1
|
Wang M, Liu Y, Zhong L, Wu F, Wang J. Advancements in the investigation of gut microbiota-based strategies for stroke prevention and treatment. Front Immunol 2025; 16:1533343. [PMID: 40103814 PMCID: PMC11914130 DOI: 10.3389/fimmu.2025.1533343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Stroke represents a predominant cause of mortality and disability on a global scale, impacting millions annually and exerting a considerable strain on healthcare systems. The incidence of stroke exhibits regional variability, with ischemic stroke accounting for the majority of occurrences. Post-stroke complications, such as cognitive impairment, motor dysfunction, and recurrent stroke, profoundly affect patients' quality of life. Recent advancements have elucidated the microbiota-gut-brain axis (MGBA), underscoring the complex interplay between gut health and brain function. Dysbiosis, characterized by an imbalance in gut microbiota, is significantly linked to an elevated risk of stroke and unfavorable outcomes. The MGBA plays a crucial role in modulating immune function, neurotransmitter levels, and metabolic byproducts, which may intensify neuroinflammation and impair cerebral health. This review elucidates the role of MGBA in stroke pathophysiology and explores potential gut-targeted therapeutic strategies to reduce stroke risk and promote recovery, including probiotics, prebiotics, pharmacological interventions, and dietary modifications. However, the current prevention and treatment strategies based on intestinal flora still face many problems, such as the large difference of individual intestinal flora, the stability of efficacy, and the long-term safety need to be considered. Further research needs to be strengthened to promote its better application in clinical practice.
Collapse
Affiliation(s)
| | | | | | | | - Jinjin Wang
- Department of Gastroenterology, The First People’s Hospital of Xiaoshan District, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Matye DJ, Wang H, Wang Y, Xiong L, Li T. Bile acid sequestrant inhibits gluconeogenesis via inducing hepatic cysteine dioxygenase type 1 to reduce cysteine availability. Am J Physiol Gastrointest Liver Physiol 2025; 328:G166-G178. [PMID: 39819116 DOI: 10.1152/ajpgi.00353.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 11/28/2024] [Accepted: 12/11/2024] [Indexed: 01/19/2025]
Abstract
Bile acid sequestrants such as cholestyramine (ChTM) are gut-restricted bile acid-binding resins that block intestine bile acid absorption and attenuate hepatic bile acid signaling. Bile acid sequestrants induce hepatic bile acid synthesis to promote cholesterol catabolism and are cholesterol-lowering drugs. Bile acid sequestrants also reduce blood glucose in clinical trials and are approved drugs for treating hyperglycemia in type-2 diabetes. However, the mechanisms mediating the glucose-lowering effect of bile acid sequestrants are still incompletely understood. Here we showed that ChTM treatment decreased hepatic glucose production in Western diet-fed mice with paradoxically induced hepatic gluconeogenic genes. Cysteine dioxygenase type 1 (CDO1) mediates cysteine conversion to taurine and its expression is repressed by bile acids. We show that ChTM induced hepatic CDO1 and selectively reduced hepatic cysteine availability. Knockdown of liver CDO1 increased liver cysteine and glucose production in mice, whereas hepatocytes cultured in cystine-deficient medium showed reduced glucose production. By using dietary protein-restricted and cystine-modified Western diets that selectively alter hepatic cysteine availability, we found that reduced hepatic cysteine availability strongly inhibited glucose production in mice. Interestingly, chronic dietary protein restriction also prevented Western diet-induced obesity, which was fully reversed by restoring dietary cystine intake alone. Consistently, reduced cysteine availability dose-dependently inhibited adipogenesis in vitro. In conclusion, we report that the glucose-lowering effect of bile acid sequestrants is mediated by a CDO1-induced hepatic cysteine restriction mimetic effect. Furthermore, the anti-obesity effect of dietary protein restriction is largely mediated by reduced dietary cysteine intake.NEW & NOTEWORTHY Hepatic cysteine availability is a key driver of hepatic gluconeogenesis. Bile acid sequestrant inhibits gluconeogenesis by inducing CDO1-mediated cysteine catabolism to reduce cysteine availability. Dietary protein restriction causes hepatic cysteine deficiency without overall amino acid deficiency. The glucose-lowering effect of dietary protein restriction is largely mediated by lower dietary cysteine intake. The anti-obesity effect of chronic dietary protein restriction is largely mediated by lower dietary cysteine intake.
Collapse
Affiliation(s)
- David J Matye
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Huaiwen Wang
- Laboratory for Molecular Biology and Cytometry Research, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Yifeng Wang
- Department of Pharmacology, Toxicology, and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Lei Xiong
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Tiangang Li
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
3
|
Tiwari RK, Ahmad A, Chadha M, Saha K, Verma H, Borgohain K, Shukla R. Modern-Day Therapeutics and Ongoing Clinical Trials against Type 2 Diabetes Mellitus: A Narrative Review. Curr Diabetes Rev 2025; 21:59-74. [PMID: 38766831 DOI: 10.2174/0115733998294919240506044544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/22/2024] [Accepted: 04/01/2024] [Indexed: 05/22/2024]
Abstract
OBJECTIVES Diabetes Mellitus (DM) is a global health concern that affects millions of people globally. The present review aims to narrate the clinical guidelines and therapeutic interventions for Type 2 Diabetes Mellitus (T2DM) patients. Furthermore, the present work summarizes the ongoing phase 1/2/3 and clinical trials against T2DM. METHODS A meticulous and comprehensive literature review was performed using various databases, such as PubMed, MEDLINE, Clinical trials database (https://clinicaltrials.gov/), and Google Scholar, to include various clinical trials and therapeutic interventions against T2DM. RESULTS Based on our findings, we concluded that most T2DM-associated clinical trials are interventional. Anti-diabetic therapeutics, including insulin, metformin, Dipeptidyl Peptidase-4 (DPP-4) inhibitors, Glucagon-Like Peptide-1 Receptor Agonists (GLP-1RAs), and Sodium- Glucose cotransporter-2 (SGLT-2) inhibitors are frontline therapeutics being clinically investigated. Currently, the therapeutics in phase IV clinical trials are mostly SGLT-2 inhibitors, implicating their critical contribution to the clinical management of T2DM. CONCLUSION Despite the success of T2DM treatments, a surge in innovative treatment options to reduce diabetic consequences and improve glycemic control is currently ongoing. More emphasis needs to be on exploring novel targeted drug candidates that can offer more sustained glycemic control.
Collapse
Affiliation(s)
- Rohit Kumar Tiwari
- Department of Clinical Research, Sharda School of Allied Health Sciences, Sharda University, Gautam Buddh Nagar, Uttar Pradesh, 201310, India
| | - Afza Ahmad
- Department of Public Health, Dr. Giri Lal Gupta Institute of Public Health and Public Affairs, University of Lucknow, Lucknow, Uttar Pradesh, 226007, India
| | - Muskan Chadha
- Department of Nutrition & Dietetics, Sharda School of Allied Health Sciences, Sharda University, Gautam Buddh Nagar, Uttar Pradesh, 201310, India
| | - Kingshuk Saha
- Department of Clinical Research, Sharda School of Allied Health Sciences, Sharda University, Gautam Buddh Nagar, Uttar Pradesh, 201310, India
| | - Harshitha Verma
- Department of Science in Biochemistry, Manasagangothri, University of Mysuru, Mysuru, 570006, Karnataka, India
| | - Kalpojit Borgohain
- Department of Clinical Research, Sharda School of Allied Health Sciences, Sharda University, Gautam Buddh Nagar, Uttar Pradesh, 201310, India
| | - Ratnakar Shukla
- Department of Clinical Research, Sharda School of Allied Health Sciences, Sharda University, Gautam Buddh Nagar, Uttar Pradesh, 201310, India
| |
Collapse
|
4
|
de Jong LA, Li X, Emamipour S, van der Werf S, Postma MJ, van Dijk PR, Feenstra TL. Evaluating the Cost-Utility of Continuous Glucose Monitoring in Individuals with Type 1 Diabetes: A Systematic Review of the Methods and Quality of Studies Using Decision Models or Empirical Data. PHARMACOECONOMICS 2024; 42:929-953. [PMID: 38904911 PMCID: PMC11343921 DOI: 10.1007/s40273-024-01388-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 06/22/2024]
Abstract
INTRODUCTION This review presents a critical appraisal of differences in the methodologies and quality of model-based and empirical data-based cost-utility studies on continuous glucose monitoring (CGM) in type 1 diabetes (T1D) populations. It identifies key limitations and challenges in health economic evaluations on CGM and opportunities for their improvement. METHODS The review and its documentation adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines for systematic reviews. Searches for articles published between January 2000 and January 2023 were conducted using the MEDLINE, Embase, Web of Science, Cochrane Library, and Econlit databases. Published studies using models and empirical data to evaluate the cost utility of all CGM devices used by T1D patients were included in the search. Two authors independently extracted data on interventions, populations, model settings (e.g., perspectives and time horizons), model types and structures, clinical outcomes used to populate the model, validation, and uncertainty analyses. They subsequently met to confirm consensus. Quality was assessed using the Philips checklist for model-based studies and the Consensus Health Economic Criteria (CHEC) checklist for empirical studies. Model validation was assessed using the Assessment of the Validation Status of Health-Economic decision models (AdViSHE) checklist. The extracted data were used to generate summary tables and figures. The study protocol is registered with PROSPERO (CRD42023391284). RESULTS In total, 34 studies satisfied the selection criteria, two of which only used empirical data. The remaining 32 studies applied 10 different models, with a substantial majority adopting the CORE Diabetes Model. Model-based studies often lacked transparency, as their assumptions regarding the extrapolation of treatment effects beyond available evidence from clinical studies and the selection and processing of the input data were not explicitly stated. Initial scores for disagreements concerning checklists were relatively high, especially for the Philips checklist. Following their resolution, overall quality scores were moderate at 56%, whereas model validation scores were mixed. Strikingly, costing approaches differed widely across studies, resulting in little consistency in the elements included in intervention costs. DISCUSSION AND CONCLUSION The overall quality of studies evaluating CGM was moderate. Potential areas of improvement include developing systematic approaches for data selection, improving uncertainty analyses, clearer reporting, and explaining choices for particular modeling approaches. Few studies provided the assurance that all relevant and feasible options had been compared, which is required by decision makers, especially for rapidly evolving technologies such as CGM and insulin administration. High scores for disagreements indicated that several checklists contained questions that were difficult to interpret consistently for quality assessment. Therefore, simpler but comprehensive quality checklists may be needed for model-based health economic evaluation studies.
Collapse
Affiliation(s)
- Lisa A de Jong
- Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Xinyu Li
- Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Sajad Emamipour
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Sjoukje van der Werf
- Central Medical Library, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Maarten J Postma
- Department of Health Sciences, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Economics, Econometrics and Finance, Faculty of Economics and Business, University of Groningen, Groningen, The Netherlands
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
- Department of Pharmacology and Therapy, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Peter R van Dijk
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
- Department of Internal Medicine, Diabetes Center, Isala, Zwolle, The Netherlands
| | - Talitha L Feenstra
- Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands.
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands.
| |
Collapse
|
5
|
Wang Y, Xu H, Zhou X, Chen W, Zhou H. Dysregulated bile acid homeostasis: unveiling its role in metabolic diseases. MEDICAL REVIEW (2021) 2024; 4:262-283. [PMID: 39135605 PMCID: PMC11317083 DOI: 10.1515/mr-2024-0020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/13/2024] [Indexed: 08/15/2024]
Abstract
Maintaining bile acid homeostasis is essential for metabolic health. Bile acid homeostasis encompasses a complex interplay between biosynthesis, conjugation, secretion, and reabsorption. Beyond their vital role in digestion and absorption of lipid-soluble nutrients, bile acids are pivotal in systemic metabolic regulation. Recent studies have linked bile acid dysregulation to the pathogenesis of metabolic diseases, including obesity, type 2 diabetes mellitus (T2DM), and metabolic dysfunction-associated steatotic liver disease (MASLD). Bile acids are essential signaling molecules that regulate many critical biological processes, including lipid metabolism, energy expenditure, insulin sensitivity, and glucose metabolism. Disruption in bile acid homeostasis contributes to metabolic disease via altered bile acid feedback mechanisms, hormonal dysregulation, interactions with the gut microbiota, and changes in the expression and function of bile acid transporters and receptors. This review summarized the essential molecular pathways and regulatory mechanisms through which bile acid dysregulation contributes to the pathogenesis and progression of obesity, T2DM, and MASLD. We aim to underscore the significance of bile acids as potential diagnostic markers and therapeutic agents in the context of metabolic diseases, providing insights into their application in translational medicine.
