101
|
Shah FA, Mahmud H, Gallego-Martin T, Jurczak MJ, O’Donnell CP, McVerry BJ. Therapeutic Effects of Endogenous Incretin Hormones and Exogenous Incretin-Based Medications in Sepsis. J Clin Endocrinol Metab 2019; 104:5274-5284. [PMID: 31216011 PMCID: PMC6763279 DOI: 10.1210/jc.2019-00296] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/13/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Sepsis, a complex disorder characterized by a dysregulated immune response to an inciting infection, affects over one million Americans annually. Dysglycemia during sepsis hospitalization confers increased risk of organ dysfunction and death, and novel targets for the treatment of sepsis and maintenance of glucose homeostasis are needed. Incretin hormones are secreted by enteroendocrine cells in response to enteral nutrients and potentiate insulin release from pancreatic β cells in a glucose-dependent manner, thereby reducing the risk of insulin-induced hypoglycemia. Incretin hormones also reduce systemic inflammation in preclinical studies, but studies of incretins in the setting of sepsis are limited. METHODS In this bench-to-bedside mini-review, we detail the evidence to support incretin hormones as a therapeutic target in patients with sepsis. We performed a PubMed search using the medical subject headings "incretins," "glucagon-like peptide-1," "gastric inhibitory peptide," "inflammation," and "sepsis." RESULTS Incretin-based therapies decrease immune cell activation, inhibit proinflammatory cytokine release, and reduce organ dysfunction and mortality in preclinical models of sepsis. Several small clinical trials in critically ill patients have suggested potential benefit in glycemic control using exogenous incretin infusions, but these studies had limited power and were performed in mixed populations. Further clinical studies examining incretins specifically in septic populations are needed. CONCLUSIONS Targeting the incretin hormone axis in sepsis may provide a means of not only promoting euglycemia in sepsis but also attenuating the proinflammatory response and improving clinical outcomes.
Collapse
Affiliation(s)
- Faraaz Ali Shah
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Veteran Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
- Correspondence and Reprint Requests: Faraaz Ali Shah, MD, MPH, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh Medical Center, 3459 Fifth Avenue NW, 628 MUH, Pittsburgh, Pennsylvania 15213. E-mail:
| | - Hussain Mahmud
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Teresa Gallego-Martin
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Michael J Jurczak
- Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher P O’Donnell
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bryan J McVerry
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- Center for Medicine and the Microbiome, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
102
|
Glucagon-like peptide-1 receptor agonist dulaglutide prevents ox-LDL-induced adhesion of monocytes to human endothelial cells: An implication in the treatment of atherosclerosis. Mol Immunol 2019; 116:73-79. [PMID: 31630078 DOI: 10.1016/j.molimm.2019.09.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 09/09/2019] [Accepted: 09/29/2019] [Indexed: 02/06/2023]
Abstract
Atherosclerosis is a common comorbidity of type II diabetes and a leading cause of death worldwide. The presence of oxidized low-density lipoprotein (ox-LDL) drives atherogenesis by inducing oxidative stress, mitochondrial dysfunction, expression of proinflammatory cytokines and chemokines including interleukin (IL)-1β, IL-6, and monocyte chemoattractant protein 1 (MCP-1), adhesion molecules including vascular cellular adhesion molecule 1 (VCAM-1) and E-selectin, and downregulating expression of the Krüppel-like factor 2 (KLF2) transcription factor. Importantly, ox-LDL induced the attachment of THP-1 monocytes to endothelial cells. In the present study, we demonstrate for the first time that the specific glucagon-like peptide 1 receptor (GLP-1R) agonist dulaglutide may prevent these atherosclerotic effects of ox-LDL by preventing suppression of KLF2 by p53 protein in human aortic endothelial cells. KLF2 has been shown to play a major role in protecting vascular endothelial cells from damage induced by ox-LDL and oscillatory shear, and therefore, therapies capable of mediating KLF2 signaling may be an attractive treatment option for preventing the development and progression of atherosclerosis.
Collapse
|
103
|
Gastaldelli A, Cusi K. From NASH to diabetes and from diabetes to NASH: Mechanisms and treatment options. JHEP Rep 2019; 1:312-328. [PMID: 32039382 PMCID: PMC7001557 DOI: 10.1016/j.jhepr.2019.07.002] [Citation(s) in RCA: 243] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/14/2019] [Accepted: 07/11/2019] [Indexed: 02/06/2023] Open
Abstract
The worldwide prevalence of non-alcoholic fatty liver disease (NAFLD) is estimated to have reached 25% or more in adults. NAFLD is prevalent in obese individuals, but may also affect non-obese insulin-resistant individuals. NAFLD is associated with a 2- to 3-fold increased risk of developing type 2 diabetes (T2D), which may be higher in patients with more severe liver disease - fibrosis increases this risk. In NAFLD, not only the close association with obesity, but also the impairment of many metabolic pathways, including decreased hepatic insulin sensitivity and insulin secretion, increase the risk of developing T2D and related comorbidities. Conversely, patients with diabetes have a higher prevalence of steatohepatitis, liver fibrosis and end-stage liver disease. Genetics and mechanisms involving dysfunctional adipose tissue, lipotoxicity and glucotoxicity appear to play a role. In this review, we discuss the altered pathophysiological mechanisms that underlie the development of T2D in NAFLD and vice versa. Although there is no approved therapy for the treatment of NASH, we discuss pharmacological agents currently available to treat T2D that could potentially be useful for the management of NASH.
Collapse
Affiliation(s)
- Amalia Gastaldelli
- Cardiometabolic Risk Unit, Institute of Clinical Physiology, National Research Council, Pisa, Italy
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, The University of Florida, and Malcom Randall Veterans Administration Medical Center, Gainesville, Florida
| |
Collapse
|
104
|
Bashier A, Bin Hussain A, Abdelgadir E, Alawadi F, Sabbour H, Chilton R. Consensus recommendations for management of patients with type 2 diabetes mellitus and cardiovascular diseases. Diabetol Metab Syndr 2019; 11:80. [PMID: 31572499 PMCID: PMC6761728 DOI: 10.1186/s13098-019-0476-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 09/19/2019] [Indexed: 02/12/2023] Open
Abstract
The recent American Diabetes Association and the European Association for the Study of Diabetes guideline mentioned glycaemia management in type 2 diabetes mellitus (T2DM) patients with cardiovascular diseases (CVDs); however, it did not cover the treatment approaches for patients with T2DM having a high risk of CVD, and treatment and screening approaches for CVDs in patients with concomitant T2DM. This consensus guideline undertakes the data obtained from all the cardiovascular outcome trials (CVOTs) to propose approaches for the T2DM management in presence of CV comorbidities. For patients at high risk of CVD, metformin is the drug of choice to manage the T2DM to achieve a patient specific HbA1c target. In case of established CVD, a combination of glucagon-like peptide-1 receptor agonist with proven CV benefits is recommended along with metformin, while for chronic kidney disease or heart failure, a sodium-glucose transporter proteins-2 inhibitor with proven benefit is advised. This document also summarises various screening and investigational approaches for the major CV events with their accuracy and specificity along with the treatment guidance to assist the healthcare professionals in selecting the best management strategies for every individual. Since lifestyle modification and management plays an important role in maintaining the effectiveness of the pharmacological therapies, authors of this consensus recommendation have also briefed on the patient-centric non-pharmacological management of T2DM and CVD.
Collapse
Affiliation(s)
- Alaaeldin Bashier
- Department of Endocrinology, Dubai Health Authority, Dubai Hospital, P.O. Box 94132, Dubai, UAE
| | - Azza Bin Hussain
- Department of Endocrinology, Dubai Health Authority, Dubai Hospital, P.O. Box 94132, Dubai, UAE
| | - Elamin Abdelgadir
- Department of Endocrinology, Dubai Health Authority, Dubai Hospital, P.O. Box 94132, Dubai, UAE
| | - Fatheya Alawadi
- Department of Endocrinology, Dubai Health Authority, Dubai Hospital, P.O. Box 94132, Dubai, UAE
| | - Hani Sabbour
- Cleveland Clinic Abu Dhabi, Heart and Vascular Institute, Al Maryah Island, Abu Dhabi, UAE
| | - Robert Chilton
- Division of Cardiology, University of Texas Health Science Center, Audie L Murphy VA Hospital, San Antonio, TX USA
| |
Collapse
|
105
|
Mathiesen DS, Bagger JI, Bergmann NC, Lund A, Christensen MB, Vilsbøll T, Knop FK. The Effects of Dual GLP-1/GIP Receptor Agonism on Glucagon Secretion-A Review. Int J Mol Sci 2019; 20:E4092. [PMID: 31443356 PMCID: PMC6747202 DOI: 10.3390/ijms20174092] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 08/16/2019] [Accepted: 08/20/2019] [Indexed: 02/07/2023] Open
Abstract
The gut-derived incretin hormones glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are secreted after meal ingestion and work in concert to promote postprandial insulin secretion. Furthermore, GLP-1 inhibits glucagon secretion when plasma glucose concentrations are above normal fasting concentrations while GIP acts glucagonotropically at low glucose levels. A dual incretin receptor agonist designed to co-activate GLP-1 and GIP receptors was recently shown to elicit robust improvements of glycemic control (mean haemoglobin A1c reduction of 1.94%) and massive body weight loss (mean weight loss of 11.3 kg) after 26 weeks of treatment with the highest dose (15 mg once weekly) in a clinical trial including overweight/obese patients with type 2 diabetes. Here, we describe the mechanisms by which the two incretins modulate alpha cell secretion of glucagon, review the effects of co-administration of GLP-1 and GIP on glucagon secretion, and discuss the potential role of glucagon in the therapeutic effects observed with novel unimolecular dual GLP-1/GIP receptor agonists. For clinicians and researchers, this manuscript offers an understanding of incretin physiology and pharmacology, and provides mechanistic insight into future antidiabetic and obesity treatments.
Collapse
Affiliation(s)
- David S Mathiesen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
| | - Jonatan I Bagger
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
- Steno Diabetes Center Copenhagen, 2820 Gentofte, Denmark
| | - Natasha C Bergmann
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
| | - Asger Lund
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
| | - Mikkel B Christensen
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
- Department of Clinical Pharmacology, Bispebjerg Hospital, University of Copenhagen, 2400 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Tina Vilsbøll
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark
- Steno Diabetes Center Copenhagen, 2820 Gentofte, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Filip K Knop
- Center for Clinical Metabolic Research, Gentofte Hospital, University of Copenhagen, 2900 Hellerup, Denmark.
- Steno Diabetes Center Copenhagen, 2820 Gentofte, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.
| |
Collapse
|
106
|
Knudsen LB. Inventing Liraglutide, a Glucagon-Like Peptide-1 Analogue, for the Treatment of Diabetes and Obesity. ACS Pharmacol Transl Sci 2019; 2:468-484. [PMID: 32259078 DOI: 10.1021/acsptsci.9b00048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Indexed: 01/08/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) has been in focus since the early 1980s as a long looked for incretin hormone, released from the gastrointestinal tract and with an important effect on glucose-dependent insulin secretion, providing efficient glucose lowering, with little risk for hypoglycemia. The enzyme dipeptidyl peptidase-4 (DPP-4) degrades GLP-1 very fast, and the remaining metabolite is cleared rapidly by the kidneys. Liraglutide is a fatty acid acylated analogue of GLP-1 that provides efficacy for 24 h/day. The mechanism of action for liraglutide is reviewed in detail with focus on pancreatic efficacy and safety, thyroid safety, and weight loss mechanism. Evolving science hypothesizes that GLP-1 has important effects on atherosclerosis, relevant for the cardiovascular benefit seen in the treatment of diabetes and obesity. Also, GLP-1 may be relevant in neurodegenerative diseases.
Collapse
Affiliation(s)
- Lotte Bjerre Knudsen
- Global Drug Discovery, Novo Nordisk, Novo Nordisk Park, DK-2760 Maaloev, Denmark
| |
Collapse
|
107
|
Yusta B, Matthews D, Koehler JA, Pujadas G, Kaur KD, Drucker DJ. Localization of Glucagon-Like Peptide-2 Receptor Expression in the Mouse. Endocrinology 2019; 160:1950-1963. [PMID: 31237617 PMCID: PMC6656427 DOI: 10.1210/en.2019-00398] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022]
Abstract
Glucagon-like peptide-2 (GLP-2), secreted from enteroendocrine cells, attenuates gut motility, enhances barrier function, and augments nutrient absorption, actions mediated by a single GLP-2 receptor (GLP-2R). Despite extensive analyses, the precise distribution and cellular localization of GLP-2R expression remains controversial, confounded by the lack of suitable GLP-2R antisera. Here, we reassessed murine Glp2r expression using regular and real-time quantitative PCR (qPCR), in situ hybridization (ISH), and a Glp2rLacZ reporter mouse. Glp2r mRNA expression was detected from the stomach to the rectum and most abundant in the jejunum. Glp2r transcripts were also detected in cerebral cortex, mesenteric lymph nodes, gallbladder, urinary bladder, and mesenteric fat. Surprisingly, Glp2r mRNA was found in testis by qPCR at levels similar to jejunum. However, the testis Glp2r transcripts, detected by different primer pairs and qPCR, lacked 5' mRNA coding sequences, and only a minute proportion of them corresponded to full-length Glp2r mRNA. Within the gut, Glp2r-driven LacZ expression was localized to enteric neurons and lamina propria stromal cells, findings confirmed by ISH analysis of the endogenous Glp2r mRNA. Unexpectedly, vascular Glp2rLacZ expression was localized to mesenteric veins and not arteries. Moreover, mesenteric fat Glp2rLacZ expression was detected within blood vessels and not adipocytes. Reporter LacZ expression was not detected in all tissues expressing an endogenous Glp2r transcript, such as gallbladder, urinary bladder, and mesenteric lymph nodes. Collectively, these findings extend our understanding of the cellular domains of Glp2r expression and highlight limitations inherent in application of commonly used technologies to infer analysis of gene expression.
