1
|
Haller N, Lutz TA. Incretin therapy in feline diabetes mellitus - A review of the current state of research. Domest Anim Endocrinol 2024; 89:106869. [PMID: 38870560 DOI: 10.1016/j.domaniend.2024.106869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/21/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Incretin hormones potentiate the glucose-induced insulin secretion following enteral nutrient intake. The best characterised incretin hormones are glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) which are produced in and secreted from the gut in response to nutrient ingestion. The property of incretins to enhance endogenous insulin secretion only at elevated blood glucose levels makes them interesting therapeutics for type 2 diabetes mellitus with a better safety profile than exogenous insulin. While incretin therapeutics (especially GLP-1 agonists, and more recently also GLP-1 / GIP dual agonists and other drugs that influence the incretin metabolism (e.g., dipeptidyl peptidase-4 (DPP-4) inhibitors)) are already widely used treatment options for human type 2 diabetes, these drugs are not yet approved for the therapy of feline diabetes mellitus. This review provides an introduction to incretins and feline diabetes mellitus in general and summarises the current study situation on incretins as therapeutics for feline diabetes mellitus to assess their possible future potential in feline medicine. Studies to date on the use of GLP-1 receptor agonists (GLP-1RA) in healthy cats largely confirm their insulinotropic effect known from other species. In diabetic cats, GLP-1RAs appear to significantly reduce glycaemic variability (GV, an indicator for the quality of glycaemic control), which is important for the management of the disease and prevention of long-term complications. However, for widespread use in feline diabetes mellitus, further studies are required that include larger numbers of diabetic cats, and that consider and test a possible need for dose adjustments to overweight and diabetic cats. Also evaluation of the outcome of GLP-1RA monotherapy will be neceessary.
Collapse
Affiliation(s)
- Nina Haller
- Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 204, CH 8057 Zurich, Switzerland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, CH 8057 Zurich, Switzerland.
| |
Collapse
|
2
|
Erta G, Gersone G, Jurka A, Tretjakovs P. The Link between Salivary Amylase Activity, Overweight, and Glucose Homeostasis. Int J Mol Sci 2024; 25:9956. [PMID: 39337444 PMCID: PMC11432655 DOI: 10.3390/ijms25189956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Butyrate, a short-chain fatty acid (SCFA) produced by the fermentation of dietary fibers in the colon, plays a pivotal role in regulating metabolic health, particularly by enhancing insulin sensitivity. Given the rising incidence of metabolic disorders, understanding the factors that influence butyrate production is of significant interest. This study explores the link between salivary amylase activity and butyrate levels in overweight women of reproductive age. Participants were categorized into low (LSA) and high (HSA) salivary amylase activity groups and further divided into two subgroups: one followed a low-starch diet (LS), and the other underwent caloric restriction (CR). We assessed salivary amylase activity and measured serum butyrate concentrations to examine their associations. Our findings showed a significant, though weak, positive correlation (ρ = 0.0486, p < 0.05), suggesting a link between salivary amylase activity and butyrate levels. The statistical significance, despite the weak correlation, implies that this relationship is not random. Moreover, higher baseline butyrate levels were observed in women with elevated salivary amylase activity. Also, women with low salivary amylase activity on a low-starch diet experienced a more pronounced increase in butyrate levels compared to those on caloric restriction. These results suggest that salivary amylase activity and dietary intake interact to influence butyrate production, with potential implications for improving insulin sensitivity and metabolic health. The study underscores the potential of butyrate in enhancing insulin sensitivity and promoting overall metabolic well-being. Further research is necessary to clarify the mechanisms involved and to understand the long-term effects of butyrate on metabolic health across different populations.
Collapse
Affiliation(s)
- Gita Erta
- Department of Human Physiology and Biochemistry, Riga Stradins University, LV-1007 Riga, Latvia
| | - Gita Gersone
- Department of Human Physiology and Biochemistry, Riga Stradins University, LV-1007 Riga, Latvia
| | - Antra Jurka
- Department of Human Physiology and Biochemistry, Riga Stradins University, LV-1007 Riga, Latvia
| | - Pēteris Tretjakovs
- Department of Human Physiology and Biochemistry, Riga Stradins University, LV-1007 Riga, Latvia
| |
Collapse
|
3
|
De Fano M, Malara M, Vermigli C, Murdolo G. Adipose Tissue: A Novel Target of the Incretin Axis? A Paradigm Shift in Obesity-Linked Insulin Resistance. Int J Mol Sci 2024; 25:8650. [PMID: 39201336 PMCID: PMC11354636 DOI: 10.3390/ijms25168650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 08/01/2024] [Indexed: 09/02/2024] Open
Abstract
Adipose tissue (AT) represents a plastic organ that can undergo significant remodeling in response to metabolic demands. With its numerous checkpoints, the incretin system seems to play a significant role in controlling glucose homeostasis and energy balance. The importance of the incretin hormones, namely the glucagon-like peptide-1 (GLP-1) and the glucose-dependent insulinotropic peptide (GIP), in controlling the function of adipose cells has been brought to light by recent studies. Notably, a "paradigm shift" in reevaluating the role of the incretin system in AT as a potential target to treat obesity-linked metabolic disorders resulted from the demonstration that a disruption of the GIP and GLP-1 signaling axis in fat is associated with adiposity-induced insulin-resistance (IR) and/or type 2 diabetes mellitus (T2D). We will briefly discuss the (patho)physiological functions of GLP-1 and GIP signaling in AT in this review, emphasizing their potential impacts on lipid storage, adipogenesis, glucose metabolism and inflammation. We will also address the conundrum with the perturbation of the incretin axis in white or brown fat tissue and the emergence of metabolic disorders. In order to reduce or avoid adiposity-related metabolic complications, we will finally go over a potential scientific rationale for suggesting AT as a novel target for GLP-1 and GIP receptor agonists and co-agonists.
Collapse
Affiliation(s)
- Michelantonio De Fano
- Complex Structure of Endocrinology and Metabolism, Department of Medicine, Azienda Ospedaliera Santa Maria Misericordia, Ospedale di Perugia, 06081 Perugia, Italy; (M.M.); (C.V.); (G.M.)
| | | | | | | |
Collapse
|
4
|
Wang Q, Du J, Ma R. White adipocyte-derived exosomal miR-23b inhibits thermogenesis by targeting Elf4 to regulate GLP-1R transcription. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5847-5860. [PMID: 38334823 DOI: 10.1007/s00210-024-02984-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 01/28/2024] [Indexed: 02/10/2024]
Abstract
Promoting non-trembling thermogenesis of brown adipose tissue (BAT) and browning of white adipose tissue (WAT) helps prevent obesity. MiR-23b is highly expressed in adipose tissue-derived exosomes obtained from obese people, but the role of exosomal miR-23b in regulating thermogenesis and obesity progression remains to be further explored. Here, a mouse obesity model was established through high-fat diet (HFD), and inguinal WAT (iWAT)-derived exosomes and miR-23b antagomir were administered by intraperitoneal injection. The results showed that WAT-derived exosomal miR-23b upregulated body weight and adipocyte hypertrophy and enhanced insulin resistance. Moreover, exosomal miR-23b restrained mtDNA copy number and the expression of genes related to thermogenesis and mitochondrial biogenesis in BAT, and suppressed the expression of WAT browning-related genes under cold stimulation, indicating that exosomal miR-23b hindered non-trembling thermogenesis of BAT and WAT browning. Mechanism studies found that miR-23b targeted Elf4 to inhibit its expression. And Elf4 bound to the GLP-1R promoter region to promote GLP-1R transcription. In addition, silencing miR-23b effectively abolished the inhibitory effect of WAT-derived exosomes on thermogenic gene expression and mitochondrial respiration in adipocytes isolated from BAT and iWAT, which was reversed by GLP-1R knockdown. In conclusion, WAT-derived exosomal miR-23b suppressed thermogenesis by targeting Elf4 to regulate GLP-1R transcription, which contributed to the progression of obesity.
Collapse
Affiliation(s)
- Qian Wang
- Functional Experiment Center, Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, China.
| | - Junkai Du
- Department of Emergency, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ruili Ma
- Functional Experiment Center, Department of Basic Medicine, Xi'an Medical University, Xi'an, 710021, China
| |
Collapse
|
5
|
Xie C, Alkhouri N, Elfeki MA. Role of incretins and glucagon receptor agonists in metabolic dysfunction-associated steatotic liver disease: Opportunities and challenges. World J Hepatol 2024; 16:731-750. [PMID: 38818288 PMCID: PMC11135259 DOI: 10.4254/wjh.v16.i5.731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 02/18/2024] [Accepted: 04/03/2024] [Indexed: 05/22/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has become the most common chronic liver disease worldwide, paralleling the rising pandemic of obesity and type 2 diabetes. Due to the growing global health burden and complex pathogenesis of MASLD, a multifaceted and innovative therapeutic approach is needed. Incretin receptor agonists, which were initially developed for diabetes management, have emerged as promising candidates for MASLD treatment. This review describes the pathophysiological mechanisms and action sites of three major classes of incretin/glucagon receptor agonists: glucagon-like peptide-1 receptor agonists, glucose-dependent insulinotropic polypeptide receptor agonists, and glucagon receptor agonists. Incretins and glucagon directly or indirectly impact various organs, including the liver, brain, pancreas, gastrointestinal tract, and adipose tissue. Thus, these agents significantly improve glycemic control and weight management and mitigate MASLD pathogenesis. Importantly, this study provides a summary of clinical trials analyzing the effectiveness and safety of incretin receptor agonists in MASLD management and provides an in-depth analysis highlighting their beneficial effects on improving liver function, hepatic steatosis, and intrahepatic inflammation. There are emerging challenges associated with the use of these medications in the real world, particularly adverse events, drug-drug interactions, and barriers to access, which are discussed in detail. Additionally, this review highlights the evolving role of incretin receptor agonists in MASLD management and suggests future research directions.
Collapse
Affiliation(s)
- Chencheng Xie
- Department of Internal Medicine, University of South Dakota Sanford School of Medicine, Sioux Falls, SD 57105, United States
- Department of Hepatology, Avera Mckennan University Hospital and Transplant Institute, Sioux Falls, SD 57105, United States
| | - Naim Alkhouri
- Department of Hepatology, Arizona Liver Health, Chandler, AZ 85712, United States
| | - Mohamed A Elfeki
- Department of Internal Medicine, University of South Dakota Sanford School of Medicine, Sioux Falls, SD 57105, United States
- Department of Hepatology, Avera McKennan University Hospital and Transplant Institute, Sioux Falls, SD 57105, United States.
| |
Collapse
|
6
|
IIDA M, ASANO A. Effects of glucagon-like peptide-1 receptor agonists on spermatogenesis-related gene expression in mouse testis and testis-derived cell lines. J Vet Med Sci 2024; 86:555-562. [PMID: 38556323 PMCID: PMC11144540 DOI: 10.1292/jvms.24-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is an incretin released into the gastrointestinal tract after food ingestion, and stimulates insulin secretion from the beta cells of the pancreatic islets. Incretins have recently been reported to have extrapancreatic actions, and they are anticipated to have potential efficacy for conditions such as male infertility as well as diabetes. However, the effects of incretins on male reproductive function remain unclear. In this study, GLP-1 receptor expression and the effects of GLP-1 on spermatogenesis-associated genes were investigated using mouse testes and testis-derived cultured cell lines. Glp1r mRNA and GLP-1 protein were expressed in mouse testes at levels comparable to or greater than those in positive control adipose tissue, and the liver and intestine, and also in a Sertoli cell line (TM4) and a Leydig cell line (MA-10) as well as the GC-1 spg and GC-2 spd (ts) germ cell lines. TM4 cells treated with the GLP-1 receptor agonist exenatide showed transiently and significantly upregulated Kitl, Pdgfa, and Glp1r mRNA expression. Furthermore, at 1 hr post-exenatide administration to male mice, Kitl and Glp1r mRNA expression levels were significantly increased, and Pdgfa mRNA expression level also showed a tendency toward increase. TM4 cells were treated with various cell-activating agents, and bucladesine elicited significantly increased Glp1r mRNA expression. We suggest that GLP-1 provides acute stimulation of Sertoli cells in the mouse testis and has a stimulatory effect on the expression of spermatogenesis-related genes.