Collapse
Affiliation(s)
- Yanyan Wang
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond Veterans Affairs Medical Center, Richmond, VA, USA
- School of Pharmaceutical Science, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Huangru Xu
- School of Life Science, Nanjing University, Nanjing, Jiangsu, China
| | - Xiqiao Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Weidong Chen
- School of Pharmaceutical Science, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond Veterans Affairs Medical Center, Richmond, VA, USA
| |
Collapse
|
6
|
Nerild HH, Brønden A, Haddouchi AE, Ellegaard AM, Hartmann B, Rehfeld JF, Holst JJ, Sonne DP, Vilsbøll T, Knop FK. Elucidating the glucose-lowering effect of the bile acid sequestrant sevelamer. Diabetes Obes Metab 2024; 26:1252-1263. [PMID: 38151760 DOI: 10.1111/dom.15421] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 12/29/2023]
Abstract
AIM Bile acid sequestrants are cholesterol-lowering drugs, which also improve glycaemic control in people with type 2 diabetes. The mechanism behind the glucose-lowering effect is unknown but has been proposed to be mediated by increased glucagon-like peptide-1 (GLP-1) secretion. Here, we investigated the glucose-lowering effects of sevelamer including any contribution from GLP-1 in people with type 2 diabetes. MATERIALS AND METHODS In a randomized, double-blind, placebo-controlled, crossover study, 15 people with type 2 diabetes on metformin monotherapy underwent two 17-day treatment periods with the bile acid sequestrant sevelamer and placebo, respectively, in a randomized order and with an interposed wash-out period of minimum 6 weeks. On days 15 and 17 of each treatment period, participants underwent experimental days with 4-h liquid meal tests and application of concomitant infusion of exendin(9-39)NH2 or saline. RESULTS Compared with placebo, sevelamer improved insulin sensitivity (assessed by homeostatic model assessment of insulin resistance) and beta-cell sensitivity to glucose and lowered fasting and postprandial plasma glucose concentrations. In both treatment periods, exendin(9-39)NH2 increased postprandial glucose excursions compared with saline but without absolute or relative difference between the two treatment periods. In contrast, exendin(9-39)NH2 abolished the sevelamer-induced improvement in beta-cell glucose sensitivity. CONCLUSIONS The bile acid sequestrant sevelamer improved insulin sensitivity and beta-cell sensitivity to glucose, but using the GLP-1 receptor antagonist exendin(9-39)NH2 we were not able to detect a GLP-1-mediated glucose-lowering effect of sevelamer in individuals with type 2 diabetes. Nevertheless, the sevelamer-induced improvement of beta-cell sensitivity to glucose was shown to be GLP-1-dependent.
Collapse
Affiliation(s)
- Henriette H Nerild
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
| | - Andreas Brønden
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
- Department of Clinical Pharmacology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Abdullah E Haddouchi
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
| | - Anne-Marie Ellegaard
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- the Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - David P Sonne
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
- Department of Clinical Pharmacology, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Copenhagen University Hospital - Herlev and Gentofte, Hellerup, Denmark
- the Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| |
Collapse
|
7
|
Gether IM, Bahne E, Nerild HH, Rehfeld JF, Hartmann B, Holst JJ, Vilsbøll T, Sonne DP, Knop FK. Colesevelam has no acute effect on postprandial GLP-1 levels but abolishes gallbladder refilling. Eur J Endocrinol 2024; 190:314-326. [PMID: 38551029 DOI: 10.1093/ejendo/lvae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/04/2024] [Accepted: 03/14/2024] [Indexed: 04/18/2024]
Abstract
OBJECTIVE Colesevelam, a bile acid sequestrant approved for the treatment of hypercholesterolaemia, improves glycaemic control in type 2 diabetes. We hypothesised that single-dose colesevelam increases postprandial GLP-1 secretion, thus, reducing postprandial glucose excursions in individuals with type 2 diabetes. Further, we explored the effects of single-dose colesevelam on ultrasonography-assessed postprandial gallbladder motility, paracetamol absorption (proxy for gastric emptying), and circulating factors known to affect gallbladder motility. METHODS In a randomised, double-blind, placebo-controlled crossover study, 12 individuals with type 2 diabetes (mean ± SD: age 61 ± 8.8 years; body mass index 29.8 ± 3.0 kg/m2) were subjected to 4 mixed meal tests on separate days; 2 with orally administered colesevelam (3.75 g) and 2 with placebo, with intravenous infusion of the GLP-1 receptor antagonist exendin(9-39)NH2 or saline. RESULTS Single-dose colesevelam had no effect on postprandial concentrations of glucose (P = .786), C-peptide (P = .440), or GLP-1 (P = .729), and exendin(9-39)NH2 administration revealed no GLP-1-mediated effects of colesevelam. Colesevelam did not affect gallbladder emptying but abolished gallbladder refilling (P = .001), increased postprandial cholecystokinin (CCK) secretion (P = .010), and decreased postprandial serum concentrations of fibroblast growth factor 19 (FGF19) (P = .035) and bile acids (P = .043). CONCLUSION Single-dose colesevelam had no effect on postprandial GLP-1 responses or glucose tolerance but disrupted postprandial gallbladder refilling by increasing CCK secretion and reducing circulating concentrations of FGF19 and bile acids. These findings leave the antidiabetic actions of colesevelam unresolved but provide mechanistic insights into its effect on gallbladder motility.
Collapse
Affiliation(s)
- Ida M Gether
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Emilie Bahne
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Henriette H Nerild
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, DK-2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, DK-2730 Herlev, Denmark
| | - David P Sonne
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, DK-2400 Copenhagen, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, DK-2730 Herlev, Denmark
| |
Collapse
|
8
|
Huber H, Schieren A, Holst JJ, Simon MC. Dietary impact on fasting and stimulated GLP-1 secretion in different metabolic conditions - a narrative review. Am J Clin Nutr 2024; 119:599-627. [PMID: 38218319 PMCID: PMC10972717 DOI: 10.1016/j.ajcnut.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/03/2024] [Accepted: 01/09/2024] [Indexed: 01/15/2024] Open
Abstract
Glucagon-like peptide 1 (GLP-1), a gastrointestinal peptide and central mediator of glucose metabolism, is secreted by L cells in the intestine in response to food intake. Postprandial secretion of GLP-1 is triggered by nutrient-sensing via transporters and G-protein-coupled receptors (GPCRs). GLP-1 secretion may be lower in adults with obesity/overweight (OW) or type 2 diabetes mellitus (T2DM) than in those with normal glucose tolerance (NGT), but these findings are inconsistent. Because of the actions of GLP-1 on stimulating insulin secretion and promoting weight loss, GLP-1 and its analogs are used in pharmacologic preparations for the treatment of T2DM. However, physiologically stimulated GLP-1 secretion through the diet might be a preventive or synergistic method for improving glucose metabolism in individuals who are OW, or have impaired glucose tolerance (IGT) or T2DM. This narrative review focuses on fasting and postprandial GLP-1 secretion in individuals with different metabolic conditions and degrees of glucose intolerance. Further, the influence of relevant diet-related factors (e.g., specific diets, meal composition, and size, phytochemical content, and gut microbiome) that could affect fasting and postprandial GLP-1 secretion are discussed. Some studies showed diminished glucose- or meal-stimulated GLP-1 response in participants with T2DM, IGT, or OW compared with those with NGT, whereas other studies have reported an elevated or unchanged GLP-1 response in T2DM or IGT. Meal composition, especially the relationship between macronutrients and interventions targeting the microbiome can impact postprandial GLP-1 secretion, although it is not clear which macronutrients are strong stimulants of GLP-1. Moreover, glucose tolerance, antidiabetic treatment, grade of overweight/obesity, and sex were important factors influencing GLP-1 secretion. The results presented in this review highlight the potential of nutritional and physiologic stimulation of GLP-1 secretion. Further research on fasting and postprandial GLP-1 concentrations and the resulting metabolic consequences under different metabolic conditions is needed.
Collapse
Affiliation(s)
- Hanna Huber
- Department of Psychiatry and Neurochemistry, The Sahlgrenska Academy at the University of Gothenburg, Institute of Neuroscience and Physiology, Mölndal, Sweden; Department Nutrition and Microbiota, University of Bonn, Institute of Nutrition and Food Science, Bonn, Germany
| | - Alina Schieren
- Department Nutrition and Microbiota, University of Bonn, Institute of Nutrition and Food Science, Bonn, Germany
| | - Jens Juul Holst
- Department of Biomedical Sciences, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Marie-Christine Simon
- Department Nutrition and Microbiota, University of Bonn, Institute of Nutrition and Food Science, Bonn, Germany.
| |
Collapse
|
9
|
Kugler BA, Cao X, Wenger M, Franczak E, McCoin CS, Von Schulze A, Morris EM, Thyfault JP. Divergence in aerobic capacity influences hepatic and systemic metabolic adaptations to bile acid sequestrant and short-term high-fat/sucrose feeding in rats. Am J Physiol Regul Integr Comp Physiol 2023; 325:R712-R724. [PMID: 37811712 PMCID: PMC11178297 DOI: 10.1152/ajpregu.00133.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023]
Abstract
High versus low aerobic capacity significantly impacts the risk for metabolic diseases. Rats selectively bred for high or low intrinsic aerobic capacity differently modify hepatic bile acid metabolism in response to high-fat diets (HFDs). Here we tested if a bile acid sequestrant would alter hepatic and whole body metabolism differently in rats with high and low aerobic capacity fed a 1-wk HFD. Male rats (8 mo of age) that were artificially selected to be high (HCR) and low-capacity runners (LCR) with divergent intrinsic aerobic capacities were transitioned from a low-fat diet (LFD, 10% fat) to an HFD (45% fat) with or without a bile acid sequestrant (BA-Seq, 2% cholestyramine resin) for 7 days while maintained in an indirect calorimetry system. HFD + BA-Seq increased fecal excretion of lipids and bile acids and prevented weight and fat mass gain in both strains. Interestingly, HCR rats had increased adaptability to enhance fecal bile acid and lipid loss, resulting in more significant energy loss than their LCR counterpart. In addition, BA-Seq induced a greater expression of hepatic CYP7A1 gene expression, the rate-limiting enzyme of bile acid synthesis in HCR rats both on HFD and HFD + BA-Seq diets. HCR displayed a more significant reduction of RQ in response to HFD than LCR, but HFD + BA-Seq lowered RQ in both groups compared with HFD alone, demonstrating a pronounced impact on metabolic flexibility. In conclusion, BA-Seq provides uniform metabolic benefits for metabolic flexibility and adiposity, but rats with higher aerobic capacity display adaptability for hepatic bile acid metabolism.NEW & NOTEWORTHY The administration of bile acid sequestrant (BA-Seq) has uniform metabolic benefits in terms of metabolic flexibility and adiposity in rats with high and low aerobic capacity. However, rats with higher aerobic capacity demonstrate greater adaptability in hepatic bile acid metabolism, resulting in increased fecal bile acid and lipid loss, as well as enhanced fecal energy loss.
Collapse
Affiliation(s)
- Benjamin A Kugler
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Xin Cao
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Madi Wenger
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Edziu Franczak
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| | - Colin S McCoin
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| | - Alex Von Schulze
- Stowers Research Institute, Kansas City, Missouri, United States
| | - E Matthew Morris
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
| | - John P Thyfault
- Department of Cell Biology and Physiology, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas Center for Metabolism and Obesity Research, Kansas City, Missouri, United States
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri, United States
- Department of Internal Medicine, Division of Endocrinology and Metabolism, KU Diabetes Institute, The University of Kansas Medical Center, Kansas City, Kansas, United States
- Kansas City Veterans Affairs Medical Center, Kansas City, Missouri, United States
| |
Collapse
|
10
|
Zhang X, Zhao F, Ma T, Zheng Y, Cao J, Li C, Zhu K. UPLC-Q-TOF/MS-based metabonomics reveals mechanisms for Holothuria leucospilota polysaccharides (HLP)-regulated serum metabolic changes in diabetic rats. Food Chem X 2023; 19:100741. [PMID: 37780338 PMCID: PMC10534105 DOI: 10.1016/j.fochx.2023.100741] [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: 07/15/2022] [Revised: 05/23/2023] [Accepted: 06/04/2023] [Indexed: 10/03/2023] Open
Abstract
This study aimed to use metabolomic methods to explore how Holothuria leucospilota polysaccharides (HLP) improved metabolism disorders in the liver of Goto-Kakizaki (GK) rats with spontaneous type 2 diabetes. The results showed that HLP effectively improved the metabolic disorder. Based on KEGG functional analysis, five key biomarkers associated with bile acid metabolism were detected and screened (P < 0.05). The results of serum total bile acid levels and liver damage in diabetic rats further showed the regulatory effects of HLP on bile acid metabolism. The results of bile acid-related gene expression in the liver showed that HLP inhibited liver farnesoid X Receptor - small heterodimer partner (FXR-SHP) signalling and increased the expression of bile acid synthesis genes (P < 0.05). Our results explored the underlying mechanisms by which HLP accelerated cholesterol consumption to anti-hypercholesterolemia and anti-diabetic by inhibiting liver FXR-SHP signaling. HLP's effect on bile acid regulation provides insights into treating T2DM.