Collapse
Affiliation(s)
- Bernardo Yusta
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, Canada
| | - Dianne Matthews
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, Canada
| | - Jacqueline A Koehler
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, Canada
| | - Gemma Pujadas
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, Canada
| | - Kiran Deep Kaur
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, Canada
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, Canada
- Correspondence: Daniel J. Drucker, MD, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, 600 University Avenue, Mailbox 39, Toronto, Ontario M5G 1X5, Canada. E-mail:
| |
Collapse
|
108
|
Capozzi ME, Wait JB, Koech J, Gordon AN, Coch RW, Svendsen B, Finan B, D'Alessio DA, Campbell JE. Glucagon lowers glycemia when β-cells are active. JCI Insight 2019; 5:129954. [PMID: 31335319 DOI: 10.1172/jci.insight.129954] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Glucagon and insulin are commonly believed to have counteracting effects on blood glucose levels. However, recent studies have demonstrated that glucagon has a physiologic role to activate β-cells and enhance insulin secretion. To date, the actions of glucagon have been studied mostly in fasting or hypoglycemic states, yet it is clear that mixed-nutrient meals elicit secretion of both glucagon and insulin, suggesting that glucagon also contributes to glucose regulation in the postprandial state. We hypothesized that the elevated glycemia seen in the fed state would allow glucagon to stimulate insulin secretion and reduce blood glucose. In fact, exogenous glucagon given under fed conditions did robustly stimulate insulin secretion and lower glycemia. Exogenous glucagon given to fed Gcgr:Glp1rβcell-/- mice failed to stimulate insulin secretion or reduce glycemia, demonstrating the importance of an insulinotropic glucagon effect. The action of endogenous glucagon to reduce glycemia in the fed state was tested with administration of alanine, a potent glucagon secretagogue. Alanine raised blood glucose in fasted WT mice or fed Gcgr:Glp1rβcell-/- mice, conditions where glucagon is unable to stimulate β-cell activity. However, alanine given to fed WT mice produced a decrease in glycemia, along with elevated insulin and glucagon levels. Overall, our data support a model in which glucagon serves as an insulinotropic hormone in the fed state and complements rather than opposes insulin action to maintain euglycemia.
Collapse
Affiliation(s)
| | | | | | | | - Reilly W Coch
- Duke Molecular Physiology Institute and.,Department of Medicine, Duke University, Durham, North Carolina, USA
| | | | - Brian Finan
- Novo Nordisk Research Center, Indianapolis, Indiana, USA
| | - David A D'Alessio
- Duke Molecular Physiology Institute and.,Department of Medicine, Duke University, Durham, North Carolina, USA
| | - Jonathan E Campbell
- Duke Molecular Physiology Institute and.,Department of Medicine, Duke University, Durham, North Carolina, USA.,Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
109
|
van Eyk HJ, Blauw LL, Bizino MB, Wang Y, van Dijk KW, de Mutsert R, Smit JWA, Lamb HJ, Jazet IM, Rensen PCN. Hepatic triglyceride content does not affect circulating CETP: lessons from a liraglutide intervention trial and a population-based cohort. Sci Rep 2019; 9:9996. [PMID: 31292457 PMCID: PMC6620358 DOI: 10.1038/s41598-019-45593-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 06/06/2019] [Indexed: 12/29/2022] Open
Abstract
Cholesteryl ester transfer protein (CETP) is mainly expressed by Kupffer cells in the liver. A reduction of hepatic triglyceride content (HTGC) by pioglitazone or caloric restriction is accompanied by a decrease in circulating CETP. Since GLP-1 analogues also reduce HTGC, we assessed whether liraglutide decreases CETP. Furthermore, we investigated the association between HTGC and CETP in a population-based cohort. In a placebo-controlled trial, 50 patients with type 2 diabetes were randomly assigned to treatment with liraglutide or placebo added to standard care. In this trial and in 1,611 participants of the Netherlands Epidemiology of Obesity (NEO) study, we measured HTGC and circulating CETP by proton magnetic resonance spectroscopy and ELISA, respectively. The HTGC was decreased in the liraglutide group (-6.3%; 95%CI of difference [-9.5, -3.0]) but also in the placebo group (-4.0%; 95%CI[-6.0, -2.0]), without between-group differences. CETP was not decreased by liraglutide (-0.05 µg/mL; 95%CI[-0.13, 0.04]) or placebo (-0.04 µg/mL; 95%CI[-0.12, 0.04]). No association was present between HTGC and CETP at baseline (β: 0.002 µg/mL per %TG, 95%CI[-0.005, 0.009]) and between the changes after treatment with liraglutide (β: 0.003 µg/mL per %TG, 95%CI[-0.010, 0.017]) or placebo (β: 0.006 µg/mL per %TG, 95%CI[-0.012,0.024]). Also, in the cohort n o association between HTGC and CETP was present (β: -0.001 µg/mL per SD TG, 95%CI[-0.005, 0.003]). A reduction of HTGC after treatment with liraglutide or placebo does not decrease circulating CETP. Also, no association between HTGC and CETP was present in a large cohort. These findings indicate that circulating CETP is not determined by HTGC.Clinical Trial Registration: Clinicaltrials.gov (NCT01761318).
Collapse
Affiliation(s)
- Huub J van Eyk
- Department Medicine, Div. Endocrinology, Leiden University Medical Center (LUMC), Leiden, The Netherlands.
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands.
| | - Lisanne L Blauw
- Department Medicine, Div. Endocrinology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Department Epidemiology, LUMC, Leiden, The Netherlands
| | - Maurice B Bizino
- Department Medicine, Div. Endocrinology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Department Radiology, LUMC, Leiden, The Netherlands
| | - Yanan Wang
- Department Medicine, Div. Endocrinology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands
| | - Ko Willems van Dijk
- Department Medicine, Div. Endocrinology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands
- Department Human Genetics, LUMC, Leiden, The Netherlands
| | | | - Johannes W A Smit
- Department Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hildo J Lamb
- Department Radiology, LUMC, Leiden, The Netherlands
| | - Ingrid M Jazet
- Department Medicine, Div. Endocrinology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands
| | - Patrick C N Rensen
- Department Medicine, Div. Endocrinology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, LUMC, Leiden, The Netherlands
| |
Collapse
|
110
|
Lee YS, Riopel M, Cabrales P, Bandyopadhyay GK. Hepatocyte-specific HIF-1α ablation improves obesity-induced glucose intolerance by reducing first-pass GLP-1 degradation. SCIENCE ADVANCES 2019; 5:eaaw4176. [PMID: 31281892 PMCID: PMC6609217 DOI: 10.1126/sciadv.aaw4176] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/30/2019] [Indexed: 05/04/2023]
Abstract
The decrease in incretin effects is an important etiologic component of type 2 diabetes with unknown mechanisms. In an attempt to understand obesity-induced changes in liver oxygen homeostasis, we found that liver HIF-1α expression was increased mainly by soluble factors released from obese adipocytes, leading to decreased incretin effects. Deletion of hepatocyte HIF-1α protected obesity-induced glucose intolerance without changes in body weight, liver steatosis, or insulin resistance. In-depth mouse metabolic phenotyping revealed that obesity increased first-pass degradation of an incretin hormone GLP-1 with increased liver DPP4 expression and decreased sinusoidal blood flow rate, reducing active GLP-1 levels in peripheral circulation. Hepatocyte HIF-1α KO blocked these changes induced by obesity. Deletion of hepatocyte HIF-2α did not change liver DPP4 expression but improved hepatic steatosis. Our results identify a previously unknown pathway for obesity-induced impaired beta cell glucose response (incretin effects) and the development of glucose intolerance through inter-organ communications.
Collapse
Affiliation(s)
- Yun Sok Lee
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Matthew Riopel
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Pedro Cabrales
- Department of Engineering, University of California San Diego, La Jolla, CA 92093, USA
| | - Guatam K. Bandyopadhyay
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
111
|
Kang Z, Zeng J, Zhang T, Lin S, Gao J, Jiang C, Fan R, Yin D. Hyperglycemia induces NF-κB activation and MCP-1 expression via downregulating GLP-1R expression in rat mesangial cells: inhibition by metformin. Cell Biol Int 2019; 43:940-953. [PMID: 31136032 DOI: 10.1002/cbin.11184] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 05/25/2019] [Indexed: 12/25/2022]
Abstract
Hyperglycemia impairs glucagon-like peptide-1 receptor (GLP-1R) signaling in multiple cell types and thereby potentially attenuates the therapeutic effects of GLP-1R agonists. We hypothesized that the downregulation of GLP-1R by hyperglycemia might reduce the renal-protective effects of GLP-1R agonists in diabetic nephropathy (DN). In this study, we examined the effects of high glucose on the expression of GLP-1R and its signaling pathways in the HBZY-1 rat mesangial cell line. We found that high glucose reduced GLP-1R messenger RNA (mRNA) levels in HBZY-1 cells and in the renal cortex in db/db mice comparing with control groups. In consistence, GLP-1R agonist exendin-4 induced CREB phosphorylation was attenuated by high glucose but not low glucose treatment, which is paralleled with abrogated anti-inflammatory functions in HBZY-1 cells linked with nuclear factor-κB (NF-κB) activation. In consistence, GLP-1R inhibition aggravated the high glucose-induced activation of NF-κB and MCP-1 protein levels in cultured HBZY-1 cells while overexpression of GLP-1R opposite effects. We further proved that metformin restored high glucose-inhibited GLP-1R mRNA expression and decreased high glucose evoked inflammation in HBZY-1 cells. On the basis of these findings, we conclude that high glucose lowers GLP-1R expression and leads to inflammatory responses in mesangial cells, which can be reversed by metformin. These data support the rationale of combinative therapy of metformin with GLP-1R agonists in DN.
Collapse
Affiliation(s)
- Zhanfang Kang
- Department of Basic Medical Research, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, Guangdong, China.,Department of Basic Medical Research, Qingyuan hospital affiliated to Jinan University, Qingyuan, 511518, Guangdong, China
| | - Jianwen Zeng
- Department of Urology, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, Guangdong, China
| | - Tian Zhang
- Department of Basic Medical Research, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, Guangdong, China
| | - Shuyun Lin
- Department of Basic Medical Research, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, Guangdong, China
| | - Jun Gao
- Department of Basic Medical Research, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, Guangdong, China
| | - Chonghe Jiang
- Department of Urology, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, Guangdong, China
| | - Rongrong Fan
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, 14183, Stockholm, Sweden
| | - Dazhong Yin
- Department of Basic Medical Research, Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, Guangdong, China
| |
Collapse
|
112
|
Liraglutide alters hepatic metabolism in high-fat fed obese mice: A bioinformatic prediction and functional analysis. Meta Gene 2019. [DOI: 10.1016/j.mgene.2019.100553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
|
113
|
Kalavalapalli S, Bril F, Guingab J, Vergara A, Garrett TJ, Sunny NE, Cusi K. Impact of exenatide on mitochondrial lipid metabolism in mice with nonalcoholic steatohepatitis. J Endocrinol 2019; 241:293-305. [PMID: 31082799 DOI: 10.1530/joe-19-0007] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 04/23/2019] [Indexed: 12/15/2022]
Abstract
Exenatide (Exe) is a glucagon-like peptide (GLP)-1 receptor agonist that enhances insulin secretion and is associated with induction of satiety with weight loss. As mitochondrial dysfunction and lipotoxicity are central features of nonalcoholic steatohepatitis (NASH), we tested whether Exe improved mitochondrial function in this setting. We studied C57BL/6J mice fed for 24 weeks either a control- or high-fructose, high-trans-fat (TFD)-diet (i.e., a NASH model previously validated by our laboratory). For the final 8 weeks, mice were treated with Exe (30 µg/kg/day) or vehicle. Mitochondrial metabolism was assessed by infusion of [13C3]propionate, [3,4-13C2]glucose and NMR-based 13C-isotopomer analysis. Exenatide significantly decreased fasting plasma glucose, free fatty acids and triglycerides, as well as adipose tissue insulin resistance. Moreover, Exe reduced 23% hepatic glucose production, 15% tri-carboxylic acid (TCA) cycle flux, 20% anaplerosis and 17% pyruvate cycling resulting in a significant 31% decrease in intrahepatic triglyceride content (P = 0.02). Exenatide improved the lipidomic profile and decreased hepatic lipid byproducts associated with insulin resistance and lipotoxicity, such as diacylglycerols (TFD: 111 ± 13 vs Exe: 64 ± 13 µmol/g protein, P = 0.03) and ceramides (TFD: 1.6 ± 0.1 vs Exe: 1.3 ± 0.1 µmol/g protein, P = 0.03). Exenatide lowered expression of hepatic lipogenic genes (Srebp1C, Cd36) and genes involved in inflammation and fibrosis (Tnfa, Timp1). In conclusion, in a diet-induced mouse model of NASH, Exe ameliorates mitochondrial TCA cycle flux and significantly decreases insulin resistance, steatosis and hepatocyte lipotoxicity. This may have significant clinical implications to the potential mechanism of action of GLP-1 receptor agonists in patients with NASH. Future studies should elucidate the relative contribution of direct vs indirect mechanisms at play.