Collapse
Affiliation(s)
- Masashi IIDA
- Laboratory of Laboratory Animal Science, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
- Safety Assessment Department, Kumamoto Laboratories, Mediford Corporation, Tokyo, Japan
| | - Atsushi ASANO
- Laboratory of Laboratory Animal Science, Joint Faculty of Veterinary Medicine, Kagoshima University, Kagoshima, Japan
| |
Collapse
|
7
|
Stafeev I, Agareva M, Michurina S, Tomilova A, Shestakova E, Zubkova E, Sineokaya M, Ratner E, Menshikov M, Parfyonova Y, Shestakova M. Semaglutide 6-months therapy of type 2 diabetes mellitus restores adipose progenitors potential to develop metabolically active adipocytes. Eur J Pharmacol 2024; 970:176476. [PMID: 38493915 DOI: 10.1016/j.ejphar.2024.176476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/20/2024] [Accepted: 03/01/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Nowadays type 2 diabetes mellitus (T2DM) leads to population mortality growth. Today glucagon-like peptide type 1 receptor agonists (GLP-1 RA) are one of the most promising glucose-lowered drugs with anorexigenic and cardioprotective effects. The present study aims to determine the effects of GLP-1 RA semaglutide 6-month therapy on T2DM patient metabolic parameters and adipose progenitor cell health. METHODS T2DM patients (N = 8) underwent clinical characterization and subcutaneous fat biopsy at start point and after semaglutide 6-month therapy. Adipose-derived stem cells (ADSC) were isolated by enzymatic method. Cell proliferation analysis was performed by MTT and immunocytochemistry. White and beige adipogenesis was analyzed by BODIPY493/503 staining and confocal microscopy. Adipocyte's metabolic properties were estimated by 3H- and 14C-based metabolic assays. Thermogenesis analysis was performed by ERthermAC staining and confocal microscopy. Protein markers were assessed by Western blotting. RESULTS Semaglutide 6-month therapy demonstrated significant anorexigenic and glucose-lowering effects. However, insulin sensitivity (HOMA-IR and M-index) was unchanged after therapy. Semaglutide 6-month therapy increased ADSC proliferation and white and beige adipogenesis. Moreover, lipid droplets fragmentation was observed in beige adipocytes. Both white and beige adipocytes after semaglutide therapy demonstrated 2-3 fold growth of glucose uptake without changes in insulin sensitivity. Newly formed white adipocytes demonstrated glucose utilization for active ATP synthesis, whereas beige adipocytes for canonical thermogenesis. CONCLUSIONS Our study has revealed that semaglutide 6-month therapy has not only systemic anorexigenic effects, but can markedly improve adipose tissue health. We have demonstrated critical restoration of ADSC renewal functions, which potentially can be involved in semaglutide based weight loss.
Collapse
Affiliation(s)
- I Stafeev
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia.
| | - M Agareva
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia; Lomonosov Moscow State University, 119991, Moscow, Russia
| | - S Michurina
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia; Lomonosov Moscow State University, 119991, Moscow, Russia
| | - A Tomilova
- Endocrinology Research Centre, 117292, Moscow, Russia
| | - E Shestakova
- Endocrinology Research Centre, 117292, Moscow, Russia
| | - E Zubkova
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia
| | - M Sineokaya
- Endocrinology Research Centre, 117292, Moscow, Russia
| | - E Ratner
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia
| | - M Menshikov
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia
| | - Ye Parfyonova
- National Medical Research Centre of Cardiology Named After Academician E.I.Chazov, 121552, Moscow, Russia; Lomonosov Moscow State University, 119991, Moscow, Russia
| | - M Shestakova
- Lomonosov Moscow State University, 119991, Moscow, Russia; Endocrinology Research Centre, 117292, Moscow, Russia
| |
Collapse
|
8
|
Bu T, Sun Z, Pan Y, Deng X, Yuan G. Glucagon-Like Peptide-1: New Regulator in Lipid Metabolism. Diabetes Metab J 2024; 48:354-372. [PMID: 38650100 PMCID: PMC11140404 DOI: 10.4093/dmj.2023.0277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/01/2024] [Indexed: 04/25/2024] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is a 30-amino acid peptide hormone that is mainly expressed in the intestine and hypothalamus. In recent years, basic and clinical studies have shown that GLP-1 is closely related to lipid metabolism, and it can participate in lipid metabolism by inhibiting fat synthesis, promoting fat differentiation, enhancing cholesterol metabolism, and promoting adipose browning. GLP-1 plays a key role in the occurrence and development of metabolic diseases such as obesity, nonalcoholic fatty liver disease, and atherosclerosis by regulating lipid metabolism. It is expected to become a new target for the treatment of metabolic disorders. The effects of GLP-1 and dual agonists on lipid metabolism also provide a more complete treatment plan for metabolic diseases. This article reviews the recent research progress of GLP-1 in lipid metabolism.
Collapse
Affiliation(s)
- Tong Bu
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ziyan Sun
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yi Pan
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xia Deng
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Guoyue Yuan
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
9
|
Kang PS, Neeland IJ. Body Fat Distribution, Diabetes Mellitus, and Cardiovascular Disease: an Update. Curr Cardiol Rep 2023; 25:1555-1564. [PMID: 37792133 DOI: 10.1007/s11886-023-01969-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/17/2023] [Indexed: 10/05/2023]
Abstract
PURPOSE OF REVIEW Specific measures of body fat distribution may have particular value in the development and treatment of cardiometabolic conditions, such as cardiovascular disease (CVD) and diabetes mellitus (DM). Here, we review the pathophysiology, epidemiology, and recent advances in the identification and management of body fat distribution as it relates to DM and CVD risk. RECENT FINDINGS Accumulation of visceral and ectopic fat is a major contributor to CVD and DM risk above and beyond the body mass index (BMI), yet implementation of fat distribution assessment into clinical practice remains a challenge. Newer imaging-based methods offer improved sensitivity and specificity for measuring specific fat depots. Lifestyle, pharmacological, and surgical interventions allow a multidisciplinary approach to reduce visceral and ectopic fat. A focus on implementation of body fat distribution measurements into clinical practice should be a priority over the next 5 to 10 years, and clinical assessment of fat distribution can be considered to refine risk evaluation and to develop improved and effective preventive and therapeutic strategies for high-risk obesity.
Collapse
Affiliation(s)
- Puneet S Kang
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Ian J Neeland
- Harrington Heart and Vascular Institute, University Hospitals Cleveland Medical Center and Case Western Reserve University School of Medicine, 11100 Euclid Ave, Cleveland, OH, 44106, USA.
| |
Collapse
|
10
|
Antoniades C, Tousoulis D, Vavlukis M, Fleming I, Duncker DJ, Eringa E, Manfrini O, Antonopoulos AS, Oikonomou E, Padró T, Trifunovic-Zamaklar D, De Luca G, Guzik T, Cenko E, Djordjevic-Dikic A, Crea F. Perivascular adipose tissue as a source of therapeutic targets and clinical biomarkers. Eur Heart J 2023; 44:3827-3844. [PMID: 37599464 PMCID: PMC10568001 DOI: 10.1093/eurheartj/ehad484] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 05/03/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
Obesity is a modifiable cardiovascular risk factor, but adipose tissue (AT) depots in humans are anatomically, histologically, and functionally heterogeneous. For example, visceral AT is a pro-atherogenic secretory AT depot, while subcutaneous AT represents a more classical energy storage depot. Perivascular adipose tissue (PVAT) regulates vascular biology via paracrine cross-talk signals. In this position paper, the state-of-the-art knowledge of various AT depots is reviewed providing a consensus definition of PVAT around the coronary arteries, as the AT surrounding the artery up to a distance from its outer wall equal to the luminal diameter of the artery. Special focus is given to the interactions between PVAT and the vascular wall that render PVAT a potential therapeutic target in cardiovascular diseases. This Clinical Consensus Statement also discusses the role of PVAT as a clinically relevant source of diagnostic and prognostic biomarkers of vascular function, which may guide precision medicine in atherosclerosis, hypertension, heart failure, and other cardiovascular diseases. In this article, its role as a 'biosensor' of vascular inflammation is highlighted with description of recent imaging technologies that visualize PVAT in clinical practice, allowing non-invasive quantification of coronary inflammation and the related residual cardiovascular inflammatory risk, guiding deployment of therapeutic interventions. Finally, the current and future clinical applicability of artificial intelligence and machine learning technologies is reviewed that integrate PVAT information into prognostic models to provide clinically meaningful information in primary and secondary prevention.