Collapse
Affiliation(s)
- Xin Zhang
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Fuqiang Zhao
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Tingting Ma
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Yuanping Zheng
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Jun Cao
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Chuan Li
- Engineering Research Center of Utilization of Tropical Polysaccharide Resources of Ministry of Education, School of Food Science and Engineering, Hainan University, Haikou 570228, China
- Collaborative Innovation Center of Provincial and Ministerial Co-construction for Marine Food Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Kexue Zhu
- Spice and Beverage Research Institute, Chinese Academy of Tropical Agricultural Sciences, Key Laboratory of Processing Suitability and Quality Control of the Special Tropical Crops of Hainan Province, Wanning 571533, China
| |
Collapse
|
11
|
Chakaroun RM, Olsson LM, Bäckhed F. The potential of tailoring the gut microbiome to prevent and treat cardiometabolic disease. Nat Rev Cardiol 2023; 20:217-235. [PMID: 36241728 DOI: 10.1038/s41569-022-00771-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2022] [Indexed: 12/12/2022]
Abstract
Despite milestones in preventive measures and treatment, cardiovascular disease (CVD) remains associated with a high burden of morbidity and mortality. The protracted nature of the development and progression of CVD motivates the identification of early and complementary targets that might explain and alleviate any residual risk in treated patients. The gut microbiota has emerged as a sentinel between our inner milieu and outer environment and relays a modified risk associated with these factors to the host. Accordingly, numerous mechanistic studies in animal models support a causal role of the gut microbiome in CVD via specific microbial or shared microbiota-host metabolites and have identified converging mammalian targets for these signals. Similarly, large-scale cohort studies have repeatedly reported perturbations of the gut microbial community in CVD, supporting the translational potential of targeting this ecological niche, but the move from bench to bedside has not been smooth. In this Review, we provide an overview of the current evidence on the interconnectedness of the gut microbiome and CVD against the noisy backdrop of highly prevalent confounders in advanced CVD, such as increased metabolic burden and polypharmacy. We further aim to conceptualize the molecular mechanisms at the centre of these associations and identify actionable gut microbiome-based targets, while contextualizing the current knowledge within the clinical scenario and emphasizing the limitations of the field that need to be overcome.
Collapse
Affiliation(s)
- Rima Mohsen Chakaroun
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Lisa M Olsson
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Region Västra Götaland, Sahlgrenska University Hospital, Department of Clinical Physiology, Gothenburg, Sweden.
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
12
|
Sah DK, Arjunan A, Park SY, Jung YD. Bile acids and microbes in metabolic disease. World J Gastroenterol 2022; 28:6846-6866. [PMID: 36632317 PMCID: PMC9827586 DOI: 10.3748/wjg.v28.i48.6846] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/01/2022] [Accepted: 12/05/2022] [Indexed: 12/26/2022] Open
Abstract
Bile acids (BAs) serve as physiological detergents that enable the intestinal absorption and transportation of nutrients, lipids and vitamins. BAs are primarily produced by humans to catabolize cholesterol and play crucial roles in gut metabolism, microbiota habitat regulation and cell signaling. BA-activated nuclear receptors regulate the enterohepatic circulation of BAs which play a role in energy, lipid, glucose, and drug metabolism. The gut microbiota plays an essential role in the biotransformation of BAs and regulates BAs composition and metabolism. Therefore, altered gut microbial and BAs activity can affect human metabolism and thus result in the alteration of metabolic pathways and the occurrence of metabolic diseases/syndromes, such as diabetes mellitus, obesity/hypercholesterolemia, and cardiovascular diseases. BAs and their metabolites are used to treat altered gut microbiota and metabolic diseases. This review explores the increasing body of evidence that links alterations of gut microbial activity and BAs with the pathogenesis of metabolic diseases. Moreover, we summarize existing research on gut microbes and BAs in relation to intracellular pathways pertinent to metabolic disorders. Finally, we discuss how therapeutic interventions using BAs can facilitate microbiome functioning and ease metabolic diseases.
Collapse
Affiliation(s)
- Dhiraj Kumar Sah
- Department of Biochemistry, Chonnam National University, Gwangju 501190, South Korea
| | - Archana Arjunan
- Department of Biochemistry, Chonnam National University, Gwangju 501190, South Korea
| | - Sun Young Park
- Department of Internal Medicine, Chonnam National University, Gwangju 501190, South Korea
| | - Young Do Jung
- Department of Biochemistry, Chonnam National University, Gwangju 501190, South Korea
| |
Collapse
|
13
|
Huang Y, Lou X, Jiang C, Ji X, Tao X, Sun J, Bao Z. Gut microbiota is correlated with gastrointestinal adverse events of metformin in patients with type 2 diabetes. Front Endocrinol (Lausanne) 2022; 13:1044030. [PMID: 36465607 PMCID: PMC9714661 DOI: 10.3389/fendo.2022.1044030] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 10/03/2022] [Indexed: 11/18/2022] Open
Abstract
Aim Gastrointestinal discomfort is the most common adverse event in metformin treatment for type 2 diabetes. The mechanism of action of metformin is associated with gut microbiota. However, the gut microbial community structure related to metformin-induced gastrointestinal adverse events remains unclear. This study aimed to investigate it. Methods 50 patients with newly diagnosed diabetes were treated with metformin 1500mg/d for 12 weeks. The patients were divided into two groups according to whether gastrointestinal adverse events occurred (group B) or did not occur (group A) after treatment. The fecal bacterial communities and short-chain fatty acids (SCFAs) were sequenced and compared. 70 diabetes mice were randomly divided into 8 groups and treated with metformin (Met), clindamycin (Clin) and/or SCFA, which were the Met+/Clin+, Met+/Clin-, Met-/Clin+, Met-/Clin-, Met+/SCFA+, Met+/SCFA-, Met-/SCFA+ and Met-/SCFA- group. After 4 weeks of metformin treatment, blood glucose, food intake, fecal SCFAs, gut microbiota and gut hormones were measured. Results Metformin increased the abundance of Phascolarctobacterium, Intestinimonas and Clostridium III. Functional prediction analysis showed that the propanoate metabolism pathway was significantly up-regulated. The concentrations of acetic acid and propanoic acid in feces were significantly increased. The abundance of Clostridium sensu stricto, Streptococcus and Akkermansia induced by metformin in group B was higher than that in group A. The propanoate metabolism pathway and propanoic acid in feces were significantly up-regulated in group B. In the animal experiments, the food intake decreased and glucose control increased in metformin groups compared with those in the control groups. The total GLP-1 level in the Met+/Clin- group was significantly higher than that in the Met-/Clin- group, while there was no statistical difference between the Met-/Clin- and Met+/Clin+ group. The total GLP-1 level in the Met-/SCFA+ group was significantly higher than that in the Met-/SCFA-group, while the levels of total GLP-1 and active GLP-1 in the Met+/SCFA- group and the Met+/SCFA+ group were significantly higher than those in the Met-/SCFA-group. Conclusions Our data suggest that metformin promotes the secretion of intestinal hormones such as GLP-1 by increasing the abundance of SCFA-producing bacteria, which not only plays an anti-diabetic role, but also may causes gastrointestinal adverse events.
Collapse
Affiliation(s)
- Yuxin Huang
- Department of Endocrinology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Xudan Lou
- Department of Endocrinology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Cuiping Jiang
- Department of Endocrinology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Xueying Ji
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Xiaoming Tao
- Department of Endocrinology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jiao Sun
- Department of Endocrinology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Zhijun Bao
- Department of Gerontology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
14
|
Lan WH, Lin TY, Yeh JA, Feng CL, Hsu JT, Lin HJ, Kuo CJ, Lai CH. Mechanism Underlying Metformin Action and Its Potential to Reduce Gastric Cancer Risk. Int J Mol Sci 2022; 23:14163. [PMID: 36430639 PMCID: PMC9695469 DOI: 10.3390/ijms232214163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/10/2022] [Accepted: 11/11/2022] [Indexed: 11/18/2022] Open
Abstract
Diabetes mellitus is associated with a high risk of developing gastric cancer (GC). Metformin, which is conventionally used to treat type 2 diabetes, induces AMP-activated protein kinase signaling and suppresses gluconeogenesis. Recent studies have reported that metformin is associated with beneficial effects in cancer prevention and treatment owing to its anti-tumor effects. This makes metformin a potential medication for GC therapy. However, contradicting reports have emerged regarding the efficacy of metformin in reducing the risk of GC. This review summarizes the impact of metformin on mitigating GC risk by analyzing clinical databases. The mechanism underlying the anti-tumor effect of metformin on GC is also discussed.
Collapse
Affiliation(s)
- Wen-Hsi Lan
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ting-Yu Lin
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Jia-Ai Yeh
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chun-Lung Feng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, China Medical University Hsinchu Hospital, Hsinchu 30272, Taiwan
- Department of Internal Medicine, Department of Medical Research, School of Medicine, China Medical University and Hospital, Taichung 40402, Taiwan
| | - Jun-Te Hsu
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of General Surgery, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
| | - Hwai-Jeng Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang-Ho Hospital, New Taipei 23562, Taiwan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chia-Jung Kuo
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
- Chang Gung Microbiota Therapy Center, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
| | - Chih-Ho Lai
- Department of Microbiology and Immunology, Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Internal Medicine, Department of Medical Research, School of Medicine, China Medical University and Hospital, Taichung 40402, Taiwan
- Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital at Linkou, Taoyuan 33305, Taiwan
- Department of Nursing, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
15
|
Cook JR, Kohan AB, Haeusler RA. An Updated Perspective on the Dual-Track Model of Enterocyte Fat Metabolism. J Lipid Res 2022; 63:100278. [PMID: 36100090 PMCID: PMC9593242 DOI: 10.1016/j.jlr.2022.100278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/08/2022] [Accepted: 08/31/2022] [Indexed: 02/04/2023] Open
Abstract
The small intestinal epithelium has classically been envisioned as a conduit for nutrient absorption, but appreciation is growing for a larger and more dynamic role for enterocytes in lipid metabolism. Considerable gaps remain in our knowledge of this physiology, but it appears that the enterocyte's structural polarization dictates its behavior in fat partitioning, treating fat differently based on its absorption across the apical versus the basolateral membrane. In this review, we synthesize existing data and thought on this dual-track model of enterocyte fat metabolism through the lens of human integrative physiology. The apical track includes the canonical pathway of dietary lipid absorption across the apical brush-border membrane, leading to packaging and secretion of those lipids as chylomicrons. However, this track also reserves a portion of dietary lipid within cytoplasmic lipid droplets for later uses, including the "second-meal effect," which remains poorly understood. At the same time, the enterocyte takes up circulating fats across the basolateral membrane by mechanisms that may include receptor-mediated import of triglyceride-rich lipoproteins or their remnants, local hydrolysis and internalization of free fatty acids, or enterocyte de novo lipogenesis using basolaterally absorbed substrates. The ultimate destinations of basolateral-track fat may include fatty acid oxidation, structural lipid synthesis, storage in cytoplasmic lipid droplets, or ultimate resecretion, although the regulation and purposes of this basolateral track remain mysterious. We propose that the enterocyte integrates lipid flux along both of these tracks in order to calibrate its overall program of lipid metabolism.
Collapse
Affiliation(s)
- Joshua R. Cook
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, NY, USA,Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Alison B. Kohan
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rebecca A. Haeusler
- Naomi Berrie Diabetes Center, Columbia University College of Physicians and Surgeons, New York, NY, USA,Department of Pathology and Cell Biology; Columbia University College of Physicians and Surgeons, New York, NY, USA,For correspondence: Rebecca A. Haeusler
| |
Collapse
|
16
|
Esan O, Viljoen A, Wierzbicki AS. Colesevelam - a bile acid sequestrant for treating hypercholesterolemia and improving hyperglycemia. Expert Opin Pharmacother 2022; 23:1363-1370. [PMID: 35968655 DOI: 10.1080/14656566.2022.2112945] [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: 11/04/2022]
Abstract
INTRODUCTION Low density Lipoprotein cholesterol)LDL-C) levels show a clear relationship with cardiovascular disease (CVD). Statins are first line agents to reduce LDL-C and CVD risk. However, combination lipid-lowering therapy is often required to achieve large reductions in LDL-C. AREA COVERED Colesevelam HCl is a bile acid sequestrant (BAS), which reduces LDL-C by 16-22% in monotherapy and adds a further 12-14% reduction in LDL-C when combined with other lipid-lowering drugs. Like statins, colesevelam reduces C-reactive protein levels by 16% in monotherapy and additional 6% when added to statins. Colesevelam also reduced HbA1c by 4mmol/mol (0.5%) when used alone and added to other hypoglycaemic drugs in studies of patients with diabetes . EXPERT OPINION Bile acid sequestrants reduce LDL-C and HbA1c and have some CVD outcome evidence. The uses of these agents are limited in patients with gastrointestinal disease or high triglycerides due to adverse effects on gut function and raising triglycerides and they interfere with the absorption of lipid-soluble drugs. Colesevelam has a higher bile acid binding capacity, and fewer adverse effects than other BAS. Colesevelam may be useful as a third line agent for treatment of hypercholesterolemia with some additional specific benefits on glycemic control.
