restricted FXR and GPBAR1 agonists and dual FXR/GPBAR1 agonists have been developed. Here we review the last decade in bile acids physiology and pharmacology.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy.
| | - Eleonora Distrutti
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Adriana Carino
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| | - Angela Zampella
- Department of Pharmacy, University of Napoli, Federico II, Napoli, Italy
| | - Michele Biagioli
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| |
Collapse
|
24
|
Win A, Delgado A, Jadeja RN, Martin PM, Bartoli M, Thounaojam MC. Pharmacological and Metabolic Significance of Bile Acids in Retinal Diseases. Biomolecules 2021; 11:292. [PMID: 33669313 PMCID: PMC7920062 DOI: 10.3390/biom11020292] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 12/21/2022] Open
Abstract
Bile acids (BAs) are amphipathic sterols primarily synthesized from cholesterol in the liver and released in the intestinal lumen upon food intake. BAs play important roles in micellination of dietary lipids, stimulating bile flow, promoting biliary phospholipid secretion, and regulating cholesterol synthesis and elimination. Emerging evidence, however, suggests that, aside from their conventional biological function, BAs are also important signaling molecules and therapeutic tools. In the last decade, the therapeutic applications of BAs in the treatment of ocular diseases have gained great interest. Despite the identification of BA synthesis, metabolism, and recycling in ocular tissues, much remains unknown with regards to their biological significance in the eye. Additionally, as gut microbiota directly affects the quality of circulating BAs, their analysis could derive important information on changes occurring in this microenvironment. This review aims at providing an overview of BA metabolism and biological function with a focus on their potential therapeutic and diagnostic use for retinal diseases.
Collapse
Affiliation(s)
- Alice Win
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.W.); (A.D.); (P.M.M.); (M.B.)
| | - Amanda Delgado
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.W.); (A.D.); (P.M.M.); (M.B.)
| | - Ravirajsinh N. Jadeja
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Pamela M. Martin
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.W.); (A.D.); (P.M.M.); (M.B.)
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Manuela Bartoli
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.W.); (A.D.); (P.M.M.); (M.B.)
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Menaka C. Thounaojam
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (A.W.); (A.D.); (P.M.M.); (M.B.)
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
25
|
Panzitt K, Fickert P, Wagner M. Regulation of autophagy by bile acids and in cholestasis - CholestoPHAGY or CholeSTOPagy. Biochim Biophys Acta Mol Basis Dis 2020; 1867:166017. [PMID: 33242590 DOI: 10.1016/j.bbadis.2020.166017] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/13/2020] [Accepted: 11/17/2020] [Indexed: 12/15/2022]
Abstract
Autophagy is a lysosomal degradation pathway in which the cell self-digests its own components to provide nutrients in harsh environmental conditions. It also represents an opportunity to rid the cell of superfluous and damaged organelles, misfolded proteins or invaded microorganisms. Liver autophagy contributes to basic hepatic functions such as lipid, glycogen and protein turnover. Deregulated hepatic autophagy has been linked to many liver diseases including alpha-1-antitrypsin deficiency, alcoholic and non-alcoholic fatty liver diseases, hepatitis B and C infections, liver fibrosis as well as liver cancer. Recently, bile acids and the bile acid receptor FXR have been implicated in the regulation of hepatic autophagy, which implies a role of autophagy also for cholestatic liver diseases. This review summarizes the current evidence of bile acid mediated effects on autophagy and how this affects cholestatic liver diseases. Although detailed studies are lacking, we suggest a concept that the activity of autophagy in cholestasis depends on the disease stage, where autophagy may be induced at early stages ("cholestophagy") but may be impaired in prolonged cholestatic states ("cholestopagy").
Collapse
Affiliation(s)
- Katrin Panzitt
- Research Unit for Translational Nuclear Receptor Research, Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria
| | - Peter Fickert
- Laboratory of Experimental and Molecular Hepatology, Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria
| | - Martin Wagner
- Research Unit for Translational Nuclear Receptor Research, Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
26
|
Distinct Postprandial Bile Acids Responses to a High-Calorie Diet in Men Volunteers Underscore Metabolically Healthy and Unhealthy Phenotypes. Nutrients 2020; 12:nu12113545. [PMID: 33228154 PMCID: PMC7699492 DOI: 10.3390/nu12113545] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/10/2020] [Accepted: 11/18/2020] [Indexed: 12/30/2022] Open
Abstract
Bile acids (BAs) regulate dietary lipid hydrolysis and absorption in the proximal intestine. Several studies have highlighted a determinant role of circulating levels and/or metabolism of BAs in the pathogenesis of major cardiometabolic diseases. Whether changes in BA profiles are causative or are consequence of these diseases remains to be determined. Healthy male volunteers (n = 71) underwent a postprandial exploration following consumption of a hypercaloric high fat typical Western meal providing 1200 kcal. We investigated variations of circulating levels of 28 BA species, together with BA synthesis marker 7α-hydroxy-4-cholesten-3-one (C4) over an approximately diurnal 12 h period. Analysis of BA variations during the postprandial time course revealed two major phenotypes with opposite fluctuations, i.e., circulating levels of each individual species of unconjugated BAs were reduced after meal consumption whereas those of tauro- and glyco-conjugated BAs were increased. By an unbiased classification strategy based on absolute postprandial changes in BA species levels, we classified subjects into three distinct clusters; the two extreme clusters being characterized by the smallest absolute changes in either unconjugated-BAs or conjugated-BAs. Finally, we demonstrated that our clustering based on postprandial changes in BA profiles was associated with specific clinical and biochemical features, including postprandial triglyceride levels, BMI or waist circumference. Altogether, our study reveals that postprandial profiles/patterns of BAs in response to a hypercaloric high fat challenge is associated with healthy or unhealthy metabolic phenotypes that may help in the early identification of subjects at risk of developing metabolic disorders.