Collapse
Affiliation(s)
- Srilaxmi Kalavalapalli
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Fernando Bril
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Joy Guingab
- Department of Pathology, University of Florida, Gainesville, Florida, USA
| | - Ariana Vergara
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Timothy J Garrett
- Department of Pathology, University of Florida, Gainesville, Florida, USA
| | - Nishanth E Sunny
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, USA
| | - Kenneth Cusi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, University of Florida, Gainesville, Florida, USA
- Division of Endocrinology, Diabetes and Metabolism, Malcom Randall Veterans Administration Medical Center (VAMC), Gainesville, Florida, USA
| |
Collapse
|
114
|
Koshibu M, Mori Y, Saito T, Kushima H, Hiromura M, Terasaki M, Takada M, Fukui T, Hirano T. Antiatherogenic effects of liraglutide in hyperglycemic apolipoprotein E-null mice via AMP-activated protein kinase-independent mechanisms. Am J Physiol Endocrinol Metab 2019; 316:E895-E907. [PMID: 30860874 DOI: 10.1152/ajpendo.00511.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1RAs) exert potent glucose-lowering effects without increasing risks for hypoglycemia and weight gain. Preclinical studies have demonstrated direct antiatherogenic effects of GLP-1RAs in normoglycemic animal models; however, the underlying mechanisms in hyperglycemic conditions have not been fully clarified. Here we aimed to elucidate the role of AMP-activated protein kinase (AMPK) in antiatherogenic effects of GLP-1RAs in hyperglycemic mice. Streptozotocin-induced hyperglycemic apolipoprotein E-null mice were treated with vehicle, low-dose liraglutide (17 nmol·kg-1·day-1), or high-dose liraglutide (107 nmol·kg-1·day-1) in experiment 1 and the AMPK inhibitor dorsomorphin, dorsomorphin + low-dose liraglutide, or dorsomorphin + high-dose liraglutide in experiment 2. Four weeks after treatment, aortas were collected to assess atherosclerosis. In experiment 1, metabolic parameters were similar among the groups. Assessment of atherosclerosis revealed that high-dose liraglutide treatments reduced lipid deposition on the aortic surface and plaque volume and intraplaque macrophage accumulation at the aortic sinus. In experiment 2, liraglutide-induced AMPK phosphorylation in the aorta was abolished by dorsomorphin; however, the antiatherogenic effects of high-dose liraglutide were preserved. In cultured human umbilical vein endothelial cells, liraglutide suppressed tumor necrosis factor-induced expression of proatherogenic molecules; these effects were maintained under small interfering RNA-mediated knockdown of AMPKα1 and in the presence of dorsomorphin. Conversely, in human monocytic U937 cells, the anti-inflammatory effects of liraglutide were abolished by dorsomorphin. In conclusion, liraglutide exerted AMPK-independent antiatherogenic effects in hyperlipidemic mice with streptozotocin-induced hyperglycemia, with the possible involvement of AMPK-independent suppression of proatherogenic molecules in vascular endothelial cells.
Collapse
Affiliation(s)
- Masakazu Koshibu
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Yusaku Mori
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Tomomi Saito
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Hideki Kushima
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Munenori Hiromura
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Michishige Terasaki
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Michiya Takada
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Tomoyasu Fukui
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Tsutomu Hirano
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| |
Collapse
|
115
|
Liu J, Yang K, Yang J, Xiao W, Le Y, Yu F, Gu L, Lang S, Tian Q, Jin T, Wei R, Hong T. Liver-derived fibroblast growth factor 21 mediates effects of glucagon-like peptide-1 in attenuating hepatic glucose output. EBioMedicine 2019; 41:73-84. [PMID: 30827929 PMCID: PMC6443026 DOI: 10.1016/j.ebiom.2019.02.037] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 01/30/2019] [Accepted: 02/18/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Glucagon-like peptide-1 (GLP-1) and its based agents improve glycemic control. Although their attenuating effect on hepatic glucose output has drawn our attention for decades, the potential mechanisms remain unclear. METHODS Cytokine array kit was used to assess cytokine profiles in db/db mice and mouse primary hepatocytes treated with exenatide (exendin-4). Two diabetic mouse models (db/db and Pax6m/+) were treated with a GLP-1 analog exenatide or liraglutide. The expression and secretion of fibroblast growth factor 21 (FGF21) in the livers of diabetic mice, primary mouse and human hepatocytes, and the human hepatic cell line HepG2 treated with or without GLP-1 analog were measured. Blockage of FGF21 with neutralizing antibody or siRNA, or hepatocytes isolated from Fgf21 knockout mice were used, and the expression and activity of key enzymes in gluconeogenesis were analyzed. Serum FGF21 level was evaluated in patients with type 2 diabetes (T2D) receiving exenatide treatment. FINDINGS Utilizing the cytokine array, we identified that FGF21 secretion was upregulated by exenatide (exendin-4). Similarly, FGF21 production in hepatocytes was stimulated by exenatide or liraglutide. FGF21 blockage attenuated the inhibitory effects of the GLP-1 analogs on hepatic glucose output. Similar results were also observed in primary hepatocytes from Fgf21 knockout mice. Furthermore, exenatide treatment increased serum FGF21 level in patients with T2D, particularly in those with better glucose control. INTERPRETATION We identify that function of GLP-1 in inhibiting hepatic glucose output is mediated via the liver hormone FGF21. Thus, we provide a new extra-pancreatic mechanism by which GLP-1 regulates glucose homeostasis. FUND: National Key Research and Development Program of China, the National Natural Science Foundation of China, the Natural Science Foundation of Beijing and Peking University Medicine Seed Fund for Interdisciplinary Research.
Collapse
Affiliation(s)
- Junling Liu
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Kun Yang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | - Jin Yang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Wenhua Xiao
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | - Yunyi Le
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | - Fei Yu
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Liangbiao Gu
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Shan Lang
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Qing Tian
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China
| | - Tianru Jin
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Banting and Best Diabetes Center, University of Toronto, Toronto, Ontario, Canada
| | - Rui Wei
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China.
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, Peking University Third Hospital, Beijing, China; Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
116
|
Zhao L, Zhu C, Lu M, Chen C, Nie X, Abudukerimu B, Zhang K, Ning Z, Chen Y, Cheng J, Xia F, Wang N, Jensen MD, Lu Y. The key role of a glucagon-like peptide-1 receptor agonist in body fat redistribution. J Endocrinol 2019; 240:271-286. [PMID: 30530905 DOI: 10.1530/joe-18-0374] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 11/08/2018] [Indexed: 01/01/2023]
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are an ideal therapy for type 2 diabetes and, as of recently, for obesity. In contrast to visceral fat, subcutaneous fat appears to be protective against metabolic diseases. Here, we aimed to explore whether liraglutide, a GLP-1RA, could redistribute body fat via regulating lipid metabolism in different fat depots. After being fed a high-fat diet for 8 weeks, 50 male Wistar and Goto-Kakizaki rats were randomly divided into a normal control group, a diabetic control group, low- and high-dose liraglutide-treated groups and a diet-control group. Different doses of liraglutide (400 μg/kg/day or 1200 μg/kg/day) or an equal volume of normal saline were administered to the rats subcutaneously once a day for 12 weeks. Body composition and body fat deposition were measured by dual-energy X-ray absorptiometry and MRI. Isotope tracers were infused to explore lipid metabolism in different fat depots. Quantitative real-time PCR and Western blot analyses were conducted to evaluate the expression of adipose-related genes. The results showed that liraglutide decreased visceral fat and relatively increased subcutaneous fat. Lipogenesis was reduced in visceral white adipose tissue (WAT) but was elevated in subcutaneous WAT. Lipolysis was also attenuated, and fatty acid oxidation was enhanced. The mRNA expression levels of adipose-related genes in different tissues displayed similar trends after liraglutide treatment. In addition, the expression of browning-related genes was upregulated in subcutaneous WAT. Taken together, the results suggested that liraglutide potentially redistributes body fat and promotes browning remodeling in subcutaneous WAT to improve metabolic disorders.
Collapse
Affiliation(s)
- Li Zhao
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Chunfang Zhu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Meng Lu
- Research Center for Clinical Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Chi Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xiaomin Nie
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Buatikamu Abudukerimu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Kun Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zhiyuan Ning
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yi Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jing Cheng
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Fangzhen Xia
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | | | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
117
|
Almutairi M, Al Batran R, Ussher JR. Glucagon-like peptide-1 receptor action in the vasculature. Peptides 2019; 111:26-32. [PMID: 30227157 DOI: 10.1016/j.peptides.2018.09.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 12/14/2022]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists augment insulin secretion and are thus used clinically to improve glycemia in subjects with type 2 diabetes (T2D). As recent data reveal marked improvements in cardiovascular outcomes in T2D subjects treated with the GLP-1R agonists liraglutide and semaglutide in the LEADER and SUSTAIN-6 clinical trials respectively, there is growing interest in delineating the mechanism(s) of action for GLP-1R agonist-induced cardioprotection. Of importance, negligible GLP-1R expression in ventricular cardiac myocytes suggests that cardiac-independent actions of GLP-1R agonists may account for the reduced death rates from cardiovascular causes in T2D subjects enrolled in the LEADER trial. Conversely, vascular smooth muscle cells (VSMCs) appear to express the canonical GLP-1R, and GLP-1/GLP-1R agonists exhibit a number of salutary actions on the vascular endothelium that could potentially contribute to GLP-1R agonists directly improving cardiovascular outcomes in subjects with T2D. We review herein the described actions of GLP-1/GLP-1R agonists on the vascular endothelium, which include antiproliferative actions on VSMCs and endothelial cells, reductions in oxidative stress, and increases in nitric oxide generation. GLP-1 also increases microvascular recruitment and microvascular blood flow. Taken together, such actions may explain the antihypertensive and/or antiatherosclerotic actions attributed to GLP-1/GLP-1R agonists in preclinical and clinical studies. Nonetheless, further mechanistic studies are still necessary to determine the relative importance of such actions in accounting for reductions in macrovascular cardiovascular disease in human subjects with T2D treated with GLP-1R agonists.
Collapse
Affiliation(s)
- Malak Almutairi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Rami Al Batran
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
118
|
Zhang Y, Parajuli KR, Fava GE, Gupta R, Xu W, Nguyen LU, Zakaria AF, Fonseca VA, Wang H, Mauvais-Jarvis F, Sloop KW, Wu H. GLP-1 Receptor in Pancreatic α-Cells Regulates Glucagon Secretion in a Glucose-Dependent Bidirectional Manner. Diabetes 2019; 68:34-44. [PMID: 30389749 PMCID: PMC6302540 DOI: 10.2337/db18-0317] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 10/15/2018] [Indexed: 01/10/2023]
Abstract
Glucagon-like peptide 1 (GLP-1) is known to suppress glucagon secretion, but the mechanism by which GLP-1 exerts this effect is unclear. In this study, we demonstrated GLP-1 receptor (GLP-1R) expression in α-cells using both antibody-dependent and antibody-independent strategies. A novel α-cell-specific GLP-1R knockout (αGLP-1R-/-) mouse model was created and used to investigate its effects on glucagon secretion and glucose metabolism. Male and female αGLP-1R-/- mice both showed higher nonfasting glucagon levels than their wild-type littermates, whereas insulin and GLP-1 levels remained similar. Female αGLP-1R-/- mice exhibited mild glucose intolerance after an intraperitoneal glucose administration and showed increased glucagon secretion in response to a glucose injection compared with the wild-type animals. Furthermore, using isolated islets, we confirmed that αGLP-1R deletion did not interfere with β-cell function but affected glucagon secretion in a glucose-dependent bidirectional manner: the αGLP-1R-/- islets failed to inhibit glucagon secretion at high glucose and failed to stimulate glucagon secretion at very low glucose condition. More interestingly, the same phenomenon was recapitulated in vivo under hypoglycemic and postprandial (fed) conditions. Taken together, this study demonstrates that GLP-1 (via GLP-1R in α-cells) plays a bidirectional role, either stimulatory or inhibitory, in glucagon secretion depending on glucose levels.
Collapse
Affiliation(s)
- Yanqing Zhang
- Section of Endocrinology, Department of Medicine, Tulane University Health Science Center, New Orleans, LA
| | - Keshab R Parajuli
- Section of Endocrinology, Department of Medicine, Tulane University Health Science Center, New Orleans, LA
| | - Genevieve E Fava
- Section of Endocrinology, Department of Medicine, Tulane University Health Science Center, New Orleans, LA
| | - Rajesh Gupta
- Section of Endocrinology, Department of Medicine, Tulane University Health Science Center, New Orleans, LA
| | - Weiwei Xu
- Section of Endocrinology, Department of Medicine, Tulane University Health Science Center, New Orleans, LA
| | - Lauren U Nguyen
- Section of Endocrinology, Department of Medicine, Tulane University Health Science Center, New Orleans, LA
| | - Anadil F Zakaria
- Section of Endocrinology, Department of Medicine, Tulane University Health Science Center, New Orleans, LA
| | - Vivian A Fonseca
- Section of Endocrinology, Department of Medicine, Tulane University Health Science Center, New Orleans, LA
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC
| | - Franck Mauvais-Jarvis
- Section of Endocrinology, Department of Medicine, Tulane University Health Science Center, New Orleans, LA
- Southeast Louisiana Veterans Healthcare Medical Center, New Orleans, LA
| | - Kyle W Sloop
- Diabetes and Complications, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN
| | - Hongju Wu
- Section of Endocrinology, Department of Medicine, Tulane University Health Science Center, New Orleans, LA
| |
Collapse
|
119
|
Yu P, Xu X, Zhang J, Xia X, Xu F, Weng J, Lai X, Shen Y. Liraglutide Attenuates Nonalcoholic Fatty Liver Disease through Adjusting Lipid Metabolism via SHP1/AMPK Signaling Pathway. Int J Endocrinol 2019; 2019:1567095. [PMID: 31236111 PMCID: PMC6545813 DOI: 10.1155/2019/1567095] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/28/2019] [Indexed: 12/12/2022] Open
Abstract
A glucagon-like peptide-1 (GLP-1) receptor agonist liraglutide (LR) had been experimentally and clinically shown to ameliorate nonalcoholic fatty liver disease (NAFLD). This study aimed to investigate the beneficial effect of LR on NAFLD in vivo and in vitro and its underlying molecular mechanism. The effects of LR were examined on the high-fat diet-induced in vivo model in mice and in vitro model of NAFLD in human HepG2 cells. Liver tissues and HepG2 cells were procured for measuring lipid metabolism, histological examination, and western blot analysis. LR administration significantly lowered the serum lipid profile and lipid disposition in vitro and in vivo because of the altered expression of enzymes on hepatic gluconeogenesis and lipid metabolism. Moreover, LR significantly decreased Src homology region 2 domain-containing phosphatase-1 (SHP1) and then increased the expression of phosphorylated-AMP-activated protein kinase (p-AMPK). However, the overexpression of SHP1 mediated by lentivirus vector reversed LR-induced improvement in lipid deposition. Moreover, SHP1 silencing could further increase the expression of p-AMPK to ameliorate lipid metabolism and relative lipogenic gene induced by LR. In addition, abrogation of AMPK by Compound C eliminated the protective effects of LR on lipid metabolism without changing the expression of SHP1. LR markedly prevented NAFLD through adjusting lipid metabolism via SHP1/AMPK signaling pathway.