Collapse
Affiliation(s)
- Oluwayemisi Esan
- Metabolic Medicine/Chemical Pathology, Guy's & St Thomas Hospitals, London SE1 7EH, UK
| | - Adie Viljoen
- Metabolic Medicine/Chemical Pathology, East & North Hertfordshire Hospitals, Lister Hospital, Stevenage, Hertfordshire SG1 4AB, UK
| | - Anthony S Wierzbicki
- Metabolic Medicine/Chemical Pathology, Guy's & St Thomas Hospitals, London SE1 7EH, UK
| |
Collapse
|
17
|
Stierwalt HD, Morris EM, Maurer A, Apte U, Phillips K, Li T, Meers GME, Koch LG, Britton SL, Graf G, Rector RS, Mercer K, Shankar K, Thyfault JP. Rats with high aerobic capacity display enhanced transcriptional adaptability and upregulation of bile acid metabolism in response to an acute high-fat diet. Physiol Rep 2022; 10:e15405. [PMID: 35923133 PMCID: PMC9350427 DOI: 10.14814/phy2.15405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 07/01/2022] [Accepted: 07/13/2022] [Indexed: 06/09/2023] Open
Abstract
Rats selectively bred for the high intrinsic aerobic capacity runner (HCR) or low aerobic capacity runner (LCR) show pronounced differences in susceptibility for high-fat/high sucrose (HFHS) diet-induced hepatic steatosis and insulin resistance, replicating the protective effect of high aerobic capacity in humans. We have previously shown multiple systemic differences in energy and substrate metabolism that impacts steatosis between HCR and LCR rats. This study aimed to investigate hepatic-specific mechanisms of action via changes in gene transcription. Livers of HCR rats had a greater number of genes that significantly changed in response to 3-day HFHS compared with LCR rats (171 vs. 75 genes: >1.5-fold, p < 0.05). HCR and LCR rats displayed numerous baseline differences in gene expression while on a low-fat control diet (CON). A 3-day HFHS diet resulted in greater expression of genes involved in the conversion of excess acetyl-CoA to cholesterol and bile acid (BA) synthesis compared with the CON diet in HCR, but not LCR rats. These results were associated with higher fecal BA loss and lower serum BA concentrations in HCR rats. Exercise studies in rats and mice also revealed higher hepatic expression of cholesterol and BA synthesis genes. Overall, these results suggest that high aerobic capacity and exercise are associated with upregulated BA synthesis paired with greater fecal excretion of cholesterol and BA, an effect that may play a role in protection against hepatic steatosis in rodents.
Collapse
Affiliation(s)
- Harrison D. Stierwalt
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
- Research ServiceKansas City VA Medical CenterKansas CityMissouriUSA
| | - E. Matthew Morris
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | - Adrianna Maurer
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | - Udayan Apte
- Department of Pharmacology, Toxicology, and TherapeuticsUniversity of Kansas Medical CenterKansas CityMissouriUSA
| | | | - Tiangang Li
- Department of PhysiologyUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Grace M. E. Meers
- Division of Gastroenterology and HepatologyUniversity of MissouriColumbiaMissouriUSA
- Division of Nutrition and Exercise PhysiologyColumbiaMissouriUSA
| | - Lauren G. Koch
- Physiology and PharmacologyThe University of ToledoToledoOhioUSA
| | | | - Greg Graf
- Department of Pharmaceutical SciencesSaha Cardiovascular Research Center, University of KentuckyLexingtonKentuckyUSA
| | - R. Scott Rector
- Division of Gastroenterology and HepatologyUniversity of MissouriColumbiaMissouriUSA
- Division of Nutrition and Exercise PhysiologyColumbiaMissouriUSA
- Research ServiceHarry S Truman Memorial VA HospitalColumbiaMissouriUSA
| | - Kelly Mercer
- Arkansas Children's Nutrition CenterUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Kartik Shankar
- Section of Nutrition, Department of PediatricsUniversity of Colorado School of Medicine Anschutz Medical CampusAuroraColoradoUSA
| | - John P. Thyfault
- Molecular and Integrative PhysiologyUniversity of Kansas Medical CenterKansas CityMissouriUSA
- Research ServiceKansas City VA Medical CenterKansas CityMissouriUSA
| |
Collapse
|
18
|
Genetic reprogramming of remnant duodenum may contribute to type 2 diabetes improvement after Roux en-Y gastric bypass. Nutrition 2022; 99-100:111631. [DOI: 10.1016/j.nut.2022.111631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 12/31/2021] [Accepted: 02/07/2022] [Indexed: 11/22/2022]
|
19
|
Miedzybrodzka EL, Gribble FM, Reimann F. Targeting the Enteroendocrine System for Treatment of Obesity. Handb Exp Pharmacol 2022; 274:487-513. [PMID: 35419620 DOI: 10.1007/164_2022_583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Mimetics of the anorexigenic gut hormone glucagon-like peptide 1 (GLP-1) were originally developed as insulinotropic anti-diabetic drugs but also evoke significant weight loss, leading to their recent approval as obesity therapeutics. Co-activation of receptors for GLP-1 and other gut hormones which reduce food intake - peptide YY (PYY3-36), cholecystokinin (CCK) and glucose-dependent insulinotropic peptide (GIP) - is now being explored clinically to enhance efficacy. An alternative approach involves pharmacologically stimulating endogenous secretion of these hormones from enteroendocrine cells (EECs) to recapitulate the metabolic consequences of bariatric surgery, where highly elevated postprandial levels of GLP-1 and PYY3-36 are thought to contribute to improved glycaemia and weight loss.
Collapse
Affiliation(s)
- Emily L Miedzybrodzka
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | - Fiona M Gribble
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
| | - Frank Reimann
- Wellcome Trust - MRC Institute of Metabolic Science, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| |
Collapse
|
20
|
Li B, Zhang J, Chen Y, Wang Q, Yan L, Wang R, Wei Y, You Z, Li Y, Miao Q, Xiao X, Lian M, Chen W, Qiu D, Fang J, Gershwin ME, Tang R, Ma X. Alterations in microbiota and their metabolites are associated with beneficial effects of bile acid sequestrant on icteric primary biliary Cholangitis. Gut Microbes 2021; 13:1946366. [PMID: 34437819 PMCID: PMC8405155 DOI: 10.1080/19490976.2021.1946366] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Background: Increasing data suggests an interaction between bile acids and intestinal microbiota in the pathogenesis of primary biliary cholangitis (PBC). Bile acid sequestrants are widely used to bind bile acids in the intestinal lumen and are therefore posited to impact gut bacteria. Herein we aimed to investigate the effects of cholestyramine on the bile acid profile and gut microbiome in a cohort of icteric PBC patients.Results: Thirty-three PBC patients were treated with cholestyramine, serum and stool samples were collected at baseline, 4 and 16 weeks. Shotgun metagenomic sequencing and targeted metabolomic profiling were performed. Following cholestyramine administration, patients exhibited a high interpersonal variability in remission of cholestasis, and were therefore dichotomized according to the decrease of total bilirubin. Gut microbial co-abundance networks showed distinct taxa interactions between subjects with superior remission (SR) and those with inferior remission (IR) at baseline. After treatment, compositional shifts of the microbiome in the SR group were characterized with enrichment of two Lachnospiraceae species, typically producing short-chain fatty acids (SCFAs). In contrast, Klebsiella pneumonia, a commensal pathobiont, was only increased in the IR group. Correspondingly, metabolome analysis demonstrated that patients with SR, but not IR, were marked by elevations of SCFAs including valeric acid and caproic acid. Finally, integrative analysis identified robust associations between the variations of microbiota, metabolites, and inflammatory cytokines in SR group, indicating potential mechanistic links.Conclusions: Beneficial responses caused by cholestyramine were closely related with compositional and functional alterations in gut commensal, highlighting the possibility of exploring bile acid-microbiota interactions for treating PBC.
Collapse
Affiliation(s)
- Bo Li
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Jun Zhang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Yong Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Qixia Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Li Yan
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Rui Wang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Yiran Wei
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Zhengrui You
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Yikang Li
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Qi Miao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Xiao Xiao
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Min Lian
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Weihua Chen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Dekai Qiu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - Jingyuan Fang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China
| | - M. Eric Gershwin
- Division of Rheumatology, Department of Medicine, Allergy and Clinical Immunology, University of California at Davis, Davis, CA, USA,M. Eric Gershwin Division of Rheumatology, Department of Medicine, Allergy and Clinical Immunology, University of California at Davis, Davis, CA, USA
| | - Ruqi Tang
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China,Ruqi Tang Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong roadShanghai 200001, China
| | - Xiong Ma
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai JiaoTong University; Shanghai Institute of Digestive Disease;145 Middle Shandong Road, Shanghai, China,CONTACT Xiong Ma
| |
Collapse
|
21
|
Zhang B, Kuipers F, de Boer JF, Kuivenhoven JA. Modulation of Bile Acid Metabolism to Improve Plasma Lipid and Lipoprotein Profiles. J Clin Med 2021; 11:jcm11010004. [PMID: 35011746 PMCID: PMC8745251 DOI: 10.3390/jcm11010004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023] Open
Abstract
New drugs targeting bile acid metabolism are currently being evaluated in clinical studies for their potential to treat cholestatic liver diseases, non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Changes in bile acid metabolism, however, translate into an alteration of plasma cholesterol and triglyceride concentrations, which may also affect cardiovascular outcomes in such patients. This review attempts to gain insight into this matter and improve our understanding of the interactions between bile acid and lipid metabolism. Bile acid sequestrants (BAS), which bind bile acids in the intestine and promote their faecal excretion, have long been used in the clinic to reduce LDL cholesterol and, thereby, atherosclerotic cardiovascular disease (ASCVD) risk. However, BAS modestly but consistently increase plasma triglycerides, which is considered a causal risk factor for ASCVD. Like BAS, inhibitors of the apical sodium-dependent bile acid transporter (ASBTi’s) reduce intestinal bile acid absorption. ASBTi’s show effects that are quite similar to those obtained with BAS, which is anticipated when considering that accelerated faecal loss of bile acids is compensated by an increased hepatic synthesis of bile acids from cholesterol. Oppositely, treatment with farnesoid X receptor agonists, resulting in inhibition of bile acid synthesis, appears to be associated with increased LDL cholesterol. In conclusion, the increasing efforts to employ drugs that intervene in bile acid metabolism and signalling pathways for the treatment of metabolic diseases such as NAFLD warrants reinforcing interactions between the bile acid and lipid and lipoprotein research fields. This review may be considered as the first step in this process.
Collapse
Affiliation(s)
- Boyan Zhang
- Department of Pediatrics, University Medical Centre Groningen, University of Groningen, 9713 AV Groningen, The Netherlands; (B.Z.); (F.K.)
| | - Folkert Kuipers
- Department of Pediatrics, University Medical Centre Groningen, University of Groningen, 9713 AV Groningen, The Netherlands; (B.Z.); (F.K.)
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Jan Freark de Boer
- Department of Pediatrics, University Medical Centre Groningen, University of Groningen, 9713 AV Groningen, The Netherlands; (B.Z.); (F.K.)
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
- Correspondence: (J.F.d.B.); (J.A.K.)
| | - Jan Albert Kuivenhoven
- Department of Pediatrics, University Medical Centre Groningen, University of Groningen, 9713 AV Groningen, The Netherlands; (B.Z.); (F.K.)
- Correspondence: (J.F.d.B.); (J.A.K.)
| |
Collapse
|
22
|
Zamek-Gliszczynski MJ, Kenworthy D, Bershas DA, Sanghvi M, Pereira AI, Mudunuru J, Crossman L, Pirhalla JL, Thorpe KM, Dennison JMTJ, McLaughlin MM, Allinder M, Swift B, O'Connor-Semmes RL, Young GC. Pharmacokinetics and ADME Characterization of Intravenous and Oral [ 14C]-Linerixibat in Healthy Male Volunteers. Drug Metab Dispos 2021; 49:1109-1117. [PMID: 34625435 DOI: 10.1124/dmd.121.000595] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/24/2021] [Indexed: 12/14/2022] Open
Abstract
Linerixibat, an oral small-molecule ileal bile acid transporter inhibitor under development for cholestatic pruritus in primary biliary cholangitis, was designed for minimal absorption from the intestine (site of pharmacological action). This study characterized the pharmacokinetics, absorption, metabolism, and excretion of [14C]-linerixibat in humans after an intravenous microtracer concomitant with unlabeled oral tablets and [14C]-linerixibat oral solution. Linerixibat exhibited absorption-limited flip-flop kinetics: longer oral versus intravenous half-life (6-7 hours vs. 0.8 hours). The short intravenous half-life was consistent with high systemic clearance (61.9 l/h) and low volume of distribution (16.3 l). In vitro studies predicted rapid hepatic clearance via cytochrome P450 3A4 metabolism, which predicted human hepatic clearance within 1.5-fold. However, linerixibat was minimally metabolized in humans after intravenous administration: ∼80% elimination via biliary/fecal excretion (>90%-97% as unchanged parent) and ∼20% renal elimination by glomerular filtration (>97% as unchanged parent). Absolute oral bioavailability of linerixibat was exceedingly low (0.05%), primarily because of a very low fraction absorbed (0.167%; fraction escaping first-pass gut metabolism (fg) ∼100%), with high hepatic extraction ratio (77.0%) acting as a secondary barrier to systemic exposure. Oral linerixibat was almost entirely excreted (>99% recovered radioactivity) in feces as unchanged and unabsorbed linerixibat. Consistent with the low oral fraction absorbed and ∼20% renal recovery of intravenous [14C]-linerixibat, urinary elimination of orally administered radioactivity was negligible (<0.04% of dose). Linerixibat unequivocally exhibited minimal gastrointestinal absorption and oral systemic exposure. Linerixibat represents a unique example of high CYP3A4 clearance in vitro but nearly complete excretion as unchanged parent drug via the biliary/fecal route. SIGNIFICANCE STATEMENT: This study conclusively established minimal absorption and systemic exposure to orally administered linerixibat in humans. The small amount of linerixibat absorbed was eliminated efficiently as unchanged parent drug via the biliary/fecal route. The hepatic clearance mechanism was mispredicted to be mediated via cytochrome P450 3A4 metabolism in vitro rather than biliary excretion of unchanged linerixibat in vivo.