Collapse
Affiliation(s)
- Charalambos Antoniades
- Acute Multidisciplinary Imaging and Interventional Centre, RDM Division of Cardiovascular Medicine, University of Oxford, Headley Way, Headington, Oxford OX39DU, UK
| | - Dimitris Tousoulis
- 1st Cardiology Department, National and Kapodistrian University of Athens, Greece
| | - Marija Vavlukis
- Medical Faculty, University Clinic for Cardiology, University Ss’ Cyril and Methodius in Skopje, Skopje, North Macedonia
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre of Molecular Medicine, Goethe University, Frankfurt, Germany
| | - Dirk J Duncker
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Etto Eringa
- Cardiovascular-Program ICCC, Research Institute—Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Olivia Manfrini
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Alexios S Antonopoulos
- Acute Multidisciplinary Imaging and Interventional Centre, RDM Division of Cardiovascular Medicine, University of Oxford, Headley Way, Headington, Oxford OX39DU, UK
- 1st Cardiology Department, National and Kapodistrian University of Athens, Greece
| | - Evangelos Oikonomou
- 1st Cardiology Department, National and Kapodistrian University of Athens, Greece
| | - Teresa Padró
- Cardiovascular Program-ICCC, Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), Barcelona, Spain
- CiberCV, Institute Carlos III, Madrid, Spain
| | | | - Giuseppe De Luca
- Division of Cardiology, AOU Policlinico G. Martino, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
- Cardiologia Ospedaliera, Nuovo Galeazzi-Sant’Ambrogio, Milan, Italy
| | - Tomasz Guzik
- Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, UK
- Department of Medicine, Jagiellonian University, Collegium Medicum, Krakow, Poland
| | - Edina Cenko
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Ana Djordjevic-Dikic
- Medical Faculty, Cardiology Clinic, University Clinical Center, University of Belgrade, Serbia
| | - Filippo Crea
- Department of Cardiology and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
11
|
Lui A, Patel RS, Krause-Hauch M, Sparks RP, Patel NA. Regulation of Human Sortilin Alternative Splicing by Glucagon-like Peptide-1 (GLP1) in Adipocytes. Int J Mol Sci 2023; 24:14324. [PMID: 37762628 PMCID: PMC10531797 DOI: 10.3390/ijms241814324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Type 2 diabetes mellitus is a chronic metabolic disease with no cure. Adipose tissue is a major site of systemic insulin resistance. Sortilin is a central component of the glucose transporter -Glut4 storage vesicles (GSV) which translocate to the plasma membrane to uptake glucose from circulation. Here, using human adipocytes we demonstrate the presence of the alternatively spliced, truncated sortilin variant (Sort_T) whose expression is significantly increased in diabetic adipose tissue. Artificial-intelligence-based modeling, molecular dynamics, intrinsically disordered region analysis, and co-immunoprecipitation demonstrated association of Sort_T with Glut4 and decreased glucose uptake in adipocytes. The results show that glucagon-like peptide-1 (GLP1) hormone decreases Sort_T. We deciphered the molecular mechanism underlying GLP1 regulation of alternative splicing of human sortilin. Using splicing minigenes and RNA-immunoprecipitation assays, the results show that GLP1 regulates Sort_T alternative splicing via the splice factor, TRA2B. We demonstrate that targeted antisense oligonucleotide morpholinos reduces Sort_T levels and improves glucose uptake in diabetic adipocytes. Thus, we demonstrate that GLP1 regulates alternative splicing of sortilin in human diabetic adipocytes.
Collapse
Affiliation(s)
- Ashley Lui
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA; (A.L.); (M.K.-H.)
| | - Rekha S. Patel
- Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (R.S.P.); (R.P.S.)
| | - Meredith Krause-Hauch
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA; (A.L.); (M.K.-H.)
| | - Robert P. Sparks
- Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (R.S.P.); (R.P.S.)
- Department of Medicine, Division of Gastroenterology, UMass Chan Medical School, Worcester, MA 01655, USA
| | - Niketa A. Patel
- Department of Molecular Medicine, University of South Florida, Tampa, FL 33612, USA; (A.L.); (M.K.-H.)
- Research Service, James A. Haley Veterans Hospital, Tampa, FL 33612, USA; (R.S.P.); (R.P.S.)
| |
Collapse
|
12
|
Petrovic A, Igrec D, Rozac K, Bojanic K, Kuna L, Kolaric TO, Mihaljevic V, Sikora R, Smolic R, Glasnovic M, Wu GY, Smolic M. The Role of GLP1-RAs in Direct Modulation of Lipid Metabolism in Hepatic Tissue as Determined Using In Vitro Models of NAFLD. Curr Issues Mol Biol 2023; 45:4544-4556. [PMID: 37367037 DOI: 10.3390/cimb45060288] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/18/2023] [Accepted: 05/22/2023] [Indexed: 06/28/2023] Open
Abstract
Glucagon-like peptide 1 receptor agonists (GLP-1RAs) have been shown to improve glucose and lipid homeostasis, promote weight loss, and reduce cardiovascular risk factors. They are a promising therapeutic option for non-alcoholic fatty liver disease (NAFLD), the most common liver disease, associated with T2DM, obesity, and metabolic syndrome. GLP-1RAs have been approved for the treatment of T2DM and obesity, but not for NAFLD. Most recent clinical trials have suggested the importance of early pharmacologic intervention with GLP-1RAs in alleviating and limiting NAFLD, as well as highlighting the relative scarcity of in vitro studies on semaglutide, indicating the need for further research. However, extra-hepatic factors contribute to the GLP-1RA results of in vivo studies. Cell culture models of NAFLD can be helpful in eliminating extrahepatic effects on the alleviation of hepatic steatosis, modulation of lipid metabolism pathways, reduction of inflammation, and prevention of the progression of NAFLD to severe hepatic conditions. In this review article, we discuss the role of GLP-1 and GLP-1RA in the treatment of NAFLD using human hepatocyte models.
Collapse
Affiliation(s)
- Ana Petrovic
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Dunja Igrec
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Karla Rozac
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Kristina Bojanic
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Health Center Osijek-Baranja County, 31000 Osijek, Croatia
| | - Lucija Kuna
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Tea Omanovic Kolaric
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Vjera Mihaljevic
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Renata Sikora
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Health Center Osijek-Baranja County, 31000 Osijek, Croatia
| | - Robert Smolic
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Marija Glasnovic
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - George Y Wu
- Department of Medicine, Division of Gastrenterology/Hepatology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Martina Smolic
- Faculty of Dental Medicine and Health Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Faculty of Medicine Osijek, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| |
Collapse
|
13
|
Mattar P, Jaque C, Teske JA, Morselli E, Kerr B, Cortés V, Baudrand R, Perez-Leighton CE. Impact of short and long exposure to cafeteria diet on food intake and white adipose tissue lipolysis mediated by glucagon-like peptide 1 receptor. Front Endocrinol (Lausanne) 2023; 14:1164047. [PMID: 37293487 PMCID: PMC10244886 DOI: 10.3389/fendo.2023.1164047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/05/2023] [Indexed: 06/10/2023] Open
Abstract
Introduction The modern food environment facilitates excessive calorie intake, a major driver of obesity. Glucagon-like peptide 1 (GLP1) is a neuroendocrine peptide that has been the basis for developing new pharmacotherapies against obesity. The GLP1 receptor (GLP1R) is expressed in central and peripheral tissues, and activation of GLP1R reduces food intake, increases the expression of thermogenic proteins in brown adipose tissue (BAT), and enhances lipolysis in white adipose tissue (WAT). Obesity decreases the efficiency of GLP1R agonists in reducing food intake and body weight. Still, whether palatable food intake before or during the early development of obesity reduces the effects of GLP1R agonists on food intake and adipose tissue metabolism remains undetermined. Further, whether GLP1R expressed in WAT contributes to these effects is unclear. Methods Food intake, expression of thermogenic BAT proteins, and WAT lipolysis were measured after central or peripheral administration of Exendin-4 (EX4), a GLP1R agonist, to mice under intermittent-short exposure to CAF diet (3 h/d for 8 days) or a longer-continuous exposure to CAF diet (24 h/d for 15 days). Ex-vivo lipolysis was measured after EX4 exposure to WAT samples from mice fed CAF or control diet for 12 weeks. . Results During intermittent-short exposure to CAF diet (3 h/d for 8 days), third ventricle injection (ICV) and intra-peritoneal administration of EX4 reduced palatable food intake. Yet, during a longer-continuous exposure to CAF diet (24 h/d for 15 days), only ICV EX4 administration reduced food intake and body weight. However, this exposure to CAF diet blocked the increase in uncoupling protein 1 (UCP1) caused by ICV EX4 administration in mice fed control diet. Finally, GLP1R expression in WAT was minimal, and EX4 failed to increase lipolysis ex-vivo in WAT tissue samples from mice fed CAF or control diet for 12 weeks. . Discussion Exposure to a CAF diet during the early stages of obesity reduces the effects of peripheral and central GLP1R agonists, and WAT does not express a functional GLP1 receptor. These data support that exposure to the obesogenic food environment, without the development or manifestation of obesity, can alter the response to GLP1R agonists. .
Collapse
Affiliation(s)
- Pamela Mattar
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian Jaque
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jennifer A. Teske
- Department of Physiology, School of Nutritional Sciences and Wellness, Graduate Interdisciplinary Programs in Physiological Sciences and Neuroscience, University of Arizona, Tucson, AZ, United States
- Department of Food Science and Nutrition at the University of Minnesota, Saint Paul, MN, United States
| | - Eugenia Morselli
- Department of Basic Sciences, Faculty of Medicine and Sciences, Universidad San Sebastián, Santiago, Chile
| | - Bredford Kerr
- Centro de Biología Celular y Biomedicina-CEBICEM, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Víctor Cortés
- Department of Nutrition, Diabetes, and Metabolism, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rene Baudrand
- Department of Endocrinology, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
- Centro Traslacional de Endocrinologia UC CETREN, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | | |
Collapse
|
14
|
Garbuzova Striukova EV, Shramko VS, Kashtanova EV, Polonskaya YV, Stakhneva EM, Kurguzov AV, Murashov IS, Chernyavsky AM, Ragino YI. Adipokine-Cytokine Profile in Patients with Unstable Atherosclerotic Plaques and Abdominal Obesity. Int J Mol Sci 2023; 24:ijms24108937. [PMID: 37240282 DOI: 10.3390/ijms24108937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
The goal of the research was to study the levels of adipokines and their associations with unstable atherosclerotic plaques in patients with coronary atherosclerosis and abdominal obesity (AO). METHODS The study included 145 men aged 38-79 with atherosclerosis of the coronary arteries (CA) and stable angina pectoris II-III FC who were hospitalized for coronary bypass surgery (2011-2022). The final analysis included 116 patients. Notably, 70 men had stable plaques in the CA (of which 44.3% had AO), and 46 men had unstable plaques in the CA (of which 43.5% had AO). Adipocytokine levels were determined using multiplex analysis (Human Metabolic Hormone V3 panel). RESULTS In the subgroup of patients with unstable plaques, patients with AO had a GLP-1 level that was 1.5 times higher and a lipocalin-2 level that was 2.1 times lower, respectively. GLP-1 is direct, and lipocalin-2 is inversely associated with AO in patients with unstable plaques. Among patients with AO, the level of lipocalin-2 in patients with unstable plaques was 2.2 times lower than in patients with stable plaques in the CA. The level of lipocalin-2 was inversely associated with the presence of unstable atherosclerotic plaques in the CA. CONCLUSION GLP-1 is directly associated with AO in patients with unstable atherosclerotic plaques. Lipocalin-2 is inversely associated with unstable atherosclerotic plaques in patients with AO.