Collapse
Affiliation(s)
- Peng Yu
- Department of Endocrinology and Metabolism, Jiangxi Institute of Endocrine and Metabolic Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xi Xu
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xuan Xia
- Department of Physiology and Pathophysiology, College of Medical Sciences, China Three Gorges University, Yichang, Hubei, China
| | - Fen Xu
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Jianping Weng
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Xiaoyang Lai
- Department of Endocrinology and Metabolism, Jiangxi Institute of Endocrine and Metabolic Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, Jiangxi Institute of Endocrine and Metabolic Diseases, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
120
|
Knudsen LB, Lau J. The Discovery and Development of Liraglutide and Semaglutide. Front Endocrinol (Lausanne) 2019; 10:155. [PMID: 31031702 PMCID: PMC6474072 DOI: 10.3389/fendo.2019.00155] [Citation(s) in RCA: 397] [Impact Index Per Article: 79.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 02/21/2019] [Indexed: 12/12/2022] Open
Abstract
The discovery of glucagon-like peptide-1 (GLP-1), an incretin hormone with important effects on glycemic control and body weight regulation, led to efforts to extend its half-life and make it therapeutically effective in people with type 2 diabetes (T2D). The development of short- and then long-acting GLP-1 receptor agonists (GLP-1RAs) followed. Our article charts the discovery and development of the long-acting GLP-1 analogs liraglutide and, subsequently, semaglutide. We examine the chemistry employed in designing liraglutide and semaglutide, the human and non-human studies used to investigate their cellular targets and pharmacological effects, and ongoing investigations into new applications and formulations of these drugs. Reversible binding to albumin was used for the systemic protraction of liraglutide and semaglutide, with optimal fatty acid and linker combinations identified to maximize albumin binding while maintaining GLP-1 receptor (GLP-1R) potency. GLP-1RAs mediate their effects via this receptor, which is expressed in the pancreas, gastrointestinal tract, heart, lungs, kidneys, and brain. GLP-1Rs in the pancreas and brain have been shown to account for the respective improvements in glycemic control and body weight that are evident with liraglutide and semaglutide. Both liraglutide and semaglutide also positively affect cardiovascular (CV) outcomes in individuals with T2D, although the precise mechanism is still being explored. Significant weight loss, through an effect to reduce energy intake, led to the approval of liraglutide (3.0 mg) for the treatment of obesity, an indication currently under investigation with semaglutide. Other ongoing investigations with semaglutide include the treatment of non-alcoholic fatty liver disease (NASH) and its use in an oral formulation for the treatment of T2D. In summary, rational design has led to the development of two long-acting GLP-1 analogs, liraglutide and semaglutide, that have made a vast contribution to the management of T2D in terms of improvements in glycemic control, body weight, blood pressure, lipids, beta-cell function, and CV outcomes. Furthermore, the development of an oral formulation for semaglutide may provide individuals with additional benefits in relation to treatment adherence. In addition to T2D, liraglutide is used in the treatment of obesity, while semaglutide is currently under investigation for use in obesity and NASH.
Collapse
Affiliation(s)
- Lotte Bjerre Knudsen
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
- *Correspondence: Lotte Bjerre Knudsen
| | - Jesper Lau
- Global Research Technology, Novo Nordisk A/S, Måløv, Denmark
| |
Collapse
|
121
|
Bangshaab M, Gutierrez A, Huynh KD, Knudsen JS, Arcanjo DDR, Petersen AG, Rungby J, Gejl M, Simonsen U. Different mechanisms involved in liraglutide and glucagon-like peptide-1 vasodilatation in rat mesenteric small arteries. Br J Pharmacol 2018; 176:386-399. [PMID: 30403290 DOI: 10.1111/bph.14534] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND AND PURPOSE Glucagon-like peptide-1 (GLP-1) is an incretin hormone that regulates insulin biosynthesis and secretion in a glucose-dependent manner and has been reported to induce vasodilatation. Here, we examined the possible vasorelaxant effect of GLP-1 and its underlying mechanisms. EXPERIMENTAL APPROACH Rat mesenteric arteries (diameter ≈ 200-400 μm) and human s.c. arteries were mounted in microvascular myographs for isometric tension recordings. The effect of GLP-1 on vascular responses was examined under normoglycaemic conditions and at high glucose concentrations. KEY RESULTS In rat mesenteric arteries and human s.c. arteries without branches, physiological concentrations (1-100 nM) of GLP-1(7-36) and liraglutide failed to cause relaxation or affect contractions evoked by electrical field stimulation. In contrast to GLP-1(7-36), liraglutide induced relaxations antagonized by the GLP-1 receptor antagonist, exendin-(9-39), in branched mesenteric arteries. In contrast to liraglutide, GLP-1 leftward shifted the concentration relaxation curves for bradykinin in s.c. arteries from patients with peripheral arterial disease, an effect resistant to exendin-(9-39). Under normoglycaemic conditions, neither GLP-1 nor liraglutide affected ACh relaxation in rat mesenteric arteries. In arteries exposed to 40 mM glucose, GLP-1, in contrast to liraglutide, potentiated ACh-induced relaxation by a mechanism that was not antagonized by exendin-(9-39). GLP-1 decreased superoxide levels measured with dihydroethidium in rat mesenteric arteries exposed to 40 mM glucose. CONCLUSIONS AND IMPLICATIONS GLP-1 receptors are involved in the liraglutide-induced relaxation of branched arteries, under normoglycaemic conditions, while GLP-1 inhibition of vascular superoxide levels contributes to GLP-1 receptor-independent potentiation of endothelium-dependent vasodilatation in hyperglycaemia.
Collapse
Affiliation(s)
- Maj Bangshaab
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Alejandro Gutierrez
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Khiem Dinh Huynh
- Department of Vascular Surgery, Aalborg University Hospital, Aalborg, Denmark
| | - Jakob Schöllhammer Knudsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Daniel Dias Rufino Arcanjo
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| | - Asbjørn G Petersen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Jørgen Rungby
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Department of Endocrinology IC, Bispebjerg University Hospital, Copenhagen, Denmark
| | - Michael Gejl
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
122
|
Rowlands J, Heng J, Newsholme P, Carlessi R. Pleiotropic Effects of GLP-1 and Analogs on Cell Signaling, Metabolism, and Function. Front Endocrinol (Lausanne) 2018; 9:672. [PMID: 30532733 PMCID: PMC6266510 DOI: 10.3389/fendo.2018.00672] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/29/2018] [Indexed: 12/22/2022] Open
Abstract
The incretin hormone Glucagon-Like Peptide-1 (GLP-1) is best known for its "incretin effect" in restoring glucose homeostasis in diabetics, however, it is now apparent that it has a broader range of physiological effects in the body. Both in vitro and in vivo studies have demonstrated that GLP-1 mimetics alleviate endoplasmic reticulum stress, regulate autophagy, promote metabolic reprogramming, stimulate anti-inflammatory signaling, alter gene expression, and influence neuroprotective pathways. A substantial body of evidence has accumulated with respect to how GLP-1 and its analogs act to restore and maintain normal cellular functions. These findings have prompted several clinical trials which have reported GLP-1 analogs improve cardiac function, restore lung function and reduce mortality in patients with obstructive lung disease, influence blood pressure and lipid storage, and even prevent synaptic loss and neurodegeneration. Mechanistically, GLP-1 elicits its effects via acute elevation in cAMP levels, and subsequent protein kinase(s) activation, pathways well-defined in pancreatic β-cells which stimulate insulin secretion in conjunction with elevated Ca2+ and ATP. More recently, new studies have shed light on additional downstream pathways stimulated by chronic GLP-1 exposure, findings which have direct relevance to our understanding of the potential therapeutic effects of longer lasting analogs recently developed for clinical use. In this review, we provide a comprehensive description of the diverse roles for GLP-1 across multiple tissues, describe downstream pathways stimulated by acute and chronic exposure, and discuss novel pleiotropic applications of GLP-1 mimetics in the treatment of human disease.
Collapse
Affiliation(s)
| | | | - Philip Newsholme
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, WA, Australia
| | - Rodrigo Carlessi
- School of Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Perth, WA, Australia
| |
Collapse
|
123
|
The GLP-1 Analogs Liraglutide and Semaglutide Reduce Atherosclerosis in ApoE -/- and LDLr -/- Mice by a Mechanism That Includes Inflammatory Pathways. JACC Basic Transl Sci 2018; 3:844-857. [PMID: 30623143 PMCID: PMC6314963 DOI: 10.1016/j.jacbts.2018.09.004] [Citation(s) in RCA: 236] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 09/10/2018] [Accepted: 09/14/2018] [Indexed: 02/06/2023]
Abstract
The GLP-1RAs liraglutide and semaglutide reduce cardiovascular risk in type 2 diabetes patients. In ApoE−/− mice and LDLr−/− mice, liraglutide and semaglutide treatment significantly attenuated plaque lesion development, in part independently of body weight and cholesterol lowering. Semaglutide decreased levels of plasma markers of systemic inflammation in an acute inflammation model (lipopolysaccharide), and transcriptomic analysis of aortic atherosclerotic tissue revealed that multiple inflammatory pathways were down-regulated by semaglutide.
The glucagon-like peptide-1 receptor agonists (GLP-1RAs) liraglutide and semaglutide reduce cardiovascular risk in type 2 diabetes patients. The mode of action is suggested to occur through modified atherosclerotic progression. In this study, both of the compounds significantly attenuated plaque lesion development in apolipoprotein E-deficient (ApoE−/−) mice and low-density lipoprotein receptor-deficient (LDLr−/−) mice. This attenuation was partly independent of weight and cholesterol lowering. In aortic tissue, exposure to a Western diet alters expression of genes in pathways relevant to the pathogenesis of atherosclerosis, including leukocyte recruitment, leukocyte rolling, adhesion/extravasation, cholesterol metabolism, lipid-mediated signaling, extracellular matrix protein turnover, and plaque hemorrhage. Treatment with semaglutide significantly reversed these changes. These data suggest GLP-1RAs affect atherosclerosis through an anti-inflammatory mechanism.
Collapse
Key Words
- CD163, cluster of differentiation 163 molecule
- GLP, glucagon-like peptide
- GLP-1
- IFN, interferon
- IL, interleukin
- LDL, low-density lipoprotein
- LPS, lipopolysaccharide
- MMP, matrix metalloproteinase
- NASH, nonalcoholic steatohepatitis
- OPN, osteopontin
- RNA, ribonucleic acid
- TIMP, tissue inhibitor of metalloproteinases
- TNF, tumor necrosis factor
- WD, Western diet
- atherosclerosis
- diabetes
- inflammation
- obesity
Collapse
|
124
|
Antidiabetic Drugs in NAFLD: The Accomplishment of Two Goals at Once? Pharmaceuticals (Basel) 2018; 11:ph11040121. [PMID: 30413050 PMCID: PMC6316860 DOI: 10.3390/ph11040121] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 02/06/2023] Open
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) is the most common cause of chronic liver disease in Western countries, accounting for 20–30% of general population and reaching a prevalence of 55% in patients with type 2 diabetes mellitus (T2DM). Insulin resistance plays a key role in pathogenic mechanisms of NAFLD. Many drugs have been tested but no medications have yet been approved. Antidiabetic drugs could have a role in the progression reduction of the disease. The aim of this review is to summarize evidence on efficacy and safety of antidiabetic drugs in patients with NAFLD. Metformin, a biguanide, is the most frequently used drug in the treatment of T2DM. To date 15 randomized controlled trials (RCTs) and four meta-analysis on the use of metformin in NAFLD are available. No significant improvement in histological liver fibrosis was shown, but it can be useful in the treatment of co-factors of NAFLD, like body weight, transaminase or cholesterol levels, and HbA1c levels. A possible protective role in various types of cancer has been reported for Metformin. Thiazolidinediones modulate insulin sensitivity by the activation of PPAR-γ. The RCTs and the meta-analysis available about the role of these drugs in NAFLD show an improvement in ballooning, lobular inflammation, and perhaps fibrosis, but some side effects, in particular cardiovascular, were showed. GLP-1 analogues stimulate insulin secretion by pancreatic beta cell and inhibit glucagon release; Liraglutide is the most used drug in this class and significantly improves steatosis, hepatocyte ballooning and transaminase levels. Scanty data about the role of DPP-4 and SGLT inhibitors were published. No data about insulin effects on NAFLD are available but it was showed a possible association between insulin use and the development of solid neoplasms, in particular HCC. In conclusion, antidiabetic drugs seem to be promising drugs, because they are able to treat both NAFLD manifestations and diabetes, preventing worsening of hepatic damage, but data are still conflicting. All antidiabetic drugs can be safely used in patients with compensated cirrhosis, while insulin is the preferred drug in decompensated Child C cirrhosis.