Collapse
Affiliation(s)
- Maciej J Zamek-Gliszczynski
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - David Kenworthy
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - David A Bershas
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - Mitesh Sanghvi
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - Adrian I Pereira
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - Jennypher Mudunuru
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - Lee Crossman
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - Jill L Pirhalla
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - Karl M Thorpe
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - Jeremy M T J Dennison
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - Megan M McLaughlin
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - Matthew Allinder
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - Brandon Swift
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - Robin L O'Connor-Semmes
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| | - Graeme C Young
- Drug Metabolism and Disposition (M.J.Z.-G., D.A.B., J.M., J.L.P.), Medicine Development (M.M.M.), and Development Biostatistics (M.A.), GlaxoSmithKline, Collegeville, Pennsylvania; Drug Metabolism and Disposition (D.K., G.C.Y.), and Bioanalysis, Immunogenicity and Biomarkers (A.I.P.), GlaxoSmithKline, Ware, United Kingdom; Pharmaron ABS Inc., Germantown, Maryland (M.S.); Covance, Harrogate, United Kingdom (L.C.); Global Clinical Development, GlaxoSmithKline, Brentford, United Kingdom (K.M.T.); Hammersmith Medicines Research, London, United Kingdom (J.M.T.J.D.); Clinical Pharmacology, Modeling and Simulation, GlaxoSmithKline, RTP, North Carolina (B.S.); and Clinical Pharmacology, Modeling and Simulation, Parexel, Durham, North Carolina (R.L.O.-S.)
| |
Collapse
|
23
|
Li R, Andreu-Sánchez S, Kuipers F, Fu J. Gut microbiome and bile acids in obesity-related diseases. Best Pract Res Clin Endocrinol Metab 2021; 35:101493. [PMID: 33707081 DOI: 10.1016/j.beem.2021.101493] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Dysbiosis has been implemented in the etiologies of obesity-related chronic diseases such as type 2 diabetes, NAFLD and cardiovascular diseases. Bile acids, a class of amphipathic steroids produced in the liver and extensively modified by the microbiome, are increasingly recognized as actors in onset and progression of these diseases. Indeed, human obesity is associated with altered bile acid metabolism. Bile acids facilitate intestinal fat absorption but also exert hormone-like functions through activation of nuclear and membrane-bound receptors and thereby modulate glucose, lipid and energy metabolism, intestinal integrity and immunity. Bile acid-signaling pathways have thus been identified as potential pharmacological targets for obesity-related diseases. Interfering with microbiome composition may also be considered, as liver- and microbiome-derived bile acid species have different signaling functions. This review summarizes recent developments in this rapidly expanding field of research and addresses potential clinical prospects of interference with bile acid signaling pathways in human diseases.
Collapse
Affiliation(s)
- Rumei Li
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Sergio Andreu-Sánchez
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Folkert Kuipers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Jingyuan Fu
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands; Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
24
|
Li X, Kimita W, Cho J, Ko J, Bharmal SH, Petrov MS. Dietary Fibre Intake in Type 2 and New-Onset Prediabetes/Diabetes after Acute Pancreatitis: A Nested Cross-Sectional Study. Nutrients 2021; 13:nu13041112. [PMID: 33805259 PMCID: PMC8066410 DOI: 10.3390/nu13041112] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/18/2022] Open
Abstract
The association between intake of dietary fibre and glucose metabolism has been extensively investigated in numerous metabolic disorders. However, little is known about this association in individuals after an attack of acute pancreatitis (AP). The aim was to investigate the associations between intake of dietary fibre and markers of glucose metabolism in individuals with new-onset prediabetes or diabetes after acute pancreatitis (NODAP), pre-exiting type 2 prediabetes or diabetes, and normoglycaemia after acute pancreatitis. This cross-sectional study was nested within the parent prospective longitudinal cohort study. The studied markers of glucose metabolism were fasting plasma glucose and glycated haemoglobin. Habitual intake of dietary fibre was determined using the EPIC-Norfolk food frequency questionnaire. Multivariable linear regression analyses were conducted. The study included a total of 108 individuals after AP. In the NODAP group, increased intakes of total fibre (β = −0.154, p = 0.006), insoluble fibre (β = −0.133, p = 0.01), and soluble fibre (β = −0.13, p = 0.02) were significantly associated with a reduction in fasting plasma glucose. Increased intakes of vegetables (β = −0.069, p = 0.004) and nuts (β = −0.039, p = 0.038) were significantly associated with a reduction in fasting plasma glucose. Increased intake of nuts (β = −0.054, p = 0.001) was also significantly associated with a reduction in glycated haemoglobin. None of the above associations were significant in the other study groups. Habitual intake of dietary fibre was inversely associated with fasting plasma glucose in individuals with NODAP. Individuals after an attack of AP may benefit from increasing their intake of dietary fibre (specifically, vegetables and nuts) with a view to preventing NODAP.
Collapse
|
25
|
Xie C, Huang W, Young RL, Jones KL, Horowitz M, Rayner CK, Wu T. Role of Bile Acids in the Regulation of Food Intake, and Their Dysregulation in Metabolic Disease. Nutrients 2021; 13:1104. [PMID: 33800566 PMCID: PMC8066182 DOI: 10.3390/nu13041104] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Bile acids are cholesterol-derived metabolites with a well-established role in the digestion and absorption of dietary fat. More recently, the discovery of bile acids as natural ligands for the nuclear farnesoid X receptor (FXR) and membrane Takeda G-protein-coupled receptor 5 (TGR5), and the recognition of the effects of FXR and TGR5 signaling have led to a paradigm shift in knowledge regarding bile acid physiology and metabolic health. Bile acids are now recognized as signaling molecules that orchestrate blood glucose, lipid and energy metabolism. Changes in FXR and/or TGR5 signaling modulates the secretion of gastrointestinal hormones including glucagon-like peptide-1 (GLP-1) and peptide YY (PYY), hepatic gluconeogenesis, glycogen synthesis, energy expenditure, and the composition of the gut microbiome. These effects may contribute to the metabolic benefits of bile acid sequestrants, metformin, and bariatric surgery. This review focuses on the role of bile acids in energy intake and body weight, particularly their effects on gastrointestinal hormone secretion, the changes in obesity and T2D, and their potential relevance to the management of metabolic disorders.
Collapse
Affiliation(s)
- Cong Xie
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
| | - Weikun Huang
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
- The ARC Center of Excellence for Nanoscale BioPhotonics, Institute for Photonics and Advanced Sensing, School of Physical Sciences, The University of Adelaide, Adelaide 5005, Australia
| | - Richard L. Young
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
- Nutrition, Diabetes & Gut Health, Lifelong Health Theme South Australian Health & Medical Research Institute, Adelaide 5005, Australia
| | - Karen L. Jones
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide 5005, Australia
| | - Michael Horowitz
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide 5005, Australia
| | - Christopher K. Rayner
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
- Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide 5005, Australia
| | - Tongzhi Wu
- Adelaide Medical School, Center of Research Excellence (CRE) in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide 5005, Australia; (C.X.); (W.H.); (R.L.Y.); (K.L.J.); (M.H.); (C.K.R.)
- Endocrine and Metabolic Unit, Royal Adelaide Hospital, Adelaide 5005, Australia
- Institute of Diabetes, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
26
|
Duca FA, Waise TMZ, Peppler WT, Lam TKT. The metabolic impact of small intestinal nutrient sensing. Nat Commun 2021; 12:903. [PMID: 33568676 PMCID: PMC7876101 DOI: 10.1038/s41467-021-21235-y] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 01/19/2021] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal tract maintains energy and glucose homeostasis, in part through nutrient-sensing and subsequent signaling to the brain and other tissues. In this review, we highlight the role of small intestinal nutrient-sensing in metabolic homeostasis, and link high-fat feeding, obesity, and diabetes with perturbations in these gut-brain signaling pathways. We identify how lipids, carbohydrates, and proteins, initiate gut peptide release from the enteroendocrine cells through small intestinal sensing pathways, and how these peptides regulate food intake, glucose tolerance, and hepatic glucose production. Lastly, we highlight how the gut microbiota impact small intestinal nutrient-sensing in normal physiology, and in disease, pharmacological and surgical settings. Emerging evidence indicates that the molecular mechanisms of small intestinal nutrient sensing in metabolic homeostasis have physiological and pathological impact as well as therapeutic potential in obesity and diabetes. The gastrointestinal tract participates in maintaining metabolic homeostasis in part through nutrient-sensing and subsequent gut-brain signalling. Here the authors review the role of small intestinal nutrient-sensing in regulation of energy intake and systemic glucose metabolism, and link high-fat diet, obesity and diabetes with perturbations in these pathways.
Collapse
Affiliation(s)
- Frank A Duca
- BIO5 Institute, University of Arizona, Tucson, AZ, USA. .,School of Animal and Comparative Biomedical Sciences, University of Arizona, Tucson, AZ, USA.
| | - T M Zaved Waise
- Toronto General Hospital Research Institute, UHN, Toronto, Canada
| | - Willem T Peppler
- Toronto General Hospital Research Institute, UHN, Toronto, Canada
| | - Tony K T Lam
- Toronto General Hospital Research Institute, UHN, Toronto, Canada. .,Department of Physiology, University of Toronto, Toronto, Canada. .,Department of Medicine, University of Toronto, Toronto, Canada. .,Banting and Best Diabetes Centre, University of Toronto, Toronto, Canada.
| |
Collapse
|
27
|
Wang S, Ji X, Zhang Z, Xue F. Relationship between Lipid Profiles and Glycemic Control Among Patients with Type 2 Diabetes in Qingdao, China. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17155317. [PMID: 32718055 PMCID: PMC7432328 DOI: 10.3390/ijerph17155317] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/22/2020] [Accepted: 07/22/2020] [Indexed: 12/18/2022]
Abstract
Glycosylated hemoglobin (HbA1c) was the best indicator of glycemic control, which did not show the dynamic relationship between glycemic control and lipid profiles. In order to guide the health management of Type 2 diabetes (T2D), we assessed the levels of lipid profiles and fasting plasma glucose (FPG) and displayed the relationship between FPG control and lipid profiles. We conducted a cross-sectional study that included 5822 participants. Descriptive statistics were conducted according to gender and glycemic status respectively. Comparisons for the control of lipid profiles were conducted according to glycemic control. Four logistic regression models were generated to analyze the relationship between lipid profiles and glycemic control according to different confounding factors. The metabolic control percentage of FPG, triglyceride (TG), total cholesterol (TC), high density lipoprotein cholesterol (HDL-C) and low density lipoprotein cholesterol (LDL-C) was 27.50%, 73.10%, 28.10%, 64.20% and 44.80% respectively. In the fourth model with the most confounding factors, the odds ratios (ORs) and 95% confidence intervals (CIs) of TG, TC, LDL-C and HDL-C were 0.989 (0.935, 1.046), 0.862 (0.823, 0.903), 0.987 (0.920, 1.060) and 2.173 (1.761, 2.683). TC and HDL-C were statistically significant, and TG and LDL-C were not statistically significant with adjustment for different confounding factors. In conclusion, FPG was significantly associated with HDL and TC and was not associated with LDL and TG. Our findings suggested that TC and HDL should be focused on in the process of T2D health management.
Collapse
Affiliation(s)
- Shukang Wang
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhuaxi Street, Jinan 250012, Shandong, China; (S.W.); (X.J.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuandong Street, Jinan 250002, Shandong, China
| | - Xiaokang Ji
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhuaxi Street, Jinan 250012, Shandong, China; (S.W.); (X.J.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuandong Street, Jinan 250002, Shandong, China
| | - Zhentang Zhang
- Qingdao West Coast New District Center for Disease Control and Prevention, 567, Lingshanwan Street, Huangdao District, Qingdao 266400, China;
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, 44, Wenhuaxi Street, Jinan 250012, Shandong, China; (S.W.); (X.J.)