Collapse
Affiliation(s)
- Evgeniia V Garbuzova Striukova
- Research Institute of Internal and Preventive Medicine-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IIPM-Branch of IC&G SB RAS), B. Bogatkova Str., 175/1, 630089 Novosibirsk, Russia
| | - Victoriya S Shramko
- Research Institute of Internal and Preventive Medicine-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IIPM-Branch of IC&G SB RAS), B. Bogatkova Str., 175/1, 630089 Novosibirsk, Russia
| | - Elena V Kashtanova
- Research Institute of Internal and Preventive Medicine-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IIPM-Branch of IC&G SB RAS), B. Bogatkova Str., 175/1, 630089 Novosibirsk, Russia
| | - Yana V Polonskaya
- Research Institute of Internal and Preventive Medicine-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IIPM-Branch of IC&G SB RAS), B. Bogatkova Str., 175/1, 630089 Novosibirsk, Russia
| | - Ekaterina M Stakhneva
- Research Institute of Internal and Preventive Medicine-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IIPM-Branch of IC&G SB RAS), B. Bogatkova Str., 175/1, 630089 Novosibirsk, Russia
| | - Alexey V Kurguzov
- Federal State Budgetary Institution "National Medical Research Center named after Academician E.N. Meshalkin" Ministry of Health of the Russian Federation, Rechkunovskaya Str., 15, 630055 Novosibirsk, Russia
| | - Ivan S Murashov
- Federal State Budgetary Institution "National Medical Research Center named after Academician E.N. Meshalkin" Ministry of Health of the Russian Federation, Rechkunovskaya Str., 15, 630055 Novosibirsk, Russia
| | - Alexander M Chernyavsky
- Federal State Budgetary Institution "National Medical Research Center named after Academician E.N. Meshalkin" Ministry of Health of the Russian Federation, Rechkunovskaya Str., 15, 630055 Novosibirsk, Russia
| | - Yuliya I Ragino
- Research Institute of Internal and Preventive Medicine-Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (IIPM-Branch of IC&G SB RAS), B. Bogatkova Str., 175/1, 630089 Novosibirsk, Russia
| |
Collapse
|
15
|
Mehdi SF, Pusapati S, Anwar MS, Lohana D, Kumar P, Nandula SA, Nawaz FK, Tracey K, Yang H, LeRoith D, Brownstein MJ, Roth J. Glucagon-like peptide-1: a multi-faceted anti-inflammatory agent. Front Immunol 2023; 14:1148209. [PMID: 37266425 PMCID: PMC10230051 DOI: 10.3389/fimmu.2023.1148209] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/26/2023] [Indexed: 06/03/2023] Open
Abstract
Inflammation contributes to many chronic conditions. It is often associated with circulating pro-inflammatory cytokines and immune cells. GLP-1 levels correlate with disease severity. They are often elevated and can serve as markers of inflammation. Previous studies have shown that oxytocin, hCG, ghrelin, alpha-MSH and ACTH have receptor-mediated anti-inflammatory properties that can rescue cells from damage and death. These peptides have been studied well in the past century. In contrast, GLP-1 and its anti-inflammatory properties have been recognized only recently. GLP-1 has been proven to be a useful adjuvant therapy in type-2 diabetes mellitus, metabolic syndrome, and hyperglycemia. It also lowers HbA1C and protects cells of the cardiovascular and nervous systems by reducing inflammation and apoptosis. In this review we have explored the link between GLP-1, inflammation, and sepsis.
Collapse
Affiliation(s)
- Syed Faizan Mehdi
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Suma Pusapati
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Muhammad Saad Anwar
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Durga Lohana
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Parkash Kumar
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | | | - Fatima Kausar Nawaz
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Kevin Tracey
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Huan Yang
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Derek LeRoith
- Division of Endocrinology, Diabetes & Bone Disease, Icahn School of Medicine at Mt. Sinai, New York, NY, United States
| | | | - Jesse Roth
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, United States
| |
Collapse
|
16
|
Morais T, Pereira SS, Andrade S, Neves D, Guimarães M, Nora M, Carreira MC, Casanueva FF, Monteiro MP. GLP-1 Increases Circulating Leptin Levels in Truncal Vagotomized Rats. Biomedicines 2023; 11:biomedicines11051322. [PMID: 37238993 DOI: 10.3390/biomedicines11051322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
GLP-1 is a gastro-intestinal hormone acting within the gut/brain axis for energy balance regulation. We aimed to evaluate the role of the vagus nerve in whole-body energy homeostasis and in mediating GLP-1 effects. For this, rats submitted to truncal vagotomy and sham-operated controls underwent a comprehensive evaluation, including eating behavior, body weight, percentage of white (WAT) and brown adipose tissue (BAT), resting energy expenditure (REE) and acute response to GLP-1. Truncal vagotomized rats had significantly lower food intake, body weight, body weight gain, WAT and BAT, with a higher BAT/WAT ratio, but no significant difference in REE when compared to controls. Vagotomized rats also had significantly higher fasting ghrelin and lower glucose and insulin levels. After GLP-1 administration, vagotomized rats depicted a blunted anorexigenic response and higher plasma leptin levels, as compared to controls. However, in vitro stimulation of VAT explants with GLP-1 resulted in no significant changes in leptin secretion. In conclusion, the vagus nerve influences whole-body energy homeostasis by modifying food intake, body weight and body composition and by mediating the GLP-1 anorectic response. The higher leptin levels in response to acute GLP-1 administration observed after truncal vagotomy suggest the existence of a putative GLP-1-leptin axis that relies on the integrity of gut-brain vagal pathway.
Collapse
Affiliation(s)
- Tiago Morais
- Endocrine and Metabolic Research, UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
| | - Sofia S Pereira
- Endocrine and Metabolic Research, UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
| | - Sara Andrade
- Endocrine and Metabolic Research, UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
| | - Diogo Neves
- Endocrine and Metabolic Research, UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
| | - Marta Guimarães
- Endocrine and Metabolic Research, UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Department of General Surgery, Centro Hospitalar de Entre o Douro e Vouga, 4520-220 Santa Maria da Feira, Portugal
| | - Mário Nora
- Endocrine and Metabolic Research, UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Department of General Surgery, Centro Hospitalar de Entre o Douro e Vouga, 4520-220 Santa Maria da Feira, Portugal
| | - Marcos C Carreira
- CIBER de Fisiopatologia Obesidad y Nutricion (CB06/03), Instituto Salud Carlos III, 15706 Santiago de Compostela, Spain
- Department of Medicine, USC University Hospital Complex, University of Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Felipe F Casanueva
- CIBER de Fisiopatologia Obesidad y Nutricion (CB06/03), Instituto Salud Carlos III, 15706 Santiago de Compostela, Spain
- Department of Medicine, USC University Hospital Complex, University of Santiago de Compostela, 15705 Santiago de Compostela, Spain
| | - Mariana P Monteiro
- Endocrine and Metabolic Research, UMIB-Unit for Multidisciplinary Research in Biomedicine, ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
17
|
Morais T, Seabra AL, Patrício BG, Carrageta DF, Guimarães M, Nora M, Oliveira PF, Alves MG, Monteiro MP. Dysglycemia Shapes Visceral Adipose Tissue's Response to GIP, GLP-1 and Glucagon in Individuals with Obesity. Metabolites 2023; 13:metabo13050587. [PMID: 37233628 DOI: 10.3390/metabo13050587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/01/2023] [Accepted: 04/22/2023] [Indexed: 05/27/2023] Open
Abstract
Visceral adipose tissue (VAT) metabolic fingerprints differ according to body mass index (BMI) and glycemic status. Glucagon-like peptide 1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP) and glucagon are gut-associated hormones that play an important role in regulating energy and glucose homeostasis, although their metabolic actions in VAT are still poorly characterized. Our aim was to assess whether GLP-1, GIP and glucagon influence the VAT metabolite profile. To achieve this goal, VAT harvested during elective surgical procedures from individuals (N = 19) with different BMIs and glycemic statuses was stimulated with GLP-1, GIP or glucagon, and culture media was analyzed using proton nuclear magnetic resonance. In the VAT of individuals with obesity and prediabetes, GLP-1 shifted its metabolic profile by increasing alanine and lactate production while also decreasing isoleucine consumption, whereas GIP and glucagon decreased lactate and alanine production and increased pyruvate consumption. In summary, GLP-1, GIP and glucagon were shown to distinctively modulate the VAT metabolic profile depending on the subject's BMI and glycemic status. In VAT from patients with obesity and prediabetes, these hormones induced metabolic shifts toward gluconeogenesis suppression and oxidative phosphorylation enhancement, suggesting an overall improvement in AT mitochondrial function.
Collapse
Affiliation(s)
- Tiago Morais
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
| | - Alexandre L Seabra
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
| | - Bárbara G Patrício
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
| | - David F Carrageta
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Laboratory of Physiology, Department of Imuno-Physiology and Pharmacology, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| | - Marta Guimarães
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Department of General Surgery, Centro Hospitalar de Entre o Douro e Vouga, 4520-220 Santa Maria da Feira, Portugal
| | - Mário Nora
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Department of General Surgery, Centro Hospitalar de Entre o Douro e Vouga, 4520-220 Santa Maria da Feira, Portugal
| | - Pedro F Oliveira
- QOPNA & LAQV, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Marco G Alves
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
- Laboratory of Physiology, Department of Imuno-Physiology and Pharmacology, ICBAS-School of Medicine and Biomedical Sciences, University of Porto, 4050-313 Porto, Portugal
| | - Mariana P Monteiro
- Endocrine and Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal
- Laboratory for Integrative and Translational Research in Population Health (ITR), University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
18
|
Vaittinen M, Ilha M, Herbers E, Wagner A, Virtanen KA, Pietiläinen KH, Pirinen E, Pihlajamäki J. Liraglutide demonstrates a therapeutic effect on mitochondrial dysfunction in human SGBS adipocytes in vitro. Diabetes Res Clin Pract 2023; 199:110635. [PMID: 36958431 DOI: 10.1016/j.diabres.2023.110635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 02/28/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
AIMS Liraglutide (LG), a glucagon-like peptide-1 receptor (GLP-1R) agonist, has been shown to improve white adipose tissue mitochondrial metabolism in mice but not in human adipocytes. Therefore, we explored whether LG has therapeutic efficacy in mitochondrial dysfunction in human adipocytes in vitro. METHODS We tested the effects of short-term (ST-LG: 24 h) and long-term (LT-LG: D0-15 days) treatments in human SGBS adipocytes on mitochondrial respiration, mRNA and protein expression. GLP-1R inhibition was investigated by the co-treatment of GLP-1R inhibitor, exendin 9-39 (Ex9-39) and ST-LG treatment. We also explored the ability of ST-LG to alleviate mitochondrial dysfunction induced by tumor necrosis factor-alpha (TNFα). RESULTS LT-LG treatment induced the formation of smaller lipid droplets and increased the expression of genes related to lipolysis. Both ST-LG and LT-LG treatments promoted mitochondrial respiration. Additionally, LT-LG treatment increased the expression of a brown adipocyte marker, uncoupling protein 1 (UCP-1), and the markers of mitochondrial biogenesis. Interestingly, ST-LG rescued TNFα-induced defects in mitochondrial energy metabolism and inflammation in SGBS adipocytes. CONCLUSION LG stimulates mitochondrial respiration and biogenesis in human adipocytes, potentially via UCP-1-mediated adipocyte browning. Importantly, our study demonstrates for the first time that LG has a therapeutic potential on mitochondrial activity in human adipocytes.