Collapse
|
125
|
Differential Effects of a Glucagon-Like Peptide 1 Receptor Agonist in Non-Alcoholic Fatty Liver Disease and in Response to Hepatectomy. Sci Rep 2018; 8:16461. [PMID: 30405191 PMCID: PMC6220318 DOI: 10.1038/s41598-018-33949-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 10/08/2018] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is associated with post-operative liver failure (PLF) and impaired liver regeneration. We investigated the effects of a glucagon-like peptide-1 (GLP-1) receptor agonist on NAFLD, PLF and liver regeneration in mice fed chow diet or methionine/choline-deficient diet (MCD) or high fat diet (HFD). Fc-GLP-1 decreased transaminases, reduced intrahepatic triglycerides (TG) and improved MCD-induced liver dysfuction. Macrophage/Kupffer cell-related markers were also reduced although Fc-GLP-1 increased expression of genes related to natural killer (NK), cytotoxic T lymphocytes and hepatic stellate cell (HSC) activation. After partial hepatectomy (PH), survival rates increased in mice receiving Fc-GLP-1 on chow or MCD diet. However, the benefit of Fc-GLP-1 on NASH-like features was attenuated 2 weeks post-PH and liver mass restoration was not improved. At this time-period, markers of NK cells and cytotoxic T lymphocytes were further elevated in Fc-GLP-1 treated mice. Increased HSC related gene expression in livers was observed together with decreased retinyl ester content and increased retinal and retinoic acid, reflecting HSC activation. Similar effects were found in mice fed HFD receiving Fc-GLP-1. Our results shed light on the differential effects of a long-acting GLP-1R agonist in improving NAFLD and PLF, but not enhancing liver regeneration in mice.
Collapse
|
126
|
Gallego-Colon E, Wojakowski W, Francuz T. Incretin drugs as modulators of atherosclerosis. Atherosclerosis 2018; 278:29-38. [DOI: 10.1016/j.atherosclerosis.2018.09.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 07/06/2018] [Accepted: 09/13/2018] [Indexed: 02/06/2023]
|
127
|
Bifari F, Manfrini R, Dei Cas M, Berra C, Siano M, Zuin M, Paroni R, Folli F. Multiple target tissue effects of GLP-1 analogues on non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Pharmacol Res 2018; 137:219-229. [PMID: 30359962 DOI: 10.1016/j.phrs.2018.09.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2018] [Revised: 09/11/2018] [Accepted: 09/24/2018] [Indexed: 12/12/2022]
Abstract
Accumulating experimental and clinical evidences over the last decade indicate that GLP-1 analogues have a series of central nervous system and peripheral target tissues actions which are able to significantly influence the liver metabolism. GLP-1 analogues pleiotropic effects proved to be efficacious in T2DM subjects not only reducing liver steatosis and ameliorating NAFLD and NASH, but also in lowering plasma glucose and liver inflammation, improving cardiac function and protecting from kidney dysfunction. While the experimental and clinical data are robust, the precise mechanisms of action potentially involved in these protective multi-target effects need further investigation. Here we present a systematic review of the most recent literature data on the multi-target effects of GLP-1 analogues on the liver, on adipose and muscular tissue and on the nervous system, all capable of influencing significant aspects of the fatty liver disease physiopathology. From this analysis, we can conclude that the multi-target beneficial action of the GLP-1 analogues could explain the positive effects observed in animal and human models on progression of NAFLD to NASH.
Collapse
Affiliation(s)
- Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Roberto Manfrini
- Department of Internal Medicine ASST Santi Paolo e Carlo, Milan, Italy
| | - Michele Dei Cas
- Laboratory of Clinical Biochemistry and Mass Spectrometry, Department of Health Science, University of Milan, Milan, Italy
| | - Cesare Berra
- Metabolic Disease and Diabetes, Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Matteo Siano
- Department of Internal Medicine ASST Santi Paolo e Carlo, Milan, Italy
| | - Massimo Zuin
- Unit of Medicine, Gastroenterology and Hepatology, Milan, Italy
| | - Rita Paroni
- Laboratory of Clinical Biochemistry and Mass Spectrometry, Department of Health Science, University of Milan, Milan, Italy
| | - Franco Folli
- Unit of Endocrinology and Metabolism ASST Santi Paolo e Carlo, Department of Health Science, University of Milan, Milan, Italy.
| |
Collapse
|
128
|
Ämmälä C, Drury WJ, Knerr L, Ahlstedt I, Stillemark-Billton P, Wennberg-Huldt C, Andersson EM, Valeur E, Jansson-Löfmark R, Janzén D, Sundström L, Meuller J, Claesson J, Andersson P, Johansson C, Lee RG, Prakash TP, Seth PP, Monia BP, Andersson S. Targeted delivery of antisense oligonucleotides to pancreatic β-cells. SCIENCE ADVANCES 2018; 4:eaat3386. [PMID: 30345352 PMCID: PMC6192685 DOI: 10.1126/sciadv.aat3386] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 09/12/2018] [Indexed: 05/03/2023]
Abstract
Antisense oligonucleotide (ASO) silencing of the expression of disease-associated genes is an attractive novel therapeutic approach, but treatments are limited by the ability to deliver ASOs to cells and tissues. Following systemic administration, ASOs preferentially accumulate in liver and kidney. Among the cell types refractory to ASO uptake is the pancreatic insulin-secreting β-cell. Here, we show that conjugation of ASOs to a ligand of the glucagon-like peptide-1 receptor (GLP1R) can productively deliver ASO cargo to pancreatic β-cells both in vitro and in vivo. Ligand-conjugated ASOs silenced target genes in pancreatic islets at doses that did not affect target gene expression in liver or other tissues, indicating enhanced tissue and cell type specificity. This finding has potential to broaden the use of ASO technology, opening up novel therapeutic opportunities, and presents an innovative approach for targeted delivery of ASOs to additional cell types.
Collapse
Affiliation(s)
- C. Ämmälä
- Cardiovascular Renal and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
- Corresponding author. (C.Ä.); (P.P.S.)
| | - W. J. Drury
- Cardiovascular Renal and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - L. Knerr
- Cardiovascular Renal and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - I. Ahlstedt
- Cardiovascular Renal and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - P. Stillemark-Billton
- Cardiovascular Renal and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - C. Wennberg-Huldt
- Cardiovascular Renal and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - E.-M. Andersson
- Cardiovascular Renal and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - E. Valeur
- Cardiovascular Renal and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - R. Jansson-Löfmark
- Cardiovascular Renal and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - D. Janzén
- Cardiovascular Renal and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - L. Sundström
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - J. Meuller
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - J. Claesson
- Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - P. Andersson
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - C. Johansson
- Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - R. G. Lee
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - T. P. Prakash
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - P. P. Seth
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, USA
- Corresponding author. (C.Ä.); (P.P.S.)
| | - B. P. Monia
- Ionis Pharmaceuticals, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - S. Andersson
- Cardiovascular Renal and Metabolic Diseases, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
129
|
Drucker DJ. The Ascending GLP-1 Road From Clinical Safety to Reduction of Cardiovascular Complications. Diabetes 2018; 67:1710-1719. [PMID: 30135132 DOI: 10.2337/dbi18-0008] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 06/01/2018] [Indexed: 11/13/2022]
Abstract
Glucagon-like peptide 1 (GLP-1) was originally identified as a gut-derived incretin hormone that lowered glycemia through potentiation of glucose-dependent insulin secretion. Subsequent studies expanded the actions of GLP-1 to include inhibition of glucagon secretion, gastric emptying, and appetite, collectively useful attributes for a glucose-lowering agent. The introduction of GLP-1 receptor (GLP-1R) agonists for the treatment of diabetes was associated with questions surrounding their safety, principally with regard to medullary thyroid cancer, pancreatitis, and pancreatic cancer, yet cardiovascular outcome trials subsequently revealed reductions in rates of stroke, myocardial infarction, and cardiovascular death with a paucity of major safety signals. We discuss the controversies, unanswered questions, and established use of GLP-1R agonists from a mechanistic and clinical perspective. We highlight methods for detection and cellular sites of GLP-1R expression, key uncertainties, recent insights, and experimental caveats surrounding the use of GLP-1R agonists for the treatment of diabetes and the reduction of diabetes-related complications.
Collapse
Affiliation(s)
- Daniel J Drucker
- Department of Medicine, Mount Sinai Hospital, Lunenfeld-Tanenbaum Research Institute, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
130
|
Hebsgaard JB, Pyke C, Yildirim E, Knudsen LB, Heegaard S, Kvist PH. Glucagon-like peptide-1 receptor expression in the human eye. Diabetes Obes Metab 2018; 20:2304-2308. [PMID: 29707863 PMCID: PMC6099507 DOI: 10.1111/dom.13339] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/13/2018] [Accepted: 04/23/2018] [Indexed: 12/20/2022]
Abstract
Semaglutide is a human glucagon-like peptide-1 (GLP-1) analogue that is in development for the treatment of type 2 diabetes. In the pre-approval cardiovascular outcomes trial SUSTAIN 6, semaglutide was associated with a significant increase in the risk of diabetic retinopathy (DR) complications vs placebo. GLP-1 receptor (GLP-1R) expression has previously been demonstrated in the retina in animals and humans; however, antibodies used to detect expression have been documented to be non-specific and fail to detect the GLP-1R using immunohistochemistry (IHC), a problem common for many G-protein coupled receptors. Using a validated GLP-1R antibody for IHC and in situ hybridization for GLP-1R mRNA in normal human eyes, GLP-1Rs were detected in a small fraction of neurons in the ganglion cell layer. In advanced stages of DR, GLP-1R expression was not detected at the protein or mRNA level. Specifically, no GLP-1R expression was found in the eyes of people with long-standing proliferative DR (PDR). In conclusion, GLP-1R expression is low in normal human eyes and was not detected in eyes exhibiting advanced stages of PDR.
Collapse
Affiliation(s)
| | | | | | | | - Steffen Heegaard
- Department of Pathology, Rigshospitalet, Eye Pathology SectionUniversity of CopenhagenCopenhagenDenmark
- Department of Ophthalmology, RigshospitaletUniversity of CopenhagenCopenhagenDenmark
| | | |
Collapse
|
131
|
Ipsen DH, Rolin B, Rakipovski G, Skovsted GF, Madsen A, Kolstrup S, Schou-Pedersen AM, Skat-Rørdam J, Lykkesfeldt J, Tveden-Nyborg P. Liraglutide Decreases Hepatic Inflammation and Injury in Advanced Lean Non-Alcoholic Steatohepatitis. Basic Clin Pharmacol Toxicol 2018; 123:704-713. [PMID: 29953740 DOI: 10.1111/bcpt.13082] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/26/2018] [Indexed: 02/06/2023]
Abstract
Although commonly associated with obesity, non-alcoholic fatty liver disease (NAFLD) is also present in the lean population representing a unique disease phenotype. Affecting 25% of the world's population, NAFLD is associated with increased mortality especially when progressed to non-alcoholic steatohepatitis (NASH). However, no approved pharmacological treatments exist. Current research focuses mainly on NASH associated with obesity, leaving the effectiveness of promising treatments in lean NASH virtually unknown. This study therefore aimed to evaluate the effect of liraglutide (glucagon-like peptide 1 analogue) and dietary intervention, alone and in combination, in guinea pigs with non-obese NASH. After 20 weeks of high-fat feeding (20% fat, 15% sucrose, 0.35% cholesterol), 40 female guinea pigs were block-randomized based on weight into four groups receiving one of four treatments for 4 weeks: continued high-fat diet (HF, control), high-fat diet and liraglutide treatment (HFL), chow diet (4% fat, 0% sucrose, 0% cholesterol; HFC) or chow diet and liraglutide treatment (HFCL). High-fat feeding induced NASH with severe fibrosis. Liraglutide decreased inflammation (p < 0.05) and hepatocyte ballooning (p < 0.05), while increasing hepatic α-tocopherol (p = 0.0154). Dietary intervention did not improve liver histopathology significantly, but decreased liver weight (p = 0.004), plasma total cholesterol (p = 0.0175), LDL-cholesterol (p = 0.0063), VLDL-cholesterol (p = 0.0034), hepatic cholesterol (p < 0.0001) and increased hepatic vitamin C (p = 0.0099). Combined liraglutide and dietary intervention induced a rapid weight loss, necessitating periodical liraglutide dose adjustment/discontinuation, limiting the strength of the findings from this group. Collectively, this pre-clinical study supports the beneficial effect of liraglutide on NASH and extends this notion to lean NASH.
Collapse
Affiliation(s)
- David H Ipsen
- Department of Veterinary & Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Bidda Rolin
- Diabetes and Cardiovascular Pharmacology, Global Research, Novo Nordisk A/S, Måløv, Denmark
| | - Günaj Rakipovski
- Diabetes and Cardiovascular Pharmacology, Global Research, Novo Nordisk A/S, Måløv, Denmark
| | - Gry F Skovsted
- Department of Veterinary & Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Anette Madsen
- Department of Veterinary & Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Stefanie Kolstrup
- Department of Veterinary & Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Anne Marie Schou-Pedersen
- Department of Veterinary & Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Josephine Skat-Rørdam
- Department of Veterinary & Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Jens Lykkesfeldt
- Department of Veterinary & Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Pernille Tveden-Nyborg
- Department of Veterinary & Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| |
Collapse
|
132
|
After the LEADER trial and SUSTAIN-6, how do we explain the cardiovascular benefits of some GLP-1 receptor agonists? DIABETES & METABOLISM 2018; 43 Suppl 1:2S3-2S12. [PMID: 28431669 DOI: 10.1016/s1262-3636(17)30067-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recent cardiovascular outcome trials - the LEADER with liragutide and SUSTAIN-6 with semaglutide - have shown significant reductions of major cardiovascular (CV) events with these glucagon-like peptide (GLP)-1 receptor agonists. Progressive separation of the treatment and placebo curves, starting clearly between 12 and 18 months of the trial period, and significant reductions in the risk of myocardial infarction and stroke, indicate that the beneficial CV effects observed with GLP-1 receptor agonists could be due to an antiatherogenic effect. So far, the reasons for such an effect of GLP-1 receptor agonists have not been entirely clear, although several hypotheses may be proposed. As the reductions in glycated haemoglobin and systolic blood pressure (SBP) in these trials were modest, and both trials lasted only a short period of time, reductions in hyperglycaemia and SBP are unlikely to be involved in the beneficial CV effects of GLP-1 receptor agonists. On the other hand, their effect on lipids and, in particular, the dramatic decrease in postprandial hypertriglyceridaemia may explain their beneficial CV actions. Reduction of body weight, including a significant decrease in visceral fat in patients using GLP-1 receptor agonists, may also have beneficial CV effects by reducing chronic proatherogenic inflammation. In addition, there are in-vitro data showing a direct anti-inflammatory effect with these agents that could also be involved in their beneficial CV effects. Moreover, studies in humans have shown significant beneficial effects on ischaemic myocardium after a very short treatment period, suggesting a direct effect of GLP-1 receptor agonists on myocardium, although the precise mechanism remains unclear. Finally, as a reduction in insulin resistance has been associated with a decrease in CV risk, it cannot be ruled out that the lowering of insulin resistance induced by GLP-1 receptor agonists might also be involved in their beneficial CV actions.