- Institute for Medical Dataology, Shandong University, 12550, Erhuandong Street, Jinan 250002, Shandong, China
- Correspondence: ; Tel.: +86-0531-88380280; Fax: +86-0531-88382553
| |
Collapse
|
28
|
Wu Y, Zhou A, Tang L, Lei Y, Tang B, Zhang L. Bile Acids: Key Regulators and Novel Treatment Targets for Type 2 Diabetes. J Diabetes Res 2020; 2020:6138438. [PMID: 32733968 PMCID: PMC7383344 DOI: 10.1155/2020/6138438] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/29/2020] [Accepted: 07/04/2020] [Indexed: 02/08/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM), characterized by insulin resistance and unclear pathogenesis, is a serious menace to human health. Bile acids are the end products of cholesterol catabolism and play an important role in maintaining cholesterol homeostasis. Furthermore, increasing studies suggest that bile acids may regulate glucose tolerance, insulin sensitivity, and energy metabolism, suggesting that bile acids may represent a potential therapeutic target for T2DM. This study summarizes the metabolism of bile acids and, more importantly, changes in their concentrations, constitution, and receptors in diabetes. Furthermore, we provide an overview of the mechanisms underlying the role of bile acids in glucose and lipid metabolism, as well as the occurrence and development of T2DM. Bile acid-targeted therapy may represent a valid approach for T2DM treatment.
Collapse
Affiliation(s)
- Yingjie Wu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510030, China
| | - An Zhou
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Li Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Yuanyuan Lei
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, China
| | - Linjing Zhang
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
29
|
Li B, Hu Y, Wang G, Liu L. The effect of exenatide on fasting bile acids in newly diagnosed type 2 diabetes mellitus patients, a pilot study. BMC Pharmacol Toxicol 2020; 21:44. [PMID: 32539783 PMCID: PMC7296654 DOI: 10.1186/s40360-020-00422-5] [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: 02/23/2020] [Accepted: 05/26/2020] [Indexed: 11/10/2022] Open
Abstract
Background Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) demonstrated good glycemic efficacy in patients with type 2 diabetes mellitus (T2DM) recent years, whereas studies on GLP-1 RAs’ biliary effects were limited. Therefore, we aimed to assess the effect of exenatide on bile acids (BAs) and investigate the role of BAs in the glycemic control effect of exenatide. Methods Thirty-eight newly diagnosed T2DM participants without glucose-lowering drugs intake were recruited. Plasma total bile acids in fasting state (FTBAs) and other parameters were tested at baseline. Then exenatide were applied to the T2DM participants for 12 weeks. FTBAs and glycemic parameters were measured again after exenatide treatment, and correlation analysis between changes of FTBAs and glycemic parameters were conducted to investigate the role of BAs in the glycemic control effect of exenatide. Results The baseline FTBAs level of T2DM patients had no significance (3.84 ± 2.06 vs. 3.87 ± 2.89, P = 0.954) compared with healthy subjects. After 12-week exenatide treatment for the T2DM patients, FTBAs were decreased from 3.84 ± 2.06 μmol/L to 3.06 ± 1.27 μmol/L (P < 0.01). The correlation analysis showed that changes of FTBAs was positively correlated with changes of FPG (r = 0.355, P < 0.05). Conclusions Our results demonstrated a decreased FTBAs level after exenatide treatment for 12 weeks, without the interference of metformin and other glucose-lowering drugs. The reduction of FTBAs might not exert a positive role in the glycemic control effect of exenatide. Trial registration Trial registration number: NCT04303819. Registered in March 11, 2020 - Retrospectively registered.
Collapse
Affiliation(s)
- Boyu Li
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongtinan Road, Chaoyang District, Beijing, 100020, China
| | - Yanjin Hu
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongtinan Road, Chaoyang District, Beijing, 100020, China
| | - Guang Wang
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongtinan Road, Chaoyang District, Beijing, 100020, China.
| | - Lihong Liu
- Department of Pharmacy, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongtinan Road, Chaoyang District, Beijing, 100020, China.
| |
Collapse
|
30
|
Stability and biological testing of taurine-conjugated bile acid antioxidant microcapsules for diabetes treatment. Ther Deliv 2020; 10:99-106. [PMID: 30729887 DOI: 10.4155/tde-2018-0034] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIM Taurine-conjugated bile acids possess positive formulation-stabilization effects, which are desirable in diabetes treatments. The taurine-conjugated bile acid, taurocholic acid (TCA), has shown promising formulation-stabilizing effects on the delivery of the antioxidant drug, probucol (PB), but success is limited due to its poor release profile. This study aimed to design new PB-TCA formulations using new polymers, and examine antioxidant and antidiabetic effects using β-cells for PB with or without TCA. MATERIALS AND METHODS Different formulations using alginate-insoluble esters of polymethylacrylate polymers encapsulating PB and TCA were developed, microencapsulated and examined for stability and biological activity. RESULTS TCA addition to new PB matrices improved osmotic and mechanical properties, and this effect was dependent on polymethylacrylate composition and concentration. CONCLUSION TCA can optimize the oral delivery of anti-diabetic compounds.
Collapse
|
31
|
Brønden A, Knop FK. Gluco-Metabolic Effects of Pharmacotherapy-Induced Modulation of Bile Acid Physiology. J Clin Endocrinol Metab 2020; 105:5601203. [PMID: 31630179 DOI: 10.1210/clinem/dgz025] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/04/2019] [Accepted: 10/04/2019] [Indexed: 02/08/2023]
Abstract
CONTEXT The discovery and characterization of the bile acid specific receptors farnesoid X receptor (FXR) and Takeda G protein-coupled receptor 5 (TGR5) have facilitated a wealth of research focusing on the link between bile acid physiology and glucose metabolism. Modulation of FXR and TGR5 activation have been demonstrated to affect the secretion of glucagon-like peptide 1, insulin, and glucagon as well as energy expenditure and gut microbiota composition, with potential beneficial effects on glucose metabolism. EVIDENCE ACQUISITION A search strategy based on literature searches in on PubMed with various combinations of the key words FXR, TGR5, agonist, apical sodium-dependent bile acid transporter (ASBT), bile acid sequestrant, metformin, and glucose metabolism has been applied to obtain material for the present review. Furthermore, manual searches including scanning of reference lists in relevant papers and conference proceedings have been performed. EVIDENCE SYNTHESIS This review provides an outline of the link between bile acid and glucose metabolism, with a special focus on the gluco-metabolic impact of treatment modalities with modulating effects on bile acid physiology; including FXR agonists, TGR5 agonists, ASBT inhibitors, bile acid sequestrants, and metformin. CONCLUSIONS Any potential beneficial gluco-metabolic effects of FXR agonists remain to be established, whereas the clinical relevance of TGR5-based treatment modalities seems limited because of substantial safety concerns of TGR5 agonists observed in animal models. The glucose-lowering effects of ASBT inhibitors, bile acid sequestrants, and metformin are at least partly mediated by modulation of bile acid circulation, which might allow an optimization of these bile acid-modulating treatment modalities. (J Clin Endocrinol Metab XX: 00-00, 2019).
Collapse
Affiliation(s)
- Andreas Brønden
- Center for Clinical M etabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
| | - Filip K Knop
- Center for Clinical M etabolic Research, Gentofte Hospital, University of Copenhagen, DK-2900 Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
- Novo Nordisk Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen N, Denmark
- Steno Diabetes Copenhagen, DK-2820 Gentofte, Denmark
| |
Collapse
|
32
|
Ahmad TR, Haeusler RA. Bile acids in glucose metabolism and insulin signalling - mechanisms and research needs. Nat Rev Endocrinol 2019; 15:701-712. [PMID: 31616073 PMCID: PMC6918475 DOI: 10.1038/s41574-019-0266-7] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/11/2019] [Indexed: 12/12/2022]
Abstract
Of all the novel glucoregulatory molecules discovered in the past 20 years, bile acids (BAs) are notable for the fact that they were hiding in plain sight. BAs were well known for their requirement in dietary lipid absorption and biliary cholesterol secretion, due to their micelle-forming properties. However, it was not until 1999 that BAs were discovered to be endogenous ligands for the nuclear receptor FXR. Since that time, BAs have been shown to act through multiple receptors (PXR, VDR, TGR5 and S1PR2), as well as to have receptor-independent mechanisms (membrane dynamics, allosteric modulation of N-acyl phosphatidylethanolamine phospholipase D). We now also have an appreciation of the range of physiological, pathophysiological and therapeutic conditions in which endogenous BAs are altered, raising the possibility that BAs contribute to the effects of these conditions on glycaemia. In this Review, we highlight the mechanisms by which BAs regulate glucose homeostasis and the settings in which endogenous BAs are altered, and provide suggestions for future research.
Collapse
Affiliation(s)
- Tiara R Ahmad
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Rebecca A Haeusler
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
33
|
Martin AM, Sun EW, Keating DJ. Mechanisms controlling hormone secretion in human gut and its relevance to metabolism. J Endocrinol 2019; 244:R1-R15. [PMID: 31751295 PMCID: PMC6892457 DOI: 10.1530/joe-19-0399] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 11/18/2019] [Indexed: 12/16/2022]
Abstract
The homoeostatic regulation of metabolism is highly complex and involves multiple inputs from both the nervous and endocrine systems. The gut is the largest endocrine organ in our body and synthesises and secretes over 20 different hormones from enteroendocrine cells that are dispersed throughout the gut epithelium. These hormones include GLP-1, PYY, GIP, serotonin, and CCK, each of whom play pivotal roles in maintaining energy balance and glucose homeostasis. Some are now the basis of several clinically used glucose-lowering and weight loss therapies. The environment in which these enteroendocrine cells exist is also complex, as they are exposed to numerous physiological inputs including ingested nutrients, circulating factors and metabolites produced from neighbouring gut microbiome. In this review, we examine the diverse means by which gut-derived hormones carry out their metabolic functions through their interactions with different metabolically important organs including the liver, pancreas, adipose tissue and brain. Furthermore, we discuss how nutrients and microbial metabolites affect gut hormone secretion and the mechanisms underlying these interactions.
Collapse
Affiliation(s)
- Alyce M Martin
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Emily W Sun
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
| | - Damien J Keating
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
- Nutrition and Metabolism, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
- Correspondence should be addressed to D J Keating:
| |
Collapse
|
34
|
Iwasaki H. Involvement of bile circulation and hepatic inflammation in glucose homeostasis in a case of primary biliary cholangitis. JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY CASE REPORTS 2019. [DOI: 10.1016/j.jecr.2019.100051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
35
|
Abstract
PURPOSE OF REVIEW The purpose of the review is to discuss recent advances in microRNA (miRNA) regulation of lipid metabolism and highlight the importance of miRNA-mediated gene regulation in dyslipidemia and fatty liver disease. This article reviews examples of miRNAs that bridge disparate metabolic pathways in the liver. For example, we highlight miRNAs that are regulated by the sterol-sensing pathway in the liver that in turn regulate cellular or systemic cholesterol, fatty acid, and glucose levels. RECENT FINDINGS The most widely studied of these miRNAs are miR-33a/b; however, we recently reported that miRNAs in the miR-183/96/182 cluster are also likely regulated by hepatic cholesterol content and mediate the observed glucose-lowering effects of the bile acid sequestrant colesevelam through the sterol-sensing pathway. In addition, several other hepatic and adipose miRNAs have been recently demonstrated to be key regulators of cellular lipid synthesis, storage, and catabolism, as well as systemic lipid metabolism. Moreover, many of these miRNAs are altered in fatty liver disease and dyslipidemia. SUMMARY miRNAs are not just fine-tuners of lipid metabolism, but critical regulatory factors in lipid homeostasis and health. Loss of these miRNA regulatory modules very likely contributes to the underlying metabolic defects observed in lipid disorders.
Collapse
Affiliation(s)
- Leslie R. Sedgeman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN. USA
| | - Danielle L. Michell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN. USA
| | - Kasey C. Vickers
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN. USA
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN. USA
| |
Collapse
|
36
|
Flynn CR, Albaugh VL, Abumrad NN. Metabolic Effects of Bile Acids: Potential Role in Bariatric Surgery. Cell Mol Gastroenterol Hepatol 2019; 8:235-246. [PMID: 31075353 PMCID: PMC6664228 DOI: 10.1016/j.jcmgh.2019.04.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 04/23/2019] [Accepted: 04/24/2019] [Indexed: 02/08/2023]
Abstract
Bariatric surgery is the most effective and durable treatment for morbid obesity, with an unexplained yet beneficial side effect of restoring insulin sensitivity and improving glycemia, often before weight loss is observed. Among the many contributing mechanisms often cited, the altered handling of intestinal bile acids is of considerable therapeutic interest. Here, we review a growing body of literature examining the metabolic effects of bile acids ranging from their physical roles in dietary fat handling within the intestine to their functions as endocrine and paracrine hormones in potentiating responses to bariatric surgery. The roles of 2 important bile acid receptors, Takeda G-protein coupled receptor (also known as G-protein coupled bile acid receptor) and farnesoid X receptor, are highlighted as is downstream signaling through glucagon-like polypeptide 1 and its cognate receptor. Additional improvements in other phenotypes and potential contributions of commensal gut bacteria, such as Akkermansia muciniphila, which are manifest after Roux-en-Y gastric bypass and other emulations, such as gallbladder bile diversion to the ileum, are also discussed.