Collapse
Affiliation(s)
- Maija Vaittinen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland.
| | - Mariana Ilha
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Elena Herbers
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland
| | - Anita Wagner
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00290 Helsinki, Finland; Research Unit for Internal Medicine, Faculty of Medicine, University of Oulu, FIN-90220 Oulu, Finland
| | - Kirsi A Virtanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Endocrinology, and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland; Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Finland; Obesity Center, Abdominal Center, Helsinki University Hospital and University of Helsinki, Finland
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, FIN-00290 Helsinki, Finland; Research Unit for Internal Medicine, Faculty of Medicine, University of Oulu, FIN-90220 Oulu, Finland
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Medicine, Endocrinology, and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
19
|
Tavares G, Rosendo-Silva D, Simões F, Eickhoff H, Marques D, Sacramento JF, Capucho AM, Seiça R, Conde SV, Matafome P. Circulating Dopamine Is Regulated by Dietary Glucose and Controls Glucagon-like 1 Peptide Action in White Adipose Tissue. Int J Mol Sci 2023; 24:ijms24032464. [PMID: 36768789 PMCID: PMC9916853 DOI: 10.3390/ijms24032464] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/21/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Dopamine directly acts in the liver and white adipose tissue (WAT) to regulate insulin signaling, glucose uptake, and catabolic activity. Given that dopamine is secreted by the gut and regulates insulin secretion in the pancreas, we aimed to determine its regulation by nutritional cues and its role in regulating glucagon-like peptide 1 (GLP-1) action in WAT. Solutions with different nutrients were administered to Wistar rats and postprandial dopamine levels showed elevations following a mixed meal and glucose intake. In high-fat diet-fed diabetic Goto-Kakizaki rats, sleeve gastrectomy upregulated dopaminergic machinery, showing the role of the gut in dopamine signaling in WAT. Bromocriptine treatment in the same model increased GLP-1R in WAT, showing the role of dopamine in regulating GLP-1R. By contrast, treatment with the GLP-1 receptor agonist Liraglutide had no impact on dopamine receptors. GLP-1 and dopamine crosstalk was shown in rat WAT explants, since dopamine upregulated GLP-1-induced AMPK activity in mesenteric WAT in the presence of the D2R and D3R inhibitor Domperidone. In human WAT, dopamine receptor 1 (D1DR) and GLP-1R expression were correlated. Our results point out a dietary and gut regulation of plasma dopamine, acting in the WAT to regulate GLP-1 action. Together with the known dopamine action in the pancreas, such results may identify new therapeutic opportunities to improve metabolic control in metabolic disorders.
Collapse
Affiliation(s)
- Gabriela Tavares
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical-Academic Center of Coimbra, 3004-531 Coimbra, Portugal
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Daniela Rosendo-Silva
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical-Academic Center of Coimbra, 3004-531 Coimbra, Portugal
| | - Flávia Simões
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Hans Eickhoff
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Daniela Marques
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Joana F. Sacramento
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Adriana M. Capucho
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Raquel Seiça
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical-Academic Center of Coimbra, 3004-531 Coimbra, Portugal
| | - Sílvia V. Conde
- NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, 1169-056 Lisbon, Portugal
| | - Paulo Matafome
- Institute of Physiology and Institute of Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical-Academic Center of Coimbra, 3004-531 Coimbra, Portugal
- Instituto Politécnico de Coimbra, Coimbra Health School, 3046-854 Coimbra, Portugal
- Correspondence:
| |
Collapse
|
20
|
Patel R, Parmar N, Palit SP, Rathwa N, Begum R. A novel combination of sitagliptin and melatonin ameliorates T2D manifestations: studies on experimental diabetic models. J Endocrinol Invest 2023:10.1007/s40618-023-02014-6. [PMID: 36692817 DOI: 10.1007/s40618-023-02014-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 01/16/2023] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Type 2 diabetes (T2D) is an endocrine disorder characterized by hyperglycemia, insulin resistance, dysregulated glucose and lipid metabolism, reduced pancreatic β-cell function and mass, and a reduced incretin effect. Circadian rhythm disruption is associated with increased T2D risk. We have investigated the therapeutic potential of a combination of melatonin (M) and sitagliptin (S), a dipeptidyl peptidase IV (DPP-IV) inhibitor, in the amelioration of T2D manifestations in high-fat diet (HFD) induced T2D mouse model and also on β-cell proliferation under gluco-lipotoxicity stress in vitro. METHODS For in vivo study, mice were fed with HFD for 25 weeks to induce T2D and were treated with monotherapies and S + M for four weeks. For the in vitro study, primary mouse islets were exposed to normal glucose and high glucose + palmitate to induce gluco-lipotoxic stress. RESULTS Our results suggest that monotherapies and S + M improve metabolic parameters and glyco-lipid metabolism in the liver and adipose tissue, respectively, and improve mitochondrial function in the skeletal muscle. Moreover, it increases peripheral insulin sensitivity. Our in vitro and in vivo studies suggest that β-cell mass was preserved in all the drug-treated groups. CONCLUSION The combination treatment is superior to monotherapies in the management of T2D.
Collapse
Affiliation(s)
- R Patel
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - N Parmar
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - S P Palit
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - N Rathwa
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India
| | - R Begum
- Department of Biochemistry, Faculty of Science, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat, 390002, India.
| |
Collapse
|
21
|
Zhu R, Chen S. Proteomic analysis reveals semaglutide impacts lipogenic protein expression in epididymal adipose tissue of obese mice. Front Endocrinol (Lausanne) 2023; 14:1095432. [PMID: 37025414 PMCID: PMC10070826 DOI: 10.3389/fendo.2023.1095432] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/28/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Obesity is a global health problem with few pharmacologic options. Semaglutide is a glucagon-like peptide-1 (GLP-1) analogue that induces weight loss. Yet, the role of semaglutide in adipose tissue has not yet been examined. The following study investigated the mechanism of semaglutide on lipid metabolism by analyzing proteomics of epididymal white adipose tissue (eWAT) in obese mice. METHODS A total of 36 C57BL/6JC mice were randomly divided into a normal-chow diet group (NCD, n = 12), high-fat diet (HFD, n = 12), and HFD+semaglutide group (Sema, n = 12). Mice in the Sema group were intraperitoneally administered semaglutide, and the HFD group and the NCD group were intraperitoneally administered an equal volume of normal saline. Serum samples were collected to detect fasting blood glucose and blood lipids. The Intraperitoneal glucose tolerance test (IPGTT) was used to measure the blood glucose value at each time point and calculate the area under the glucose curve. Tandem Mass Tag (TMT) combined with liquid chromatography-tandem mass spectrometry (LC-MS/MS) were used to study the expression of eWAT, while cellular processes, biological processes, corresponding molecular functions, and related network molecular mechanisms were analyzed by bioinformatics. RESULTS Compared with the model group, the semaglutide-treated mice presented 640 differentially expressed proteins (DEPs), including 292 up-regulated and 348 down-regulated proteins. Bioinformatics analysis showed a reduction of CD36, FABP5, ACSL, ACOX3, PLIN2, ANGPTL4, LPL, MGLL, AQP7, and PDK4 involved in the lipid metabolism in the Sema group accompanied by a decrease in visceral fat accumulation, blood lipids, and improvement in glucose intolerance. CONCLUSION Semaglutide can effectively reduce visceral fat and blood lipids and improve glucose metabolism in obese mice. Semaglutide treatment might have beneficial effects on adipose tissues through the regulation of lipid uptake, lipid storage, and lipolysis in white adipose tissue.
Collapse
Affiliation(s)
- Ruiyi Zhu
- Department of Internal Medical, Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Internal Medical, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Shuchun Chen
- Department of Internal Medical, Hebei Medical University, Shijiazhuang, Hebei, China
- Department of Internal Medical, Hebei General Hospital, Shijiazhuang, Hebei, China
- *Correspondence: Shuchun Chen,
| |
Collapse
|
22
|
Martins FF, Marinho TS, Cardoso LEM, Barbosa-da-Silva S, Souza-Mello V, Aguila MB, Mandarim-de-Lacerda CA. Semaglutide (GLP-1 receptor agonist) stimulates browning on subcutaneous fat adipocytes and mitigates inflammation and endoplasmic reticulum stress in visceral fat adipocytes of obese mice. Cell Biochem Funct 2022; 40:903-913. [PMID: 36169111 DOI: 10.1002/cbf.3751] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/24/2022] [Accepted: 09/13/2022] [Indexed: 12/15/2022]
Abstract
Semaglutide (GLP-1 agonist) was approved for treating obesity. Although the effects on weight loss and metabolism are known, the responses of adipocytes to semaglutide are yet limited. C57BL/6 male mice (n = 20/group) were fed a control diet (C) or a high-fat (HF) diet for 16 weeks and then separated into four groups (n = 10/group) for an additional four weeks: C, C diet and semaglutide, HF, and HF diet and semaglutide. Epididymal white adipose tissue (eWAT) and subcutaneous white adipose tissue (sWAT) fat pads were studied with biochemistry, immunohistochemistry/fluorescence, stereology, and reverse transcription-quantitative polymerase chain reaction. In obese mice, semaglutide reduced the fat pad masses (eWAT, -55%; sWAT, -40%), plasmatic cytokines, and proinflammatory gene expressions: tumor necrosis factor-alpha (-60%); interleukin (IL)-6 (-55%); IL-1 beta (-40%); monocyte chemoattractant protein-1 (-90%); and leptin (-80%). Semaglutide also lessened endoplasmic reticulum (ER) stress genes of activating transcription factor-4 (-85%), CCAAT enhancer-binding protein homologous protein (-55%), and growth arrest and DNA damage-inducible gene 45 (-45%). The obese mice's adipocyte hypertrophy and macrophage infiltration were equally reduced by semaglutide. Semaglutide enhanced multiloculation and uncoupled protein 1 (UCP1) labeling in obese mice: peroxisome proliferator-activated receptor-alpha (+560%) and gamma (+150%), fibronectin type III domain-containing protein 5 (+215%), peroxisome proliferator-activated receptor-alpha coactivator (+110%), nuclear respiratory factor 1 (+260%), and mitochondrial transcription factor A (+120%). Semaglutide also increased thermogenetic gene expressions for the browning phenotype maintenance: beta-3 adrenergic receptor (+520%), PR domain containing 16 (+90%), and Ucp1 (+110%). In conclusion, semaglutide showed significant beneficial effects beyond weight loss, directly on fat pads and adipocytes of obese mice, remarkably anti-inflammatory, and reduced adipocyte size and ER stress. Besides, semaglutide activated adipocyte browning, improving UCP1, mitochondrial biogenesis, and thermogenic marker expressions help weight loss.