Collapse
|
27
|
Chiang JY, Ferrell JM. Up to date on cholesterol 7 alpha-hydroxylase (CYP7A1) in bile acid synthesis. LIVER RESEARCH 2020; 4:47-63. [PMID: 34290896 PMCID: PMC8291349 DOI: 10.1016/j.livres.2020.05.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cholesterol 7 alpha-hydroxylase (CYP7A1, EC1.14) is the first and rate-limiting enzyme in the classic bile acid synthesis pathway. Much progress has been made in understanding the transcriptional regulation of CYP7A1 gene expression and the underlying molecular mechanisms of bile acid feedback regulation of CYP7A1 and bile acid synthesis in the last three decades. Discovery of bile acid-activated receptors and their roles in the regulation of lipid, glucose and energy metabolism have been translated to the development of bile acid-based drug therapies for the treatment of liver-related metabolic diseases such as alcoholic and non-alcoholic fatty liver diseases, liver cirrhosis, diabetes, obesity and hepatocellular carcinoma. This review will provide an update on the advances in our understanding of the molecular biology and mechanistic insights of the regulation of CYP7A1 in bile acid synthesis in the last 40 years.
Collapse
|
28
|
Hofmann AF. Quantifying the Complexities of Bile Acid Metabolism in Man: Continued Progress. Cell Mol Gastroenterol Hepatol 2020; 10:201-202. [PMID: 32240639 PMCID: PMC7296227 DOI: 10.1016/j.jcmgh.2020.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/04/2020] [Indexed: 12/10/2022]
Affiliation(s)
- Alan F. Hofmann
- Correspondence Address correspondence to: Alan F. Hofmann, MD, Department of Medicine, University of California, San Diego, La Jolla, California 92093.
| |
Collapse
|
29
|
Chiang JYL, Ferrell JM, Wu Y, Boehme S. Bile Acid and Cholesterol Metabolism in Atherosclerotic Cardiovascular Disease and Therapy. CARDIOLOGY PLUS 2020; 5:159-170. [PMID: 34350368 PMCID: PMC8330388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Dysregulation of lipid metabolism is a major factor contributing to atherosclerotic cardiovascular disease (ACVD), which is the number one cause of death in western countries. The liver plays a central role in maintaining whole body cholesterol homeostasis via catabolism of cholesterol to bile acids, as well as biliary cholesterol excretion. The liver synthesizes lipoproteins that transport dietary cholesterol and fats to muscle and adipose tissue, directs reverse cholesterol transport of excess cholesterol from extrahepatic tissues and macrophages to the liver to convert to bile acids, and thus, protects against metabolism-related nonalcoholic fatty liver disease (NAFLD) and ACVD. Liver fibrosis/nonalcoholic steatohepatitis increases the risk and prevalence of cardiovascular disease morbidity and mortality. Bile acids are signaling molecules and metabolic regulators that activate farnesoid X receptor and G protein-coupled bile acid receptor-1 to regulate lipid, glucose, and energy metabolism. The bidirectional regulation of bile acids and the gut microbiota determine the rate of bile acid synthesis, the bile acid pool size, and the composition of the circulating bile acid pool. The liver-intestine-heart axis regulates lipid metabolism, inflammation, and the pathogenesis of metabolic diseases such as ACVD, NAFLD, diabetes, and obesity. This review focuses on the roles of liver-to-intestine, liver-to-heart and intestine-to-heart axes in cholesterol, lipoprotein, and bile acid metabolism; signaling in heart health and ACVD; and drug therapies for atherosclerosis.
Collapse
Affiliation(s)
- John Y. L. Chiang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Jessica M. Ferrell
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Yue Wu
- Department of Cardiology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Shannon Boehme
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, USA
| |
Collapse
|