Collapse
|
133
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) is very common in patients with type 2 diabetes (T2D), with approximately two-thirds having a diagnosis of the disease. Currently, the only validated treatment for NAFLD is weight loss. A number of studies of animal models and human trials have evaluated the effects of glucagon-like peptide-1 receptor agonists (GLP-1RAs) on liver fat content and suggest that the treatment could represent a new alternative for NAFLD management. In this review, our focus is on the main studies regarding the effects of GLP-1RAs on NAFLD. Also, the mechanisms that might explain their beneficial effects on liver diseases are analyzed.
Collapse
Affiliation(s)
- J-M Petit
- Université de Bourgogne, Centre de Recherche INSERM LNC-UMR1231; Service de Diabétologie et Endocrinologie, CHU François Mitterand, BP 77908, Dijon cedex 21079, France.
| | - B Vergès
- Université de Bourgogne, Centre de Recherche INSERM LNC-UMR1231; Service de Diabétologie et Endocrinologie, CHU François Mitterand, BP 77908, Dijon cedex 21079, France
| |
Collapse
|
134
|
Yang SH, Xu RX, Cui CJ, Wang Y, Du Y, Chen ZG, Yao YH, Ma CY, Zhu CG, Guo YL, Wu NQ, Sun J, Chen BX, Li JJ. Liraglutide downregulates hepatic LDL receptor and PCSK9 expression in HepG2 cells and db/db mice through a HNF-1a dependent mechanism. Cardiovasc Diabetol 2018; 17:48. [PMID: 29618348 PMCID: PMC5885408 DOI: 10.1186/s12933-018-0689-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/17/2018] [Indexed: 02/06/2023] Open
Abstract
Background Proprotein convertase subtilisin/kexin type 9 (PCSK9), a major regulator of cholesterol homeostasis, is associated with glucose metabolism. Liraglutide, a glucagon-like peptide-1 receptor agonist, can increase insulin secretion in a glucose-dependent manner and lower blood glucose. We aimed to investigate the relationship between liraglutide and PCSK9. Methods At the cellular level, the expressions of PCSK9 and hepatocyte nuclear factor 1 alpha (HNF1α) protein in HepG2 cells stimulated by liraglutide was examined using Western blot. Seven-week old db/db mice and wild type (WT) mice were administered either liraglutide (200 μg/kg) or equivoluminal saline subcutaneously, twice daily for 7 weeks. Fasting glucose level, food intake and body weight were measured every week. After the 7-week treatment, the blood was collected for lipid and PCSK9 levels detection and the liver was removed from the mice for oil red O staining, immunohistochemical analysis, immunofluorescence test and Western bolt. Results Firstly, liraglutide suppressed both PCSK9 and HNF1α expression in HepG2 cells in a time and concentration dependent manner. Secondly, liraglutide induced weight loss in WT and db/db mice, decreased serum PCSK9, glucose and lipid levels and improved hepatic accumulation in db/db but not WT mice. Thirdly, liraglutide reduced both hepatic PCSK9 and low-density lipoprotein receptor (LDLR) expression with a decrease in HNF1α in db/db mice but not in WT mice. Conclusions Liraglutide suppressed PCSK9 expression through HNF1α-dependent mechanism in HepG2 cells and db/db mice, and decreased LDLR possibly via PCSK9-independent pathways in db/db mice. Electronic supplementary material The online version of this article (10.1186/s12933-018-0689-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sheng-Hua Yang
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China.,Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Rui-Xia Xu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Chuan-Jue Cui
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Yin Wang
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Ying Du
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Zhi-Guo Chen
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Yu-Hong Yao
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Chun-Yan Ma
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Cheng-Gang Zhu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Yuan-Lin Guo
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Na-Qiong Wu
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Jing Sun
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China
| | - Bu-Xing Chen
- Department of Cardiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China.
| | - Jian-Jun Li
- Division of Dyslipidemia, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, BeiLiShi Road 167, Beijing, 100037, China.
| |
Collapse
|
135
|
Abstract
Glucagon-like peptide-1 (GLP-1) released from gut enteroendocrine cells controls meal-related glycemic excursions through augmentation of insulin and inhibition of glucagon secretion. GLP-1 also inhibits gastric emptying and food intake, actions maximizing nutrient absorption while limiting weight gain. Here I review the circuits engaged by endogenous versus pharmacological GLP-1 action, highlighting key GLP-1 receptor (GLP-1R)-positive cell types and pathways transducing metabolic and non-glycemic GLP-1 signals. The role(s) of GLP-1 in the benefits and side effects associated with bariatric surgery are discussed and actions of GLP-1 controlling islet function, appetite, inflammation, and cardiovascular pathophysiology are highlighted. Refinement of the risk-versus-benefit profile of GLP-1-based therapies for the treatment of diabetes and obesity has stimulated development of orally bioavailable agonists, allosteric modulators, and unimolecular multi-agonists, all targeting the GLP-1R. This review highlights established and emerging concepts, unanswered questions, and future challenges for development and optimization of GLP-1R agonists in the treatment of metabolic disease.
Collapse
Affiliation(s)
- Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, 600 University Avenue, Mailbox 39, Toronto, ON M5G 1X5, Canada.
| |
Collapse
|
136
|
Mulvihill EE. Regulation of intestinal lipid and lipoprotein metabolism by the proglucagon-derived peptides glucagon like peptide 1 and glucagon like peptide 2. Curr Opin Lipidol 2018; 29:95-103. [PMID: 29432213 PMCID: PMC5882252 DOI: 10.1097/mol.0000000000000495] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW The intestine is highly efficient at absorbing and packaging dietary lipids onto the structural protein apoB48 for distribution throughout the body. Here, we summarize recent advances into understanding the physiological and pharmacological actions of the proglucagon-derived peptides: glucagon like peptide 1 (GLP-1) and glucagon like peptide 2 (GLP-2) on intestinal lipoprotein secretion. RECENT FINDINGS Several recent studies have elucidated mechanisms underlying the paradoxical effects of GLP-1 and GLP-2 on intestinal production of triglyceride-rich lipoproteins (TRLs). Both gut-derived peptides are secreted on an equimolar basis in response to the same nutrient stimulus. Despite neither receptor demonstrating clear localization to enterocytes, a single injection of a GLP-1R agonist rapidly decreases delivery of intestinally packaged fatty acids into the plasma, while conversely GLP-2 receptor (GLP-2R) activation acutely increases TRL concentrations in plasma. SUMMARY The regulation of TRL secretion is dependent on the coordination of many processes: fatty acid availability uptake, assembly onto the apoB48 polypeptide backbone, secretion and reuptake, which the hormonal, neural, inflammatory and metabolic milieu can all strongly influence. Understanding of how GLP-1 and GLP-2 receptor agonists control TRL production has clinical importance given that GLP1R agonists were recently demonstrated not only to provide glycemic control but also to prevent major adverse cardiovascular events in patients with T2DM and the success of GLP-2R agonists in treating short bowel disease.
Collapse
Affiliation(s)
- Erin E Mulvihill
- University of Ottawa Heart Institute, University of Ottawa, Department of Biochemistry, Microbiology and Immunology, Ottawa, Ontario, Canada
| |
Collapse
|
137
|
Decara J, Rivera P, Arrabal S, Vargas A, Serrano A, Pavón FJ, Dieguez C, Nogueiras R, Rodríguez de Fonseca F, Suárez J. Cooperative role of the glucagon-like peptide-1 receptor and β3-adrenergic-mediated signalling on fat mass reduction through the downregulation of PKA/AKT/AMPK signalling in the adipose tissue and muscle of rats. Acta Physiol (Oxf) 2018; 222:e13008. [PMID: 29193738 DOI: 10.1111/apha.13008] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/13/2017] [Accepted: 11/23/2017] [Indexed: 12/19/2022]
Abstract
AIM To explore the cooperation of GLP-1 receptor and β3-adrenergic receptor (β3-AR)-mediated signalling in the control of fat mass/feeding behaviour by studying the effects of a combined therapy composed of the GLP-1R agonist liraglutide and the β3-AR agonist CL316243. METHODS The study included the analysis of key mechanisms regulating lipid/cholesterol metabolism, and thermogenesis in brown (BAT) and epididymal white (eWAT) adipose tissues, abdominal muscle and liver of male rats. RESULTS CL316243 (1 mg kg-1 ) and liraglutide (100 μg kg-1 ) co-administration over 6 days potentiated an overall negative energy balance (reduction in food intake, body weight gain, fat/non-fat mass ratio, liver fat content, and circulating levels of non-essential fatty acids, triglycerides, very low-density lipoprotein-cholesterol and leptin). These effects were accompanied by increased plasma levels of insulin and IL6. We also observed increased gene expression of uncoupling proteins regulating thermogenesis in BAT/eWAT (Ucp1) and muscle (Ucp2/3). Expression of transcription factor and enzymes involved either in de novo lipogenesis (Chrebp, Acaca, Fasn, Scd1, Insig1, Srebp1) or in fatty acid β-oxidation (Cpt1b) was enhanced in eWAT and/or muscle but decreased in BAT. Pparα and Pparγ, essentials in lipid flux/storage, were decreased in BAT/eWAT but increased in the muscle and liver. Cholesterol synthesis regulators (Insig2, Srebp2, Hmgcr) were particularly over-expressed in muscle. These GLP-1R/β3-AR-induced metabolic effects were associated with the downregulation of cAMP-dependent signalling pathways (PKA/AKT/AMPK). CONCLUSION Combined activation of GLP-1 and β3-ARs potentiate changes in peripheral pathways regulating lipid/cholesterol metabolism in a tissue-specific manner that favours a switch in energy availability/expenditure and may be useful for obesity treatment.
Collapse
Affiliation(s)
- J. Decara
- Instituto de Investigación Biomédica de Málaga (IBIMA); UGC Salud Mental; Universidad de Málaga, Hospital Universitario Regional de Málaga; Málaga Spain
| | - P. Rivera
- Department of Endocrinology; Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús; Madrid Spain
| | - S. Arrabal
- Instituto de Investigación Biomédica de Málaga (IBIMA); UGC Salud Mental; Universidad de Málaga, Hospital Universitario Regional de Málaga; Málaga Spain
| | - A. Vargas
- Instituto de Investigación Biomédica de Málaga (IBIMA); UGC Salud Mental; Universidad de Málaga, Hospital Universitario Regional de Málaga; Málaga Spain
| | - A. Serrano
- Instituto de Investigación Biomédica de Málaga (IBIMA); UGC Salud Mental; Universidad de Málaga, Hospital Universitario Regional de Málaga; Málaga Spain
| | - F. J. Pavón
- Instituto de Investigación Biomédica de Málaga (IBIMA); UGC Salud Mental; Universidad de Málaga, Hospital Universitario Regional de Málaga; Málaga Spain
| | - C. Dieguez
- Department of Physiology; School of Medicine-CIMUS; University of Santiago De Compostela-Instituto De Investigación Sanitaria; Santiago De Compostela Spain
- CIBER OBN; Instituto de Salud Carlos III; Madrid Spain
| | - R. Nogueiras
- Department of Physiology; School of Medicine-CIMUS; University of Santiago De Compostela-Instituto De Investigación Sanitaria; Santiago De Compostela Spain
- CIBER OBN; Instituto de Salud Carlos III; Madrid Spain
| | - F. Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga (IBIMA); UGC Salud Mental; Universidad de Málaga, Hospital Universitario Regional de Málaga; Málaga Spain
| | - J. Suárez
- Instituto de Investigación Biomédica de Málaga (IBIMA); UGC Salud Mental; Universidad de Málaga, Hospital Universitario Regional de Málaga; Málaga Spain
- Departamento de Biología Celular; Genética y Fisiología; Facultad de Ciencias; IBIMA; Universidad de Málaga; Málaga Spain
| |
Collapse
|
138
|
Baggio LL, Yusta B, Mulvihill EE, Cao X, Streutker CJ, Butany J, Cappola TP, Margulies KB, Drucker DJ. GLP-1 Receptor Expression Within the Human Heart. Endocrinology 2018; 159:1570-1584. [PMID: 29444223 PMCID: PMC5939638 DOI: 10.1210/en.2018-00004] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 02/06/2018] [Indexed: 12/25/2022]
Abstract
Glucagonlike peptide-1 receptor (GLP-1R) agonists, which are used to treat type 2 diabetes and obesity, reduce the rates of myocardial infarction and cardiovascular death. GLP-1R has been localized to the human sinoatrial node; however, its expression in ventricular tissue remains uncertain. Here we studied GLP-1R expression in the human heart using GLP-1R-directed antisera, quantitative polymerase chain reaction (PCR), reverse transcription PCR to detect full-length messenger RNA (mRNA) transcripts, and in situ hybridization (ISH). GLP1R mRNA transcripts, encompassing the entire open reading frame, were detected in all four cardiac chambers from 15 hearts at levels approximating those detected in human pancreas. In contrast, cardiac GLP2R expression was relatively lower, and cardiac GCGR expression was sporadic and not detected in the left ventricle. GLP1R mRNA transcripts were not detected in RNA from human cardiac fibroblasts, coronary artery endothelial, or vascular smooth muscle cells. Human Brunner glands and pancreatic islets exhibited GLP-1R immunopositivity and abundant expression of GLP1R mRNA transcripts by ISH. GLP1R transcripts were also detected by ISH in human cardiac sinoatrial node tissue. However, definitive cellular localization of GLP1R mRNA transcripts or immunoreactive GLP-1R protein within human cardiomyocytes or cardiac blood vessels remained elusive. Moreover, validated GLP-1R antisera lacked sufficient sensitivity to detect expression of the endogenous islet or cardiac GLP-1R by Western blotting. Hence, although human cardiac ventricles express the GLP1R, the identity of one or more ventricular cell type(s) that express a translated GLP1R protein requires further clarification with highly sensitive methods of detection.