Collapse
Affiliation(s)
- Charles R. Flynn
- Correspondence Address correspondence to: Charles R. Flynn, PhD, 1161 21st Avenue S, CCC-2308 MCN, Nashville, Tennessee 37232-2730. fax: (615) 343-6456.
| | | | | |
Collapse
|
37
|
Jin LH, Fang ZP, Fan MJ, Huang WD. Bile-ology: from bench to bedside. J Zhejiang Univ Sci B 2019; 20:414-427. [PMID: 31090267 PMCID: PMC6568232 DOI: 10.1631/jzus.b1900158] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 04/05/2019] [Indexed: 12/13/2022]
Abstract
Bile acids (BAs) are originally known as detergents essential for the digestion and absorption of lipids. In recent years, extensive research has unveiled new functions of BAs as gut hormones that modulate physiological and pathological processes, including glucose and lipid metabolism, energy expenditure, inflammation, tumorigenesis, cardiovascular disease, and even the central nervous system in addition to cholesterol homeostasis, enterohepatic protection and liver regeneration. BAs are closely linked with gut microbiota which might explain some of their crucial roles in organs. The signaling actions of BAs can also be mediated through specific nuclear receptors and membrane-bound G protein-coupled receptors. Several pharmacological agents or bariatric surgeries have demonstrated efficacious therapeutic effects on metabolic diseases through targeting BA signaling. In this mini-review, we summarize recent advances in bile-ology, focusing on its translational studies.
Collapse
Affiliation(s)
- Li-hua Jin
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- State Key Laboratory of Cellular Stress Biology; Innovation Center for Cell Signaling Network; School of Life Sciences, Xiamen University, Xiamen 361005, China
| | - Zhi-peng Fang
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Min-jie Fan
- College of Life Science, Zhejiang University, Hangzhou 310058, China
| | - Wen-dong Huang
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
38
|
Abstract
Bile acids have important roles in the regulation of lipid, glucose and energy metabolism. Metabolic diseases linked to obesity, including type 2 diabetes mellitus and non-alcoholic fatty liver disease, are associated with dysregulation of bile acid homeostasis. Here, the basic chemistry and regulation of bile acids as well as their metabolic effects will be reviewed. Changes in circulating bile acids associated with obesity and related diseases will be reviewed. Finally, pharmaceutical manipulation of bile acid homeostasis as therapy for metabolic diseases will be outlined.
Collapse
Affiliation(s)
- Emma Rose McGlone
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Stephen R Bloom
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| |
Collapse
|
39
|
Albaugh VL, Banan B, Antoun J, Xiong Y, Guo Y, Ping J, Alikhan M, Clements BA, Abumrad NN, Flynn CR. Role of Bile Acids and GLP-1 in Mediating the Metabolic Improvements of Bariatric Surgery. Gastroenterology 2019; 156:1041-1051.e4. [PMID: 30445014 PMCID: PMC6409186 DOI: 10.1053/j.gastro.2018.11.017] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/19/2018] [Accepted: 11/01/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND & AIMS Bile diversion to the ileum (GB-IL) has strikingly similar metabolic and satiating effects to Roux-en-Y gastric bypass (RYGB) in rodent obesity models. The metabolic benefits of these procedures are thought to be mediated by increased bile acids, although parallel changes in body weight and other confounding variables limit this interpretation. METHODS Global G protein-coupled bile acid receptor-1 null (Tgr5-/-) and intestinal-specific farnesoid X receptor null (FxrΔ/E) mice on high-fat diet as well as wild-type C57BL/6 and glucagon-like polypeptide 1 receptor deficient (Glp-1r-/-) mice on chow diet were characterized following GB-IL. RESULTS GB-IL induced weight loss and improved oral glucose tolerance in Tgr5-/-, but not FxrΔ/E mice fed a high-fat diet, suggesting a role for intestinal Fxr. GB-IL in wild-type, chow-fed mice prompted weight-independent improvements in glycemia and glucose tolerance secondary to augmented insulin responsiveness. Improvements were concomitant with increased levels of lymphatic GLP-1 in the fasted state and increased levels of intestinal Akkermansia muciniphila. Improvements in fasting glycemia after GB-IL were mitigated with exendin-9, a GLP-1 receptor antagonist, or cholestyramine, a bile acid sequestrant. The glucoregulatory effects of GB-IL were lost in whole-body Glp-1r-/- mice. CONCLUSIONS Bile diversion to the ileum improves glucose homeostasis via an intestinal Fxr-Glp-1 axis. Altered intestinal bile acid availability, independent of weight loss, and intestinal Akkermansia muciniphila appear to mediate the metabolic changes observed after bariatric surgery and might be manipulated for treatment of obesity and diabetes.
Collapse
Affiliation(s)
- Vance L. Albaugh
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Babak Banan
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Joseph Antoun
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Yanhua Xiong
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | - Yan Guo
- Department of Cancer Biology, Vanderbilt University, Nashville, TN
| | - Jie Ping
- Department of Cancer Biology, Vanderbilt University, Nashville, TN
| | - Muhammed Alikhan
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | | | - Naji N. Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN
| | | |
Collapse
|
40
|
Jendle J, Pöhlmann J, de Portu S, Smith-Palmer J, Roze S. Cost-Effectiveness Analysis of the MiniMed 670G Hybrid Closed-Loop System Versus Continuous Subcutaneous Insulin Infusion for Treatment of Type 1 Diabetes. Diabetes Technol Ther 2019; 21:110-118. [PMID: 30785311 DOI: 10.1089/dia.2018.0328] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hybrid closed-loop (HCL) systems combine continuous glucose monitoring with continuous subcutaneous insulin infusion (CSII) to continuously self-adjust basal insulin delivery. Relative to CSII, HCL improves glycemic control and reduces the risk of hypoglycemia but has higher acquisition costs. The aim of this analysis was to assess the cost-effectiveness of the MiniMed™ 670G HCL system versus CSII in people with type 1 diabetes (T1D) in Sweden. METHODS Cost-effectiveness analysis, from a societal perspective, was performed over patient lifetimes using the IQVIA CORE Diabetes Model. Clinical data were sourced from a study comparing the MiniMed 670G system with CSII in people with T1D. Cost data, expressed in 2018 Swedish krona (SEK), were obtained from Swedish reference prices and published literature. RESULTS The MiniMed 670G system was associated with a quality-adjusted life-year (QALY) gain of 1.90 but higher overall costs versus CSII, leading to an incremental cost-effectiveness ratio (ICER) of SEK 164,236 per QALY gained. Use of the HCL system resulted in a lower cumulative incidence of diabetes-related complications. Higher HCL system acquisition costs were partially offset by reduced complication costs and productivity losses. In people with T1D poorly controlled at baseline, the MiniMed 670G system was associated with 2.25 incremental QALYs versus CSII, yielding an ICER of SEK 15,830 per QALY gained. CONCLUSIONS The MiniMed 670G system was associated with clinical benefits and quality-of-life improvements in people with T1D relative to CSII. At a willingness-to-pay threshold of SEK 300,000 per QALY gained, this HCL system likely represents a cost-effective treatment option for people with T1D in Sweden.
Collapse
Affiliation(s)
- Johan Jendle
- 1 Faculty of Medical Sciences, Örebro University, Örebro, Sweden
| | - Johannes Pöhlmann
- 2 Ossian Health Economics and Communications GmbH, Basel, Switzerland
| | - Simona de Portu
- 3 Medtronic International Trading Sàrl, Tolochenaz, Switzerland
| | | | | |
Collapse
|
41
|
Donkers JM, Roscam Abbing RLP, van de Graaf SFJ. Developments in bile salt based therapies: A critical overview. Biochem Pharmacol 2018; 161:1-13. [PMID: 30582898 DOI: 10.1016/j.bcp.2018.12.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Accepted: 12/20/2018] [Indexed: 01/06/2023]
Abstract
Bile acids, amphipathic molecules known for their facilitating role in fat absorption, are also recognized as signalling molecules acting via nuclear and membrane receptors. Of the bile acid-activated receptors, the Farnesoid X Receptor (FXR) and the G protein-coupled bile acid receptor-1 (Gpbar1 or TGR5) have been studied most extensively. Bile acid signaling is critical in the regulation of bile acid metabolism itself, but it also plays a significant role in glucose, lipid and energy metabolism. Activation of FXR and TGR5 leads to reduced hepatic bile salt load, improved insulin sensitivity and glucose regulation, increased energy expenditure, and anti-inflammatory effects. These beneficial effects render bile acid signaling an interesting therapeutic target for the treatment of diseases such as cholestasis, non-alcoholic fatty liver disease, and diabetes. Here, we summarize recent findings on bile acid signaling and discuss potential and current limitations of bile acid receptor agonist and modulators of bile acid transport as future therapeutics for a wide-spectrum of diseases.
Collapse
Affiliation(s)
- Joanne M Donkers
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam, the Netherlands
| | - Reinout L P Roscam Abbing
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam, the Netherlands
| | - Stan F J van de Graaf
- Amsterdam UMC, University of Amsterdam, Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology and Metabolism, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Amsterdam Gastroenterology and Metabolism, Amsterdam, the Netherlands.
| |
Collapse
|
42
|
Kosmas CE, Silverio D, Sourlas A, Garcia F, Montan PD, Guzman E. Impact of lipid-lowering therapy on glycemic control and the risk for new-onset diabetes mellitus. Drugs Context 2018; 7:212562. [PMID: 30515229 PMCID: PMC6267678 DOI: 10.7573/dic.212562] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/04/2018] [Accepted: 11/06/2018] [Indexed: 12/24/2022] Open
Abstract
Lipid-lowering therapy is used very commonly nowadays not only for the optimization of the lipid profile but also to reduce cardiovascular risk. However, some studies have linked the use of certain lipid-lowering agents with an increased risk for impaired glycemic control and new-onset diabetes mellitus, a condition well established as an important risk factor for cardiovascular disease. On the other hand, some other lipid-lowering agents have been shown to have a beneficial effect on glucose metabolism. Profound knowledge of these differences would enable the clinician to choose the right lipid-lowering medication for each individual patient, so that the benefits would outweigh the risk of side effects. This review aims to present and discuss the clinical and scientific data pertaining to the impact of lipid-lowering therapy on glycemic control and the risk for new-onset diabetes mellitus.
Collapse
Affiliation(s)
- Constantine E Kosmas
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Bronx, NY, USA
| | - Delia Silverio
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY, USA
| | | | - Frank Garcia
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY, USA
| | - Peter D Montan
- Cardiology Clinic, Cardiology Unlimited, PC, New York, NY, USA
| | - Eliscer Guzman
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Bronx, NY, USA
| |
Collapse
|
43
|
Sedgeman LR, Beysen C, Allen RM, Ramirez Solano MA, Turner SM, Vickers KC. Intestinal bile acid sequestration improves glucose control by stimulating hepatic miR-182-5p in type 2 diabetes. Am J Physiol Gastrointest Liver Physiol 2018; 315:G810-G823. [PMID: 30160993 PMCID: PMC6415711 DOI: 10.1152/ajpgi.00238.2018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Colesevelam is a bile acid sequestrant approved to treat both hyperlipidemia and type 2 diabetes, but the mechanism for its glucose-lowering effects is not fully understood. The aim of this study was to investigate the role of hepatic microRNAs (miRNAs) as regulators of metabolic disease and to investigate the link between the cholesterol and glucose-lowering effects of colesevelam. To quantify the impact of colesevelam treatment in rodent models of diabetes, metabolic studies were performed in Zucker diabetic fatty (ZDF) rats and db/db mice. Colesevelam treatments significantly decreased plasma glucose levels and increased glycolysis in the absence of changes to insulin levels in ZDF rats and db/db mice. High-throughput sequencing and real-time PCR were used to quantify hepatic miRNA and mRNA changes, and the cholesterol-sensitive miR-96/182/183 cluster was found to be significantly increased in livers from ZDF rats treated with colesevelam compared with vehicle controls. Inhibition of miR-182 in vivo attenuated colesevelam-mediated improvements to glycemic control in db/db mice. Hepatic expression of mediator complex subunit 1 (MED1), a nuclear receptor coactivator, was significantly decreased with colesevelam treatments in db/db mice, and MED1 was experimentally validated to be a direct target of miR-96/182/183 in humans and mice. In summary, these results support that colesevelam likely improves glycemic control through hepatic miR-182-5p, a mechanism that directly links cholesterol and glucose metabolism. NEW & NOTEWORTHY Colesevelam lowers systemic glucose levels in Zucker diabetic fatty rats and db/db mice and increases hepatic levels of the sterol response element binding protein 2-responsive microRNA cluster miR-96/182/183. Inhibition of miR-182 in vivo reverses the glucose-lowering effects of colesevelam in db/db mice. Mediator complex subunit 1 (MED1) is a novel, direct target of the miR-96/182/183 cluster in mice and humans.