Collapse
Affiliation(s)
- Fabiane F Martins
- Biomedical Center, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thatiany S Marinho
- Biomedical Center, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz E M Cardoso
- Biomedical Center, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sandra Barbosa-da-Silva
- Biomedical Center, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Biomedical Center, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia B Aguila
- Biomedical Center, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Biomedical Center, Laboratory of Morphometry, Metabolism and Cardiovascular Diseases, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Wu Y, Sun B, Guo X, Wu L, Hu Y, Qin L, Yang T, Li M, Qin T, Jiang M, Liu T. Zishen Pill alleviates diabetes in Db/db mice via activation of PI3K/AKT pathway in the liver. Chin Med 2022; 17:128. [PMID: 36352450 PMCID: PMC9647929 DOI: 10.1186/s13020-022-00683-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
Background The rising global incidence of type 2 diabetes mellitus (T2DM) highlights a need for new therapies. The Zishen Pill (ZSP) is a traditional Chinese herbal decoction that has previously shown hypoglycemic effects in C57BL/KsJ-db/db mice, although the therapeutic mechanism remains unknown. This study aims to explore the underlying mechanisms of ZSP’s hypoglycemic effects using db/db mice. Methods Db/db mice were divided into two groups: model group and ZSP group, while wt/wt mice were used as a normal control. ZSP was given to mice by gavage for 40 days. During treatment, blood glucose level and body weight were monitored continuously. Oral glucose tolerance test (OGTT) was performed at day 35. Blood and tissue samples were collected at the end of treatment for further analyses. Mice liver samples were analyzed with mRNA transcriptomics using functional annotation and pathway enrichment to identify potential mechanisms that were then explored with qPCR and Western Blot techniques. Results ZSP treatment significantly reduced weight gain and glycemic severity in db/db mice. ZSP also partially restored the glucose homeostasis in db/db mice and increased the hepatic glycogen content. Transcriptomic analyses showed ZSP increased expression of genes involved in glycolysis including Hk2, Hk3, Gck and Pfkb1, and decreased expression of G6pase. Additionally, the gene and protein expression of phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway, and Csf1 and Flt3 mRNA expression were significantly upregulated in ZSP group. Conclusion ZSP treatment reduced the severity of diabetic symptoms in db/db mice. ZSP increased expression of genes associated with glycogen synthesis and glycolysis, and decreased gluconeogenesis via the enhancement of the PI3K/AKT signaling in the liver. Supplementary Information The online version contains supplementary material available at 10.1186/s13020-022-00683-8.
Collapse
|
24
|
Anti-Inflammatory Effects of GLP-1 Receptor Activation in the Brain in Neurodegenerative Diseases. Int J Mol Sci 2022; 23:ijms23179583. [PMID: 36076972 PMCID: PMC9455625 DOI: 10.3390/ijms23179583] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/17/2022] [Accepted: 08/21/2022] [Indexed: 11/16/2022] Open
Abstract
The glucagon-like peptide-1 (GLP-1) is a pleiotropic hormone well known for its incretin effect in the glucose-dependent stimulation of insulin secretion. However, GLP-1 is also produced in the brain and displays a critical role in neuroprotection and inflammation by activating the GLP-1 receptor signaling pathways. Several studies in vivo and in vitro using preclinical models of neurodegenerative diseases show that GLP-1R activation has anti-inflammatory properties. This review explores the molecular mechanistic action of GLP-1 RAS in relation to inflammation in the brain. These findings update our knowledge of the potential benefits of GLP-1RAS actions in reducing the inflammatory response. These molecules emerge as a potential therapeutic tool in treating neurodegenerative diseases and neuroinflammatory pathologies.
Collapse
|
25
|
Epicardial Adipose Tissue: A Novel Potential Imaging Marker of Comorbidities Caused by Chronic Inflammation. Nutrients 2022; 14:nu14142926. [PMID: 35889883 PMCID: PMC9316118 DOI: 10.3390/nu14142926] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/09/2022] [Indexed: 12/11/2022] Open
Abstract
The observation of correlations between obesity and chronic metabolic and cardiovascular diseases has led to the emergence of strong interests in “adipocyte biology”, in particular in relation to a specific visceral adipose tissue that is the epicardial adipose tissue (EAT) and its pro-inflammatory role. In recent years, different imaging techniques frequently used in daily clinical practice have tried to obtain an EAT quantification. We provide a useful update on comorbidities related to chronic inflammation typical of cardiac adiposity, analyzing how the EAT assessment could impact and provide data on the patient prognosis. We assessed for eligibility 50 papers, with a total of 10,458 patients focusing the review on the evaluation of EAT in two main contexts: cardiovascular and metabolic diseases. Given its peculiar properties and rapid responsiveness, EAT could act as a marker to investigate the basal risk factor and follow-up conditions. In the future, EAT could represent a therapeutic target for new medications. The assessment of EAT should become part of clinical practice to help clinicians to identify patients at greater risk of developing cardiovascular and/or metabolic diseases and to provide information on their clinical and therapeutic outcomes.
Collapse
|
26
|
Li M, Chi X, Wang Y, Setrerrahmane S, Xie W, Xu H. Trends in insulin resistance: insights into mechanisms and therapeutic strategy. Signal Transduct Target Ther 2022; 7:216. [PMID: 35794109 PMCID: PMC9259665 DOI: 10.1038/s41392-022-01073-0] [Citation(s) in RCA: 186] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 02/06/2023] Open
Abstract
The centenary of insulin discovery represents an important opportunity to transform diabetes from a fatal diagnosis into a medically manageable chronic condition. Insulin is a key peptide hormone and mediates the systemic glucose metabolism in different tissues. Insulin resistance (IR) is a disordered biological response for insulin stimulation through the disruption of different molecular pathways in target tissues. Acquired conditions and genetic factors have been implicated in IR. Recent genetic and biochemical studies suggest that the dysregulated metabolic mediators released by adipose tissue including adipokines, cytokines, chemokines, excess lipids and toxic lipid metabolites promote IR in other tissues. IR is associated with several groups of abnormal syndromes that include obesity, diabetes, metabolic dysfunction-associated fatty liver disease (MAFLD), cardiovascular disease, polycystic ovary syndrome (PCOS), and other abnormalities. Although no medication is specifically approved to treat IR, we summarized the lifestyle changes and pharmacological medications that have been used as efficient intervention to improve insulin sensitivity. Ultimately, the systematic discussion of complex mechanism will help to identify potential new targets and treat the closely associated metabolic syndrome of IR.
Collapse
Affiliation(s)
- Mengwei Li
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | - Xiaowei Chi
- Development Center for Medical Science & Technology National Health Commission of the People's Republic of China, 100044, Beijing, China
| | - Ying Wang
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | | | - Wenwei Xie
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, 210009, China
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China
| | - Hanmei Xu
- The Engineering Research Center of Synthetic Peptide Drug Discovery and Evaluation of Jiangsu Province, China Pharmaceutical University, Nanjing, 210009, China.
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
27
|
Helgeland Ø, Vaudel M, Sole-Navais P, Flatley C, Juodakis J, Bacelis J, Koløen IL, Knudsen GP, Johansson BB, Magnus P, Kjennerud TR, Juliusson PB, Stoltenberg C, Holmen OL, Andreassen OA, Jacobsson B, Njølstad PR, Johansson S. Characterization of the genetic architecture of infant and early childhood body mass index. Nat Metab 2022; 4:344-358. [PMID: 35315439 DOI: 10.1038/s42255-022-00549-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 02/09/2022] [Indexed: 02/08/2023]
Abstract
Early childhood obesity is a growing global concern; however, the role of common genetic variation on infant and child weight development is unclear. Here, we identify 46 loci associated with early childhood body mass index at specific ages, matching different child growth phases, and representing four major trajectory patterns. We perform genome-wide association studies across 12 time points from birth to 8 years in 28,681 children and their parents (27,088 mothers and 26,239 fathers) in the Norwegian Mother, Father and Child Cohort Study. Monogenic obesity genes are overrepresented near identified loci, and several complex association signals near LEPR, GLP1R, PCSK1 and KLF14 point towards a major influence for common variation affecting the leptin-melanocortin system in early life, providing a link to putative treatment strategies. We also demonstrate how different polygenic risk scores transition from birth to adult profiles through early child growth. In conclusion, our results offer a fine-grained characterization of a changing genetic landscape sustaining early childhood growth.
Collapse
Affiliation(s)
- Øyvind Helgeland
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Genetics and Bioinformatics, Health Data and Digitalization, Norwegian Institute of Public Health, Oslo, Norway
| | - Marc Vaudel
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pol Sole-Navais
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christopher Flatley
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Julius Juodakis
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jonas Bacelis
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingvild L Koløen
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | | | - Bente B Johansson
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Per Magnus
- Centre for Fertility and Health, Norwegian Institute of Public Health, Oslo, Norway
| | - Ted Reichborn Kjennerud
- Department of Mental Disorders, Norwegian Institute of Public Health, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Petur B Juliusson
- Department of Health Registry Research and Development, National Institute of Public Health, Bergen, Norway
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
| | | | - Oddgeir L Holmen
- HUNT Research Centre, Department of Public Health and Nursing, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ole A Andreassen
- NORMENT Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Mental Health and Addiction, Oslo University Hospital, Oslo, Norway
| | - Bo Jacobsson
- Department of Genetics and Bioinformatics, Health Data and Digitalization, Norwegian Institute of Public Health, Oslo, Norway
- Department of Obstetrics and Gynecology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pål R Njølstad
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway.
| | - Stefan Johansson
- Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway.
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
28
|
Iacobellis G, Baroni MG. Cardiovascular risk reduction throughout GLP-1 receptor agonist and SGLT2 inhibitor modulation of epicardial fat. J Endocrinol Invest 2022; 45:489-495. [PMID: 34643917 DOI: 10.1007/s40618-021-01687-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/01/2021] [Indexed: 12/17/2022]
Abstract
Epicardial adipose tissue is a novel cardiovascular risk factor. It plays a role in the progression of coronary artery disease, heart failure and atrial fibrillation. Given its rapid metabolism, clinical measurability, and modifiability, epicardial fat works well as therapeutic target of drugs modulating the adipose tissue. Epicardial fat responds to glucagon-like peptide 1 receptor agonists (GLP1A) and sodium glucose co-transporter 2 inhibitors (SGLT2i). GLP-1A and SGLT2i provide weight loss and cardiovascular protective effects beyond diabetes control, as recently demonstrated. The potential of modulating the epicardial fat morphology and genetic profile with targeted pharmacological agents can open new avenues in the pharmacotherapy of diabetes and obesity, with particular focus on cardiovascular risk reduction.
Collapse
Affiliation(s)
- G Iacobellis
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, 1400 NW 10th Ave, Dominion Tower suite 805-807, Miami, FL, 33136, USA.
| | - M G Baroni
- Endocrinology and Diabetes, Department of Clinical Medicine, Public Health, Life and Environmental Sciences (MeSVA), University of L'Aquila, L'Aquila, Italy
| |
Collapse
|
29
|
Dysregulated Epicardial Adipose Tissue as a Risk Factor and Potential Therapeutic Target of Heart Failure with Preserved Ejection Fraction in Diabetes. Biomolecules 2022; 12:biom12020176. [PMID: 35204677 PMCID: PMC8961672 DOI: 10.3390/biom12020176] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/12/2022] [Accepted: 01/18/2022] [Indexed: 02/01/2023] Open
Abstract
Cardiovascular (CV) disease and heart failure (HF) are the leading cause of mortality in type 2 diabetes (T2DM), a metabolic disease which represents a fast-growing health challenge worldwide. Specifically, T2DM induces a cluster of systemic metabolic and non-metabolic signaling which may promote myocardium derangements such as inflammation, fibrosis, and myocyte stiffness, which represent the hallmarks of heart failure with preserved ejection fraction (HFpEF). On the other hand, several observational studies have reported that patients with T2DM have an abnormally enlarged and biologically transformed epicardial adipose tissue (EAT) compared with non-diabetic controls. This expanded EAT not only causes a mechanical constriction of the diastolic filling but is also a source of pro-inflammatory mediators capable of causing inflammation, microcirculatory dysfunction and fibrosis of the underlying myocardium, thus impairing the relaxability of the left ventricle and increasing its filling pressure. In addition to representing a potential CV risk factor, emerging evidence shows that EAT may guide the therapeutic decision in diabetic patients as drugs such as metformin, glucagon-like peptide‑1 (GLP-1) receptor agonists and sodium-glucose cotransporter 2 inhibitors (SGLT2-Is), have been associated with attenuation of EAT enlargement.