Collapse
Affiliation(s)
- Laurie L Baggio
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | - Bernardo Yusta
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | - Erin E Mulvihill
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | - Xiemin Cao
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
| | | | - Jagdish Butany
- University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Thomas P Cappola
- University of Pennsylvania Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kenneth B Margulies
- University of Pennsylvania Perelman School of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, Toronto, Ontario, Canada
- Correspondence: Daniel J. Drucker, MD, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, 25 Orde Street, TCP5-1004, Toronto, Ontario M5G 1X5, Canada. E-mail:
| |
Collapse
|
139
|
Brubaker PL. The Cardiac Glucagonlike Peptide-1 Receptor: Whither Art Thou? Endocrinology 2018; 159:1842-1843. [PMID: 29509920 DOI: 10.1210/en.2018-00186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 02/24/2018] [Indexed: 11/19/2022]
Affiliation(s)
- Patricia L Brubaker
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
140
|
Lee SJ, Sanchez-Watts G, Krieger JP, Pignalosa A, Norell PN, Cortella A, Pettersen KG, Vrdoljak D, Hayes MR, Kanoski SE, Langhans W, Watts AG. Loss of dorsomedial hypothalamic GLP-1 signaling reduces BAT thermogenesis and increases adiposity. Mol Metab 2018; 11:33-46. [PMID: 29650350 PMCID: PMC6001878 DOI: 10.1016/j.molmet.2018.03.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 03/09/2018] [Accepted: 03/14/2018] [Indexed: 12/15/2022] Open
Abstract
Objective Glucagon-like peptide-1 (GLP-1) neurons in the hindbrain densely innervate the dorsomedial hypothalamus (DMH), a nucleus strongly implicated in body weight regulation and the sympathetic control of brown adipose tissue (BAT) thermogenesis. Therefore, DMH GLP-1 receptors (GLP-1R) are well placed to regulate energy balance by controlling sympathetic outflow and BAT function. Methods We investigate this possibility in adult male rats by using direct administration of GLP-1 (0.5 ug) into the DMH, knocking down DMH GLP-1R mRNA with viral-mediated RNA interference, and by examining the neurochemical phenotype of GLP-1R expressing cells in the DMH using in situ hybridization. Results GLP-1 administered into the DMH increased BAT thermogenesis and hepatic triglyceride (TG) mobilization. On the other hand, Glp1r knockdown (KD) in the DMH increased body weight gain and adiposity, with a concomitant reduction in energy expenditure (EE), BAT temperature, and uncoupling protein 1 (UCP1) expression. Moreover, DMH Glp1r KD induced hepatic steatosis, increased plasma TG, and elevated liver specific de-novo lipogenesis, effects that collectively contributed to insulin resistance. Interestingly, DMH Glp1r KD increased neuropeptide Y (NPY) mRNA expression in the DMH. GLP-1R mRNA in the DMH, however, was found in GABAergic not NPY neurons, consistent with a GLP-1R-dependent inhibition of NPY neurons that is mediated by local GABAergic neurons. Finally, DMH Glp1r KD attenuated the anorexigenic effects of the GLP-1R agonist exendin-4, highlighting an important role of DMH GLP-1R signaling in GLP-1-based therapies. Conclusions Collectively, our data show that DMH GLP-1R signaling plays a key role for BAT thermogenesis and adiposity. DMH GLP-1R stimulation acutely increases BAT thermogenesis. DMH GLP-1R mRNA knockdown decreases EE and BAT thermogenesis. DMH GLP-1R mRNA knockdown impairs lipid and glucose metabolism. Reduced DMH GLP-1R signaling blunts the anorexigenic responses to Ex-4. DMH GLP-1R signaling indirectly regulates NPY gene expression.
Collapse
Affiliation(s)
- Shin J Lee
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland.
| | - Graciela Sanchez-Watts
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | | | - Angelica Pignalosa
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Puck N Norell
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Alyssa Cortella
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Klaus G Pettersen
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Dubravka Vrdoljak
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Matthew R Hayes
- Department of Psychiatry, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Scott E Kanoski
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Wolfgang Langhans
- Physiology and Behavior Laboratory, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Alan G Watts
- Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
141
|
Marlet IR, Ölmestig JNE, Vilsbøll T, Rungby J, Kruuse C. Neuroprotective Mechanisms of Glucagon-like Peptide-1-based Therapies in Ischaemic Stroke: A Systematic Review based on Pre-Clinical Studies. Basic Clin Pharmacol Toxicol 2018; 122:559-569. [PMID: 29411931 DOI: 10.1111/bcpt.12974] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 01/19/2018] [Indexed: 12/12/2022]
Abstract
Glucagon-like peptide-1 (GLP-1)-based therapies, GLP-1 receptor agonists (GLP-1RAs) and dipeptidyl peptidase-4 inhibitors (DPP-4Is) are widely used for the treatment of type 2 diabetes. Increasing evidence suggests that they may provide neuroprotection. The aim of this MiniReview was to systematically evaluate the proposed mechanism of action for GLP-1-based therapies in ischaemic brain damage in animals. We performed a literature search using MEDLINE, EMBASE and The Cochrane Library. GLP-1-based therapies administered before, during or after experimental stroke in diabetic and non-diabetic animals were evaluated. We reviewed 27 studies comprised of 20 involving GLP-1RAs and seven involving DPP-4Is. Both GLP-1RAs and DPP-4Is affected the acute inflammatory response secondary to ischaemia by reducing inflammation, endothelial leakage and excitotoxicity. Both treatments also reduced oxidative stress and apoptosis. GLP-1RAs significantly reduced infarct volume when administered acutely, but not later after stroke. The reported effects of DPP-4Is on infarct volume were inconsistent. GLP-1-RAs reliably improved functional outcome, but the effects on cerebral blood flow were inconclusive. These neuroprotective effects were often attributed to activation of the GLP-1 receptor, but non-GLP-1R-mediated effects have also been suggested. Both GLP-1RAs and DPP-4Is significantly affected inflammation, oxidative stress and apoptosis in animal stroke models; however, data from clinical trials only report therapeutic efficacy for GLP-1RAs. Thus, GLP-1RA administration is the most promising treatment to pursue for patients at risk of stroke or immediately after stroke. Future studies should address acute and prophylactic treatments in stroke patients with and without diabetes.
Collapse
Affiliation(s)
- Ida R Marlet
- Neurovascular Research Unit, Dept. of Neurology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Joakim N E Ölmestig
- Neurovascular Research Unit, Dept. of Neurology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Tina Vilsbøll
- Centre for Diabetes Research, Herlev and Gentofte Hospital, University of Copenhagen, Hellerup, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jørgen Rungby
- Department of Endocrinology IC, Bispebjerg University Hospital, University of Copenhagen, Copenhagen, NV, Denmark
| | - Christina Kruuse
- Neurovascular Research Unit, Dept. of Neurology, Herlev and Gentofte Hospital, University of Copenhagen, Herlev, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
142
|
Biggs EK, Liang L, Naylor J, Madalli S, Collier R, Coghlan MP, Baker DJ, Hornigold DC, Ravn P, Reimann F, Gribble FM. Development and characterisation of a novel glucagon like peptide-1 receptor antibody. Diabetologia 2018; 61:711-721. [PMID: 29119245 PMCID: PMC5890879 DOI: 10.1007/s00125-017-4491-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/05/2017] [Indexed: 02/06/2023]
Abstract
AIMS/HYPOTHESIS Glucagon like peptide-1 (GLP-1) enhances glucose-dependent insulin secretion by binding to GLP-1 receptors (GLP1Rs) on pancreatic beta cells. GLP-1 mimetics are used in the clinic for the treatment of type 2 diabetes, but despite their therapeutic success, several clinical effects of GLP-1 remain unexplained at a mechanistic level, particularly in extrapancreatic tissues. The aim of this study was to generate and characterise a monoclonal antagonistic antibody for the GLP1R for use in vivo. METHODS A naive phage display selection strategy was used to isolate single-chain variable fragments (ScFvs) that bound to GLP1R. The ScFv with the highest affinity, Glp1R0017, was converted into a human IgG1 and characterised further. In vitro antagonistic activity was assessed in a number of assays: a cAMP-based homogenous time-resolved fluorescence assay in GLP1R-overexpressing cell lines, a live cell cAMP imaging assay and an insulin secretion assay in INS-1 832/3 cells. Glp1R0017 was further tested in immunostaining of mouse pancreas, and the ability of Glp1R0017 to block GLP1R in vivo was assessed by both IPGTT and OGTT in C57/Bl6 mice. RESULTS Antibodies to GLP1R were selected from naive antibody phage display libraries. The monoclonal antibody Glp1R0017 antagonised mouse, human, rat, cynomolgus monkey and dog GLP1R. This antagonistic activity was specific to GLP1R; no antagonistic activity was found in cells overexpressing the glucose-dependent insulinotropic peptide receptor (GIPR), glucagon like peptide-2 receptor or glucagon receptor. GLP-1-stimulated cAMP and insulin secretion was attenuated in INS-1 832/3 cells by Glp1R0017 incubation. Immunostaining of mouse pancreas tissue with Glp1R0017 showed specific staining in the islets of Langerhans, which was absent in Glp1r knockout tissue. In vivo, Glp1R0017 reversed the glucose-lowering effect of liraglutide during IPGTTs, and reduced glucose tolerance by blocking endogenous GLP-1 action in OGTTs. CONCLUSIONS/INTERPRETATION Glp1R0017 is a monoclonal antagonistic antibody to the GLP1R that binds to GLP1R on pancreatic beta cells and blocks the actions of GLP-1 in vivo. This antibody holds the potential to be used in investigating the physiological importance of GLP1R signalling in extrapancreatic tissues where cellular targets and signalling pathways activated by GLP-1 are poorly understood.
Collapse
Affiliation(s)
- Emma K Biggs
- Department of Cardiovascular and Metabolic Disease, MedImmune Ltd, Granta Park, Cambridge, UK
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK
- University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK
| | - Lihuan Liang
- Department of Cardiovascular and Metabolic Disease, MedImmune Ltd, Granta Park, Cambridge, UK
| | - Jacqueline Naylor
- Department of Cardiovascular and Metabolic Disease, MedImmune Ltd, Granta Park, Cambridge, UK
| | - Shimona Madalli
- Department of Cardiovascular and Metabolic Disease, MedImmune Ltd, Granta Park, Cambridge, UK
| | - Rachel Collier
- In Vivo Sciences - UK, AstraZeneca, The Babraham Institute, Cambridge, UK
| | - Matthew P Coghlan
- Department of Cardiovascular and Metabolic Disease, MedImmune Ltd, Granta Park, Cambridge, UK
| | - David J Baker
- Department of Cardiovascular and Metabolic Disease, MedImmune Ltd, Granta Park, Cambridge, UK
| | - David C Hornigold
- Department of Cardiovascular and Metabolic Disease, MedImmune Ltd, Granta Park, Cambridge, UK
| | - Peter Ravn
- Department of Antibody Discovery and Protein Engineering, MedImmune Ltd, Granta Park, Cambridge, CB21 6GH, UK.
| | - Frank Reimann
- University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK.
| | - Fiona M Gribble
- University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Hills Road, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
143
|
Coppolino G, Leporini C, Rivoli L, Ursini F, di Paola ED, Cernaro V, Arturi F, Bolignano D, Russo E, De Sarro G, Andreucci M. Exploring the effects of DPP-4 inhibitors on the kidney from the bench to clinical trials. Pharmacol Res 2018; 129:274-294. [DOI: 10.1016/j.phrs.2017.12.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 10/15/2017] [Accepted: 12/01/2017] [Indexed: 02/06/2023]
|
144
|
Podewin T, Ast J, Broichhagen J, Fine NHF, Nasteska D, Leippe P, Gailer M, Buenaventura T, Kanda N, Jones BJ, M’Kadmi C, Baneres JL, Marie J, Tomas A, Trauner D, Hoffmann-Röder A, Hodson DJ. Conditional and Reversible Activation of Class A and B G Protein-Coupled Receptors Using Tethered Pharmacology. ACS CENTRAL SCIENCE 2018; 4:166-179. [PMID: 29532016 PMCID: PMC5832994 DOI: 10.1021/acscentsci.7b00237] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Indexed: 05/21/2023]
Abstract
Understanding the activation and internalization of G protein-coupled receptors (GPCRs) using conditional approaches is paramount to developing new therapeutic strategies. Here, we describe the design, synthesis, and testing of ExONatide, a benzylguanine-linked peptide agonist of the glucagon-like peptide-1 receptor (GLP-1R), a class B GPCR required for maintenance of glucose levels in humans. ExONatide covalently binds to SNAP-tagged GLP-1R-expressing cells, leading to prolonged cAMP generation, Ca2+ rises, and intracellular retention of the receptor. These effects were readily switched OFF following cleavage of the introduced disulfide bridge using the cell-permeable reducing agent beta-mercaptoethanol (BME). A similar approach could be extended to a class A GPCR using GhrelON, a benzylguanine-linked peptide agonist of the growth hormone secretagogue receptor 1a (GHS-R1a), which is involved in food intake and growth. Thus, ExONatide and GhrelON allow SNAP-tag-directed activation of class A and B GPCRs involved in gut hormone signaling in a reversible manner. This tactic, termed reductively cleavable agONist (RECON), may be useful for understanding GLP-1R and GHS-R1a function both in vitro and in vivo, with applicability across GPCRs.