Collapse
Affiliation(s)
- Leslie R. Sedgeman
- 1Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | | | - Ryan M. Allen
- 3Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | - Kasey C. Vickers
- 1Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee,3Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
44
|
Nerild HH, Christensen MB, Knop FK, Brønden A. Preclinical discovery and development of colesevelam for the treatment of type 2 diabetes. Expert Opin Drug Discov 2018; 13:1161-1167. [DOI: 10.1080/17460441.2018.1538206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Henriette Holst Nerild
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Mikkel Bring Christensen
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Filip Krag Knop
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Andreas Brønden
- Clinical Metabolic Physiology, Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| |
Collapse
|
45
|
Mooranian A, Negrulj R, Takechi R, Mamo J, Al-Sallami H, Al-Salami H. The biological effects of the hypolipidaemic drug probucol microcapsules fed daily for 4 weeks, to an insulin-resistant mouse model: potential hypoglycaemic and anti-inflammatory effects. Drug Deliv Transl Res 2018; 8:543-551. [PMID: 29313296 DOI: 10.1007/s13346-017-0473-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Probucol (PB) is an hypolipidaemic drug with potential antidiabetic effects. We showed recently using in vitro studies that when PB was incorporated with stabilising lipophilic bile acids and microencapsulated using the polymer sodium alginate, the microcapsules showed good stability but poor and irregular PB release. This suggests that PB microcapsules may exhibit better release profile and hence better absorption, if more hydrophilic bile acids were used, such as ursodeoxycholic acid (UDCA). Accordingly, this study aimed to produce PB-UDCA microcapsules and examine PB absorption and antidiabetic effects in our mouse-model of insulin-resistance and diabetes (fed high-fat diet; HFD). The study also aimed to examine the effects of the microcapsules on the bile acid profile. Healthy mice (fed low-fat diet; LFD) were used as control. Seventy mice were randomly allocated into seven equal groups: LFD, HFD given empty microcapsules, HFD given metformin (M), HFD given standard-dose probucol (PB-SD), HFD given high-dose probucol (PB-H), HFD given UDCA microcapsules and HFD given PB-UDCA microcapsules. Blood glucose (BG), inflammatory biomarkers (TNF-α, IFN-γ, IL-1β, IL-6, IL-10, IL-12 and IL-17), plasma cholesterol, non-esterified fatty acids and triglycerides were analysed together with plasma bile acid and probucol concentrations. PB-UDCA microcapsules reduced BG in HFD mice, but did not reduce inflammation or improve lipid profile, compared with positive control (HFD) group. Although PB-UDCA microcapsules did not exert hypolipidaemic or antiinflammatory effects, they resulted in significant hypoglycaemic effects in a mouse model of insulin resistance, which suggests potential applications in insulin-resistance and glucose haemostasis.
Collapse
Affiliation(s)
- Armin Mooranian
- Biotechnology and Drug Development Research Laboratory, School of Pharmacy, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Rebecca Negrulj
- Biotechnology and Drug Development Research Laboratory, School of Pharmacy, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - Ryu Takechi
- School of Public Health, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | - John Mamo
- School of Public Health, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia
| | | | - Hani Al-Salami
- Biotechnology and Drug Development Research Laboratory, School of Pharmacy, Curtin Health Innovation Research Institute, Curtin University, Perth, WA, Australia.
| |
Collapse
|
46
|
Bustos AY, Font de Valdez G, Fadda S, Taranto MP. New insights into bacterial bile resistance mechanisms: the role of bile salt hydrolase and its impact on human health. Food Res Int 2018; 112:250-262. [DOI: 10.1016/j.foodres.2018.06.035] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/14/2018] [Accepted: 06/18/2018] [Indexed: 01/18/2023]
|
47
|
Brønden A, Mikkelsen K, Sonne DP, Hansen M, Våben C, Gabe MN, Rosenkilde M, Tremaroli V, Wu H, Bäckhed F, Rehfeld JF, Holst JJ, Vilsbøll T, Knop FK. Glucose-lowering effects and mechanisms of the bile acid-sequestering resin sevelamer. Diabetes Obes Metab 2018; 20:1623-1631. [PMID: 29493868 DOI: 10.1111/dom.13272] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/19/2018] [Accepted: 02/23/2018] [Indexed: 12/18/2022]
Abstract
AIMS Sevelamer, a non-absorbable amine-based resin used for treatment of hyperphosphataemia, has been demonstrated to have a marked bile acid-binding potential alongside beneficial effects on lipid and glucose metabolism. The aim of this study was to investigate the glucose-lowering effect and mechanism(s) of sevelamer in patients with type 2 diabetes. MATERIALS AND METHODS In this double-blinded randomized controlled trial, we randomized 30 patients with type 2 diabetes to sevelamer (n = 20) or placebo (n = 10). Participants were subjected to standardized 4-hour liquid meal tests at baseline and after 7 days of treatment. The main outcome measure was plasma glucagon-like peptide-1 excursions as measured by area under the curve. In addition, blood was sampled for measurements of glucose, lipids, glucose-dependent insulinotropic polypeptide, C-peptide, glucagon, fibroblast growth factor-19, cholecystokinin and bile acids. Assessments of gastric emptying, resting energy expenditure and gut microbiota composition were performed. RESULTS Sevelamer elicited a significant placebo-corrected reduction in plasma glucose with concomitant reduced fibroblast growth factor-19 concentrations, increased de novo synthesis of bile acids, a shift towards a more hydrophilic bile acid pool and increased lipogenesis. No glucagon-like peptide-1-mediated effects on insulin, glucagon or gastric emptying were evident, which points to a limited contribution of this incretin hormone to the glucose-lowering effect of sevelamer. Furthermore, no sevelamer-mediated effects on gut microbiota composition or resting energy expenditure were observed. CONCLUSIONS Sevelamer reduced plasma glucose concentrations in patients with type 2 diabetes by mechanisms that seemed to involve decreased intestinal and hepatic bile acid-mediated farnesoid X receptor activation.
Collapse
Affiliation(s)
- Andreas Brønden
- Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Kristian Mikkelsen
- Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - David P Sonne
- Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Morten Hansen
- Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Christoffer Våben
- Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
| | - Maria N Gabe
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mette Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Valentina Tremaroli
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Hao Wu
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Bäckhed
- The Wallenberg Laboratory, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Filip K Knop
- Steno Diabetes Center Copenhagen, Gentofte Hospital, University of Copenhagen, Hellerup, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
48
|
Bauer PV, Duca FA, Waise TMZ, Dranse HJ, Rasmussen BA, Puri A, Rasti M, O'Brien CA, Lam TKT. Lactobacillus gasseri in the Upper Small Intestine Impacts an ACSL3-Dependent Fatty Acid-Sensing Pathway Regulating Whole-Body Glucose Homeostasis. Cell Metab 2018. [PMID: 29514066 DOI: 10.1016/j.cmet.2018.01.013] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Long-chain acyl-CoA synthetase (ACSL)-dependent upper small intestinal lipid metabolism activates pre-absorptive pathways to regulate metabolic homeostasis, but whether changes in the upper small intestinal microbiota alter specific fatty acid-dependent pathways to impact glucose homeostasis remains unknown. We here first find that upper small intestinal infusion of Intralipid, oleic acid, or linoleic acid pre-absorptively increases glucose tolerance and lowers glucose production in rodents. High-fat feeding impairs pre-absorptive fatty acid sensing and reduces upper small intestinal Lactobacillus gasseri levels and ACSL3 expression. Transplantation of healthy upper small intestinal microbiota to high-fat-fed rodents restores L. gasseri levels and fatty acid sensing via increased ACSL3 expression, while L. gasseri probiotic administration to non-transplanted high-fat-fed rodents is sufficient to restore upper small intestinal ACSL3 expression and fatty acid sensing. In summary, we unveil a glucoregulatory role of upper small intestinal L. gasseri that impacts an ACSL3-dependent glucoregulatory fatty acid-sensing pathway.
Collapse
Affiliation(s)
- Paige V Bauer
- Toronto General Hospital Research Institute, UHN, MaRS Centre, Toronto Medical Discovery Tower, Room 10-705, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Frank A Duca
- Toronto General Hospital Research Institute, UHN, MaRS Centre, Toronto Medical Discovery Tower, Room 10-705, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - T M Zaved Waise
- Toronto General Hospital Research Institute, UHN, MaRS Centre, Toronto Medical Discovery Tower, Room 10-705, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Helen J Dranse
- Toronto General Hospital Research Institute, UHN, MaRS Centre, Toronto Medical Discovery Tower, Room 10-705, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Brittany A Rasmussen
- Toronto General Hospital Research Institute, UHN, MaRS Centre, Toronto Medical Discovery Tower, Room 10-705, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Akshita Puri
- Princess Margaret Cancer Centre, UHN, Toronto, ON M5G 2M9, Canada
| | - Mozhgan Rasti
- Toronto General Hospital Research Institute, UHN, MaRS Centre, Toronto Medical Discovery Tower, Room 10-705, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Catherine A O'Brien
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Princess Margaret Cancer Centre, UHN, Toronto, ON M5G 2M9, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Tony K T Lam
- Toronto General Hospital Research Institute, UHN, MaRS Centre, Toronto Medical Discovery Tower, Room 10-705, 101 College Street, Toronto, ON M5G 1L7, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada; Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada.
| |
Collapse
|
49
|
Brønden A, Albér A, Rohde U, Gasbjerg LS, Rehfeld JF, Holst JJ, Vilsbøll T, Knop FK. The bile acid-sequestering resin sevelamer eliminates the acute GLP-1 stimulatory effect of endogenously released bile acids in patients with type 2 diabetes. Diabetes Obes Metab 2018; 20:362-369. [PMID: 28786523 DOI: 10.1111/dom.13080] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 08/01/2017] [Accepted: 08/02/2017] [Indexed: 12/27/2022]
Abstract
AIMS Discovery of the specific bile acid receptors farnesoid X receptor (FXR) and Takeda G protein-coupled receptor 5 (TGR5) in enteroendocrine L cells has prompted research focusing on the impact of bile acids on glucagon-like peptide-1 (GLP-1) secretion and glucose metabolism. The aim of the present study was to assess the GLP-1 secretory and gluco-metabolic effects of endogenously released bile, with and without concomitant administration of the bile acid-sequestering resin, sevelamer, in patients with type 2 diabetes. MATERIALS AND METHODS We performed a randomized, placebo-controlled, double-blinded cross-over study including 15 metformin-treated patients with type 2 diabetes. During 4 experimental study days, either sevelamer 3200 mg or placebo in combination with intravenous infusion of cholecystokinin (CCK) (0.4 pmol sulfated CCK-8/kg/min) or saline was administered in randomized order. The primary endpoint was plasma GLP-1 excursions as measured by incremental area under the curve. Secondary endpoints included plasma responses of glucose, triglycerides, insulin, CCK, fibroblast growth factor-19 and 7α-hydroxy-4-cholesten-3-one (C4). In addition, gallbladder dynamics, gastric emptying, resting energy expenditure, appetite and ad libitum food intake were assessed. RESULTS CCK-mediated gallbladder emptying was demonstrated to elicit a significant induction of GLP-1 secretion compared to saline, whereas concomitant single-dose administration of the bile acid sequestrant sevelamer was shown to eliminate the acute bile acid-induced increase in plasma GLP-1 excursions. CONCLUSIONS Single-dose administration of sevelamer eliminated bile acid-mediated GLP-1 secretion in patients with type 2 diabetes, which could be explained by reduced bile acid stimulation of the basolaterally localized TGR5 on enteroendocrine L cells.
Collapse
Affiliation(s)
- Andreas Brønden
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Anders Albér
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ulrich Rohde
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Laerke S Gasbjerg
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens F Rehfeld
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Steno Diabetes Center Copenhagen, University of Copenhagen, Copenhagen, Denmark
| | - Filip K Knop
- Center for Diabetes Research, Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
50
|
Molinaro A, Wahlström A, Marschall HU. Role of Bile Acids in Metabolic Control. Trends Endocrinol Metab 2018; 29:31-41. [PMID: 29195686 DOI: 10.1016/j.tem.2017.11.002] [Citation(s) in RCA: 311] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/04/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023]
Abstract
Bile acids are endocrine molecules that in addition to facilitating the absorption of fat-soluble nutrients regulate numerous metabolic processes, including glucose, lipid, and energy homeostasis. The signaling actions of bile acids are mediated through specific bile-acid-activated nuclear and membrane-bound receptors. These receptors are not only expressed by tissues within the enterohepatic circulation such as the liver and the intestine, but also in other organs where bile acids mediate their systemic actions. In this review, we discuss bile acid signaling and the interplay with the gut microbiota in the pathophysiology of obesity, type 2 diabetes, and non-alcoholic fatty liver disease, and the role of surgical and pharmacological interventions on bile acid profiles and metabolism.
Collapse
Affiliation(s)
- Antonio Molinaro
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, S-413 45 Gothenburg, Sweden
| | - Annika Wahlström
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, S-413 45 Gothenburg, Sweden
| | - Hanns-Ulrich Marschall
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Sahlgrenska Academy, University of Gothenburg, S-413 45 Gothenburg, Sweden.
| |
Collapse
|