Collapse
|
30
|
Oliveira FCB, Bauer EJ, Ribeiro CM, Pereira SA, Beserra BTS, Wajner SM, Maia AL, Neves FAR, Coelho MS, Amato AA. Liraglutide Activates Type 2 Deiodinase and Enhances β3-Adrenergic-Induced Thermogenesis in Mouse Adipose Tissue. Front Endocrinol (Lausanne) 2022; 12:803363. [PMID: 35069450 PMCID: PMC8771968 DOI: 10.3389/fendo.2021.803363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/13/2021] [Indexed: 12/02/2022] Open
Abstract
Aims Liraglutide is a long-acting glucagon-like peptide 1 (GLP-1) receptor agonist used as an anti-hyperglycemic agent in type 2 diabetes treatment and recently approved for obesity management. Weight loss is attributed to appetite suppression, but therapy may also increase energy expenditure. To further investigate the effect of GLP-1 signaling in thermogenic fat, we assessed adipose tissue oxygen consumption and type 2 deiodinase (D2) activity in mice treated with liraglutide, both basally and after β3-adrenergic treatment. Methods Male C57BL/6J mice were randomly assigned to receive liraglutide (400 μg/kg, n=12) or vehicle (n=12). After 16 days, mice in each group were co-treated with the selective β3-adrenergic agonist CL316,243 (1 mg/kg, n=6) or vehicle (n=6) for 5 days. Adipose tissue depots were assessed for gene and protein expression, oxygen consumption, and D2 activity. Results Liraglutide increased interscapular brown adipose tissue (iBAT) oxygen consumption and enhanced β3-adrenergic-induced oxygen consumption in iBAT and inguinal white adipose tissue (ingWAT). These effects were accompanied by upregulation of UCP-1 protein levels in iBAT and ingWAT. Notably, liraglutide increased D2 activity without significantly upregulating its mRNA levels in iBAT and exhibited additive effects to β3-adrenergic stimulation in inducing D2 activity in ingWAT. Conclusions Liraglutide exhibits additive effects to those of β3-adrenergic stimulation in thermogenic fat and increases D2 activity in BAT, implying that it may activate this adipose tissue depot by increasing intracellular thyroid activation, adding to the currently known mechanisms of GLP-1A-induced weight loss.
Collapse
Affiliation(s)
- Fernanda C. B. Oliveira
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil
| | - Eduarda J. Bauer
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil
| | - Carolina M. Ribeiro
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil
| | - Sidney A. Pereira
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil
| | - Bruna T. S. Beserra
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil
| | - Simone M. Wajner
- Endocrine Division, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Ana L. Maia
- Endocrine Division, Hospital de Clinicas de Porto Alegre, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Francisco A. R. Neves
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil
| | - Michella S. Coelho
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil
| | - Angelica A. Amato
- Laboratory of Molecular Pharmacology, School of Health Sciences, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
31
|
Iacobellis G, Basilico S, Malavazos AE. Targeting Epicardial Fat in Obesity and Diabetes Pharmacotherapy. Handb Exp Pharmacol 2022; 274:93-108. [PMID: 35156138 DOI: 10.1007/164_2021_577] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Epicardial adipose tissue surrounds and infiltrates the heart. Epicardial fat displays unique anatomic, genetic, and biomolecular properties. People with obesity and in particular, those with abdominal obesity and associated type 2 diabetes mellitus, have an increased amount of epicardial adipose tissue (EAT). Epicardial fat works well as therapeutic target due to its fast-responding metabolism, organ fat specificity, and easy measurability. Epicardial fat responds to thiazolidinediones (TZD), glucagon-like peptide 1-receptor agonists (GLP1A), sodium-glucose cotransporter 2 inhibitors (SGLT2i), dipeptidyl peptidase-4 inhibitors (DPP4i), and statins. Modulating epicardial fat morphology and genetic profile with targeted pharmacological agents suggests novel strategies in the pharmacotherapy of diabetes and obesity.
Collapse
Affiliation(s)
- Gianluca Iacobellis
- Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, University of Miami, Miller School of Medicine, Miami, FL, USA.
| | - Sara Basilico
- Endocrinology Unit, Clinical Nutrition and Cardiovascular Prevention Service, IRCCS Policlinico San Donato, Milan, Italy
| | - Alexis Elias Malavazos
- Endocrinology Unit, Clinical Nutrition and Cardiovascular Prevention Service, IRCCS Policlinico San Donato, Milan, Italy
| |
Collapse
|
32
|
Modulation of Adipocyte Metabolism by Microbial Short-Chain Fatty Acids. Nutrients 2021; 13:nu13103666. [PMID: 34684670 PMCID: PMC8538331 DOI: 10.3390/nu13103666] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/17/2021] [Accepted: 10/17/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity and its complications—including type 2 diabetes, cardiovascular disease, and certain cancers—constitute a rising global epidemic that has imposed a substantial burden on health and healthcare systems over the years. It is becoming increasingly clear that there is a link between obesity and the gut microbiota. Gut dysbiosis, characterized as microbial imbalance, has been consistently associated with obesity in both humans and animal models, and can be reversed with weight loss. Emerging evidence has shown that microbial-derived metabolites such as short-chain fatty acids (SCFAs)—including acetate, propionate, and butyrate—provide benefits to the host by impacting organs beyond the gut, including adipose tissue. In this review, we summarize what is currently known regarding the specific mechanisms that link gut-microbial-derived SCFAs with adipose tissue metabolism, such as adipogenesis, lipolysis, and inflammation. In addition, we explore indirect mechanisms by which SCFAs can modulate adipose tissue metabolism, such as via perturbation of gut hormones, as well as signaling to the brain and the liver. Understanding how the modulation of gut microbial metabolites such as SCFAs can impact adipose tissue function could lead to novel therapeutic strategies for the prevention and treatment of obesity.
Collapse
|
33
|
Nauck MA, Quast DR, Wefers J, Pfeiffer AFH. The evolving story of incretins (GIP and GLP-1) in metabolic and cardiovascular disease: A pathophysiological update. Diabetes Obes Metab 2021; 23 Suppl 3:5-29. [PMID: 34310013 DOI: 10.1111/dom.14496] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 11/27/2022]
Abstract
The incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) have their main physiological role in augmenting insulin secretion after their nutrient-induced secretion from the gut. A functioning entero-insular (gut-endocrine pancreas) axis is essential for the maintenance of a normal glucose tolerance. This is exemplified by the incretin effect (greater insulin secretory response to oral as compared to "isoglycaemic" intravenous glucose administration due to the secretion and action of incretin hormones). GIP and GLP-1 have additive effects on insulin secretion. Local production of GIP and/or GLP-1 in islet α-cells (instead of enteroendocrine K and L cells) has been observed, and its significance is still unclear. GLP-1 suppresses, and GIP increases glucagon secretion, both in a glucose-dependent manner. GIP plays a greater physiological role as an incretin. In type 2-diabetic patients, the incretin effect is reduced despite more or less normal secretion of GIP and GLP-1. While insulinotropic effects of GLP-1 are only slightly impaired in type 2 diabetes, GIP has lost much of its acute insulinotropic activity in type 2 diabetes, for largely unknown reasons. Besides their role in glucose homoeostasis, the incretin hormones GIP and GLP-1 have additional biological functions: GLP-1 at pharmacological concentrations reduces appetite, food intake, and-in the long run-body weight, and a similar role is evolving for GIP, at least in animal studies. Human studies, however, do not confirm these findings. GIP, but not GLP-1 increases triglyceride storage in white adipose tissue not only through stimulating insulin secretion, but also by interacting with regional blood vessels and GIP receptors. GIP, and to a lesser degree GLP-1, play a role in bone remodelling. GLP-1, but not GIP slows gastric emptying, which reduces post-meal glycaemic increments. For both GIP and GLP-1, beneficial effects on cardiovascular complications and neurodegenerative central nervous system (CNS) disorders have been observed, pointing to therapeutic potential over and above improving diabetes complications. The recent finding that GIP/GLP-1 receptor co-agonists like tirzepatide have superior efficacy compared to selective GLP-1 receptor agonists with respect to glycaemic control as well as body weight has renewed interest in GIP, which previously was thought to be without any therapeutic potential. One focus of this research is into the long-term interaction of GIP and GLP-1 receptor signalling. A GLP-1 receptor antagonist (exendin [9-39]) and, more recently, a GIP receptor agonist (GIP [3-30] NH2 ) and, hopefully, longer-acting GIP receptor agonists for human use will be helpful tools to shed light on the open questions. A detailed knowledge of incretin physiology and pathophysiology will be a prerequisite for designing more effective incretin-based diabetes drugs.
Collapse
Affiliation(s)
- Michael A Nauck
- Diabetes Division, Katholisches Klinikum Bochum, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Daniel R Quast
- Diabetes Division, Katholisches Klinikum Bochum, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Jakob Wefers
- Diabetes Division, Katholisches Klinikum Bochum, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Andreas F H Pfeiffer
- Charité - Universitätsmedizin Berlin, Klinik für Endokrinologie, Stoffwechsel- und Ernährungsmedizin, Berlin, Germany
| |
Collapse
|
34
|
Mamontova ED, Michurina SS, Stafeev IS, Sorkina EL, Sklyanik IA, Koksharova EO, Menshikov MY, Shestakova MV, Parfyonova YV. Direct Effect of the Synthetic Analogue of Glucagon-Like Peptide Type 1, Liraglutide, on Mature Adipocytes Is Realized through Adenylate-Cyclase-Dependent Enhancing of Insulin Sensitivity. BIOCHEMISTRY (MOSCOW) 2021; 86:350-360. [PMID: 33838634 DOI: 10.1134/s000629792103010x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Incretin hormones analogues, including glucagon-like peptide type 1 (GLP-1), exhibit complex glucose-lowering, anorexigenic, and cardioprotective properties. Mechanisms of action of GLP-1 and its analogues are well known for pancreatic β-cells, hepatocytes, and other tissues. Nevertheless, local effects of GLP-1 and its analogues in adipose tissue remain unclear. In the present work effects of the GLP-1 synthetic analogue, liraglutide, on adipogenesis and insulin sensitivity of the 3T3-L1 adipocytes were examined. Enhancement of insulin sensitivity of mature adipocytes by the GLP-1 synthetic analogue liraglutide mediated by adenylate cyclase was demonstrated. The obtained results imply existence of the positive direct insulin-sensitizing effect of liraglutide on mature adipocytes.