Collapse
Affiliation(s)
- Tom Podewin
- Department
of Chemistry and Center for Integrated Protein Science, LMU Munich, 81377 Munich, Germany
| | - Julia Ast
- Institute
of Metabolism and Systems Research (IMSR), University of Birmingham, B15 2TT, Birmingham, United Kingdom
- Centre
for Endocrinology, Diabetes and Metabolism, Birmingham
Health Partners, Birmingham, B15 2TH, United Kingdom, and COMPARE University of Birmingham and University of Nottingham
Midlands
| | - Johannes Broichhagen
- Department
of Chemistry and Center for Integrated Protein Science, LMU Munich, 81377 Munich, Germany
| | - Nicholas H. F. Fine
- Institute
of Metabolism and Systems Research (IMSR), University of Birmingham, B15 2TT, Birmingham, United Kingdom
- Centre
for Endocrinology, Diabetes and Metabolism, Birmingham
Health Partners, Birmingham, B15 2TH, United Kingdom, and COMPARE University of Birmingham and University of Nottingham
Midlands
| | - Daniela Nasteska
- Institute
of Metabolism and Systems Research (IMSR), University of Birmingham, B15 2TT, Birmingham, United Kingdom
- Centre
for Endocrinology, Diabetes and Metabolism, Birmingham
Health Partners, Birmingham, B15 2TH, United Kingdom, and COMPARE University of Birmingham and University of Nottingham
Midlands
| | - Philipp Leippe
- Department
of Chemistry and Center for Integrated Protein Science, LMU Munich, 81377 Munich, Germany
| | - Manuel Gailer
- Department
of Chemistry and Center for Integrated Protein Science, LMU Munich, 81377 Munich, Germany
| | - Teresa Buenaventura
- Section
of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - Nisha Kanda
- Section
of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - Ben J. Jones
- Section
of Investigative Medicine, Division of Diabetes, Endocrinology and
Metabolism, Imperial College London, London, W12 0NN, United Kingdom
| | - Celine M’Kadmi
- Institut des Biomolécules
Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM,
Faculté de Pharmacie, 15 Avenue
Charles Flahault, BP 14491, 34093 Montpellier Cedex 05, France
| | - Jean-Louis Baneres
- Institut des Biomolécules
Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM,
Faculté de Pharmacie, 15 Avenue
Charles Flahault, BP 14491, 34093 Montpellier Cedex 05, France
| | - Jacky Marie
- Institut des Biomolécules
Max Mousseron, UMR 5247 CNRS-Université Montpellier-ENSCM,
Faculté de Pharmacie, 15 Avenue
Charles Flahault, BP 14491, 34093 Montpellier Cedex 05, France
| | - Alejandra Tomas
- Section
of Cell Biology and Functional Genomics, Department of Medicine, Imperial College London, London, W12 0NN, United Kingdom
| | - Dirk Trauner
- Department
of Chemistry and Center for Integrated Protein Science, LMU Munich, 81377 Munich, Germany
- (D.T.) E-mail:
| | - Anja Hoffmann-Röder
- Department
of Chemistry and Center for Integrated Protein Science, LMU Munich, 81377 Munich, Germany
- (A.H.-R.) E-mail:
| | - David J. Hodson
- Institute
of Metabolism and Systems Research (IMSR), University of Birmingham, B15 2TT, Birmingham, United Kingdom
- Centre
for Endocrinology, Diabetes and Metabolism, Birmingham
Health Partners, Birmingham, B15 2TH, United Kingdom, and COMPARE University of Birmingham and University of Nottingham
Midlands
- (D.J.H.)
E-mail:
| |
Collapse
|
145
|
Pancreatic alpha cells in diabetic rats express active GLP-1 receptor: Endosomal co-localization of GLP-1/GLP-1R complex functioning through intra-islet paracrine mechanism. Sci Rep 2018; 8:3725. [PMID: 29487355 PMCID: PMC5829082 DOI: 10.1038/s41598-018-21751-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 02/09/2018] [Indexed: 01/03/2023] Open
Abstract
Glucagon-like peptide-1 (GLP-1) stimulates insulin secretion from pancreatic beta cells and suppresses glucagon secretion from alpha cells. It remains controversial, however, whether GLP-1 receptor (GLP-1R) is expressed in mature alpha cells. In this study, unlike previous studies using non-diabetic animals, we demonstrated using diabetic model rats and confocal laser scanning microscopy that the GLP-1/GLP-1R complex was located in the endosome of diabetic islets. In addition, we showed that GLP-1 and GLP-1R co-localized with various endosomal markers and adenylate cyclase in the alpha cells of diabetic rats. Diabetic rats had endosomal signaling pathway but normal rats had classical signaling pathway for activated GLP-1R. Furthermore, we performed pancreatic perfusion to assess the functional activity of GLP-1R when stimulated by exendin-4 (EX4). In a pancreas perfusion study, EX4 significantly stimulated glucagon secretion in diabetic rats but not normal rats. However, such glucagon secretion was immediately suppressed, probably due to concomitantly secreted insulin. The GLP-1/GLP-1R complex appears to function through an intra-islet paracrine mechanism in diabetic conditions which could explain, at least in part, the mechanism of paradoxical hyperglucagonaemia in type 2 diabetes.
Collapse
|
146
|
Jensen CB, Pyke C, Rasch MG, Dahl AB, Knudsen LB, Secher A. Characterization of the Glucagonlike Peptide-1 Receptor in Male Mouse Brain Using a Novel Antibody and In Situ Hybridization. Endocrinology 2018; 159:665-675. [PMID: 29095968 DOI: 10.1210/en.2017-00812] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/23/2017] [Indexed: 01/30/2023]
Abstract
Glucagonlike peptide 1 (GLP-1) is a physiological regulator of appetite, and long-acting GLP-1 receptor (GLP-1R) agonists lower food intake and body weight in both human and animal studies. The effects are mediated through brain GLP-1Rs, and several brain nuclei expressing the GLP-1R may be involved. To date, the mapping of the complete location of GLP-1R protein in the brain has been challenged by lack of good antibodies and the discrepancy between mRNA and protein, especially relevant in neuronal axonal processes. Here, we present a specific monoclonal GLP-1R antibody for immunohistochemistry with murine tissue and show detailed distribution of GLP-1R expression, as well as mapping of GLP-1R mRNA by nonradioactive in situ hybridization. Semiautomated image analysis was performed to map the GLP-1R distribution to atlas plates from the Allen Institute for Brain Science. The GLP-1R was abundantly expressed in numerous regions, including the septal nucleus, hypothalamus, and brain stem. GLP-1R protein expression was also observed on neuronal projections in brain regions devoid of any mRNA that has not been observed in earlier reports. Taken together, these findings provide knowledge on GLP-1R expression in neuronal cell bodies and neuronal projections.
Collapse
Affiliation(s)
- Casper Bo Jensen
- Global Research, Novo Nordisk A/S, Måløv, Denmark
- Image Analysis & Computer Graphics, Department of Applied Mathematics and Computer Science, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Charles Pyke
- Global Research, Novo Nordisk A/S, Måløv, Denmark
| | | | - Anders Bjorholm Dahl
- Image Analysis & Computer Graphics, Department of Applied Mathematics and Computer Science, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Anna Secher
- Global Research, Novo Nordisk A/S, Måløv, Denmark
| |
Collapse
|
147
|
Patel V, Joharapurkar A, Kshirsagar S, Sutariya B, Patel M, Pandey D, Patel H, Ranvir R, Kadam S, Patel D, Bahekar R, Jain M. Coagonist of GLP-1 and glucagon decreases liver inflammation and atherosclerosis in dyslipidemic condition. Chem Biol Interact 2018; 282:13-21. [DOI: 10.1016/j.cbi.2018.01.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 12/13/2017] [Accepted: 01/05/2018] [Indexed: 12/25/2022]
|
148
|
Al Batran R, Almutairi M, Ussher JR. Glucagon-like peptide-1 receptor mediated control of cardiac energy metabolism. Peptides 2018; 100:94-100. [PMID: 29412838 DOI: 10.1016/j.peptides.2017.12.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 12/16/2022]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists are frequently used to improve glycemia in patients with type 2 diabetes (T2D). Recent data from cardiovascular outcomes trials for the GLP-1R agonists, liraglutide and semaglutide, have also demonstrated significant reductions in death rates from cardiovascular causes in patients with T2D. As cardiovascular death is the number one cause of death in patients with T2D, understanding the mechanisms by which GLP-1R agonists such as liraglutide and semaglutide improve cardiac function is essential. Yet despite strong evidence from preclinical and clinical studies supporting the cardioprotective actions of GLP-1R agonists, the precise mechanism(s) by which this drug-class for T2D may produce these beneficial actions remains enigmatic. Negligible GLP-1R expression in ventricular cardiac myocytes suggests that GLP-1R agonist-induced cardioprotection is likely partially attributed to indirect actions on peripheral tissues other than the heart. Because insulin increases glucose oxidation, whereas glucagon increases fatty acid oxidation in the heart, GLP-1R agonist-induced increases and decreases in insulin and glucagon secretion, respectively, may modify cardiac energy metabolism in T2D patients. This may represent a potential mechanism for GLP-1R agonist-induced cardioprotection in T2D, as increases in fatty acid oxidation and decreases in glucose oxidation are frequently observed in the hearts of animals and human subjects with T2D.
Collapse
Affiliation(s)
- Rami Al Batran
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB Canada
| | - Malak Almutairi
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB Canada; Alberta Diabetes Institute, University of Alberta, Edmonton, AB Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, AB Canada.
| |
Collapse
|
149
|
Drucker DJ, Habener JF, Holst JJ. Discovery, characterization, and clinical development of the glucagon-like peptides. J Clin Invest 2017; 127:4217-4227. [PMID: 29202475 DOI: 10.1172/jci97233] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The discovery, characterization, and clinical development of glucagon-like-peptide-1 (GLP-1) spans more than 30 years and includes contributions from multiple investigators, science recognized by the 2017 Harrington Award Prize for Innovation in Medicine. Herein, we provide perspectives on the historical events and key experimental findings establishing the biology of GLP-1 as an insulin-stimulating glucoregulatory hormone. Important attributes of GLP-1 action and enteroendocrine science are reviewed, with emphasis on mechanistic advances and clinical proof-of-concept studies. The discovery that GLP-2 promotes mucosal growth in the intestine is described, and key findings from both preclinical studies and the GLP-2 clinical development program for short bowel syndrome (SBS) are reviewed. Finally, we summarize recent progress in GLP biology, highlighting emerging concepts and scientific insights with translational relevance.
Collapse
Affiliation(s)
- Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Joel F Habener
- Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Harvard University, Boston, Massachusetts, USA
| | - Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
150
|
Bruen R, Curley S, Kajani S, Crean D, O’Reilly ME, Lucitt MB, Godson CG, McGillicuddy FC, Belton O. Liraglutide dictates macrophage phenotype in apolipoprotein E null mice during early atherosclerosis. Cardiovasc Diabetol 2017; 16:143. [PMID: 29110715 PMCID: PMC5674826 DOI: 10.1186/s12933-017-0626-3] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/28/2017] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Macrophages play a pivotal role in atherosclerotic plaque development. Recent evidence has suggested the glucagon-like peptide-1 receptor (GLP-1R) agonist, liraglutide, can attenuate pro-inflammatory responses in macrophages. We hypothesized that liraglutide could limit atherosclerosis progression in vivo via modulation of the inflammatory response. METHODS Human THP-1 macrophages and bone marrow-derived macrophages, from both wild-type C57BL/6 (WT) and apolipoprotein E null mice (ApoE-/-) were used to investigate the effect of liraglutide on the inflammatory response in vitro. In parallel, ApoE-/- mice were fed a high-fat (60% calories from fat) high-cholesterol (1%) diet for 8 weeks to induce atherosclerotic disease progression with/without daily 300 μg/kg liraglutide administration for the final 6 weeks. Macrophages were analysed for MΦ1 and MΦ2 macrophage markers by Western blotting, RT-qPCR, ELISA and flow cytometry. Atherosclerotic lesions in aortae from ApoE-/- mice were analysed by en face staining and monocyte and macrophage populations from bone marrow derived cells analysed by flow cytometry. RESULTS Liraglutide decreased atherosclerotic lesion formation in ApoE-/- mice coincident with a reduction in pro-inflammatory and increased anti-inflammatory monocyte/macrophage populations in vivo. Liraglutide decreased IL-1beta in MΦ0 THP-1 macrophages and bone marrow-derived macrophages from WT mice and induced a significant increase in the MΦ2 surface marker mannose receptor in both MΦ0 and MΦ2 macrophages. Significant reduction in total lesion development was found with once daily 300 μg/kg liraglutide treatment in ApoE-/- mice. Interestingly, liraglutide inhibited disease progression at the iliac bifurcation suggesting that it retards the initiation and development of disease. These results corresponded to attenuated MΦ1 markers (CCR7, IL-6 and TNF-alpha), augmented MΦ2 cell markers (Arg-1, IL-10 and CD163) and finally decreased MΦ1-like monocytes and macrophages from bone marrow-derived cells. CONCLUSIONS This data supports a therapeutic role for liraglutide as an atheroprotective agent via modulating macrophage cell fate towards MΦ2 pro-resolving macrophages.
Collapse
Affiliation(s)
- Robyn Bruen
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Sean Curley
- Diabetes Complications Research Centre, School of Medicine, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Sarina Kajani
- Diabetes Complications Research Centre, School of Medicine, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Daniel Crean
- Diabetes Complications Research Centre, School of Veterinary Medicine, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Marcella E. O’Reilly
- Diabetes Complications Research Centre, School of Medicine, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Margaret B. Lucitt
- School of Medicine, Department of Pharmacology and Therapeutics, Trinity College Dublin, The University of Dublin, College Green, Dublin 2, Ireland
| | - Catherine G. Godson
- Diabetes Complications Research Centre, School of Medicine, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Fiona C. McGillicuddy
- Diabetes Complications Research Centre, School of Medicine, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Orina Belton
- Diabetes Complications Research Centre, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| |
Collapse
|