Collapse
Affiliation(s)
- Elizaveta D Mamontova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.,Institute of Experimental Cardiology, National Medical Research Centre for Cardiology, Moscow, 121552, Russia.,Diabetes Institute, Endocrinology Research Centre, Moscow, 117036, Russia
| | - Svetlana S Michurina
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.,Institute of Experimental Cardiology, National Medical Research Centre for Cardiology, Moscow, 121552, Russia
| | - Iurii S Stafeev
- Institute of Experimental Cardiology, National Medical Research Centre for Cardiology, Moscow, 121552, Russia.
| | | | - Igor A Sklyanik
- Diabetes Institute, Endocrinology Research Centre, Moscow, 117036, Russia
| | | | - Mikhail Y Menshikov
- Institute of Experimental Cardiology, National Medical Research Centre for Cardiology, Moscow, 121552, Russia
| | | | - Yelena V Parfyonova
- Institute of Experimental Cardiology, National Medical Research Centre for Cardiology, Moscow, 121552, Russia.,Faculty of Basic Medicine, Lomonosov Moscow State University, Moscow, 119234, Russia
| |
Collapse
|
35
|
Chilelli NC, Faggian A, Favaretto F, Milan G, Compagnin C, Dassie F, Bettini S, Roverso M, Seraglia R, Lapolla A, Vettor R. In vitro chronic glycation induces AGEs accumulation reducing insulin-stimulated glucose uptake and increasing GLP1R in adipocytes. Am J Physiol Endocrinol Metab 2021; 320:E976-E988. [PMID: 33779307 DOI: 10.1152/ajpendo.00156.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intracellular AGEs accumulation increases RAGE and GLP1R and reduces glucose uptake in adipocytes.
Collapse
Affiliation(s)
| | - Alessia Faggian
- Department of Medicine, University of Padua, Internal Medicine 3, Padua, Italy
| | - Francesca Favaretto
- Department of Medicine, University of Padua, Internal Medicine 3, Padua, Italy
| | - Gabriella Milan
- Department of Medicine, University of Padua, Internal Medicine 3, Padua, Italy
| | - Chiara Compagnin
- Department of Medicine, University of Padua, Internal Medicine 3, Padua, Italy
| | - Francesca Dassie
- Department of Medicine, University of Padua, Internal Medicine 3, Padua, Italy
| | - Silvia Bettini
- Department of Medicine, University of Padua, Internal Medicine 3, Padua, Italy
| | - Marco Roverso
- Department of Chemical Sciences, University of Padua, Padua, Italy
| | - Roberta Seraglia
- Consiglio Nazionale delle Ricerche-Istituto di Chimica della Materia Condensata e di Tecnologie per l'Energia (CNR-ICMATE), Padua, Italy
| | - Annunziata Lapolla
- Department of Medicine, University of Padua, Diabetology and Dietetics, Padua, Italy
| | - Roberto Vettor
- Department of Medicine, University of Padua, Internal Medicine 3, Padua, Italy
| |
Collapse
|
36
|
Muzurović EM, Vujošević S, Mikhailidis DP. Can We Decrease Epicardial and Pericardial Fat in Patients With Diabetes? J Cardiovasc Pharmacol Ther 2021; 26:415-436. [PMID: 33844605 DOI: 10.1177/10742484211006997] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Diabetes mellitus (DM) is a chronic and complex metabolic disorder and also an important cause of cardiovascular (CV) disease (CVD). Patients with type 2 DM (T2DM) and obesity show a greater propensity for visceral fat deposition (and excessive fat deposits elsewhere) and the link between adiposity and CVD risk is greater for visceral than for subcutaneous (SC) adipose tissue (AT). There is growing evidence that epicardial AT (EAT) and pericardial AT (PAT) play a role in the development of DM-related atherosclerosis, atrial fibrillation (AF), myocardial dysfunction, and heart failure (HF). In this review, we will highlight the importance of PAT and EAT in patients with DM. We also consider therapeutic interventions that could have a beneficial effect in terms of reducing the amount of AT and thus CV risk. EAT is biologically active and a likely determinant of CV morbidity and mortality in patients with DM, given its anatomical characteristics and proinflammatory secretory pattern. Consequently, modification of EAT/PAT may become a therapeutic target to reduce the CV burden. In patients with DM, a low calorie diet, exercise, antidiabetics and statins may change the quantity of EAT, PAT or both, alter the secretory pattern of EAT, improve the metabolic profile, and reduce inflammation. However, well-designed studies are needed to clearly define CV benefits and a therapeutic approach to EAT/PAT in patients with DM.
Collapse
Affiliation(s)
- Emir M Muzurović
- Department of Internal Medicine, Endocrinology Section, 274294Clinical Centre of Montenegro, Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Snežana Vujošević
- Department of Internal Medicine, Endocrinology Section, 274294Clinical Centre of Montenegro, Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, 9687Royal Free Hospital Campus, University College London Medical School, University College London (UCL), Pond Street, London, UK.,Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
37
|
Model JFA, Lima MV, Ohlweiler R, Lopes Vogt É, Rocha DS, Souza SKD, Türck P, Araújo ASDR, Vinagre AS. Liraglutide improves lipid and carbohydrate metabolism of ovariectomized rats. Mol Cell Endocrinol 2021; 524:111158. [PMID: 33444670 DOI: 10.1016/j.mce.2021.111158] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 12/16/2022]
Abstract
Considering that post-menopausal women and ovariectomized rodents develop obesity associated with increased visceral fat, this study was developed to investigate if liraglutide, a glucagon-like peptide 1 (GLP1) analogue, could improve the metabolism of estrogen (E2) deficient females. Wistar rats were ovariectomized (OVX), and subdivided in four groups: sham saline, sham liraglutide, OVX saline, and OVX liraglutide. After sixty days, metabolic parameters of blood, heart, liver, brown (BAT) and white adipose tissue (WAT) visceral depots, and, heart oxidative homeostasis, were evaluated. Castration increased the animals' body weight, the relative weight of the WAT depots, hepatic triglycerides and cardiac glycogen content. Liraglutide treatment reversed these effects, decreased WAT depots weight and increased glucose oxidation and lipogenesis in BAT and WAT. In addition, liraglutide enhanced adrenalin (A) lipolytic effect. These results indicate that liraglutide may be a promising treatment to restore lipid homeostasis and prevent weight gain associated with E2 deficiency.
Collapse
Affiliation(s)
| | - Matheus Vieira Lima
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Renata Ohlweiler
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Éverton Lopes Vogt
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Débora Santos Rocha
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Samir Khal de Souza
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Patrick Türck
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | | | - Anapaula Sommer Vinagre
- Department of Physiology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil.
| |
Collapse
|
38
|
Zhang N, Liu C, Zhang Y, Xu D, Gui L, Lu Y, Zhang Q. Liraglutide regulates lipid metabolism via FGF21- LKB1- AMPK- ACC1 pathway in white adipose tissues and macrophage of type 2 diabetic mice. Biochem Biophys Res Commun 2021; 548:120-126. [PMID: 33640604 DOI: 10.1016/j.bbrc.2021.02.065] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 02/16/2021] [Indexed: 01/25/2023]
Abstract
Liraglutide (LRG), a glucagon-like peptide 1 analogue (GLP1A), could decrease body mass of type 2 diabetes (T2DM), but the exact molecular mechanism of LRG has not been elucidated. This study was performed to explore whether LRG regulated TG synthesis via secretion of FGF21 and modulating AMP-dependent protein kinase (AMPK) pathway in an autocrine mode. Two-month-old male C57BL/6 mice were fed high-fat diet (HFD) for 4 months followed by injection of 30 mg/kg streptozotocin (STZ) to induce state of T2DM. Then DM mice were given LRG (0.4 mg/kg/d) for 4 months. Body mass, serum lipids and FGF21 levels, related gene expression were analyzed. Lipopolysaccharide (LPS)-induced RAW264.7 cells were treated with palmitic acid and different concentrations of LRG. Then Exendin (9-39), siRNA targeted to liver kinase B1 (LKB1) and Compound C were used to confirm the signaling pathway. LRG decreased adipocyte size, increased secretion of FGF21, and promoted phosphorylation of LKB1, AMPK and Acetyl coenzyme A carboxylase 1 (ACC1) in white adipose tissue (WAT) of DM mice. LRG also increased phosphorylation of fibroblast growth factor receptor 3 (FGFR3), LKB1, AMPK and ACC1 via FGF21 secretion, which ultimately inhibited synthesis of TG in macrophage. In conclusion, FGF21 is induced to be expressed in macrophage by LRG, which then activates LKB1-AMPK-ACC1 pathway in an autocrine manner.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chao Liu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, Hefei, China
| | - Dongmei Xu
- Department of Biochemistry and Molecular Biology, China
| | - Li Gui
- The Comprehensive Laboratory, School of Basic Medical Science, Anhui Medical University, Hefei, China
| | - Yunxia Lu
- Department of Biochemistry and Molecular Biology, China; The Comprehensive Laboratory, School of Basic Medical Science, Anhui Medical University, Hefei, China.
| | - Qiu Zhang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
39
|
The methionine aminopeptidase 2 inhibitor, TNP-470, enhances the antidiabetic properties of sitagliptin in mice by upregulating xenin. Biochem Pharmacol 2020; 183:114355. [PMID: 33279496 DOI: 10.1016/j.bcp.2020.114355] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/12/2020] [Accepted: 12/01/2020] [Indexed: 12/24/2022]
Abstract
The therapeutic mechanism of action of methionine aminopeptidase 2 (MetAP2) inhibitors for obesity-diabetes has not yet been fully defined. Xenin, a K-cell derived peptide hormone, possesses an N-terminal Met amino acid residue. Thus, elevated xenin levels could represent a potential pharmacological mechanism of MetAP2 inhibitors, since long-acting xenin analogues have been shown to improve obesity-diabetes. The present study has assessed the ability of the MetAP2 inhibitor, TNP-470, to augment the antidiabetic utility of the incretin-enhancer drug, sitagliptin, in high fat fed (HFF) mice. TNP-470 (1 mg/kg) and sitagliptin (25 mg/kg) were administered once-daily alone, or in combination, to diabetic HFF mice (n = 10) for 18 days. Individual therapy with TNP-470 or sitagliptin resulted in numerous metabolic benefits including reduced blood glucose, increased circulating and pancreatic insulin and improved glucose tolerance, insulin sensitivity, pyruvate tolerance and overall pancreatic islet architecture. Further assessment of metabolic rate revealed that all treatments reduced respiratory exchange ratio and increased locomotor activity. All sitagliptin treated mice also exhibited increased energy expenditure. In addition, treatment with TNP-470 alone, or in combination with sitagliptin, reduced food intake and body weight, as well as elevating plasma and intestinal xenin. Importantly, combined sitagliptin and TNP-470 therapy was associated with further significant benefits beyond that observed by either treatment alone. This included more rapid restoration of normoglycaemia, superior glucose tolerance, increased circulating GIP concentrations and an enhanced pancreatic beta:alpha cell ratio. In conclusion, these data demonstrate that TNP-470 increases plasma and intestinal xenin levels, and augments the antidiabetic advantages of sitagliptin.
Collapse
|
40
|
Malbert CH, Chauvin A, Horowitz M, Jones KL. Pancreatic GLP-1r binding potential is reduced in insulin-resistant pigs. BMJ Open Diabetes Res Care 2020; 8:8/2/e001540. [PMID: 33132211 PMCID: PMC7607594 DOI: 10.1136/bmjdrc-2020-001540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/11/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION The insulinotropic capacity of exogenous glucagon like peptide-1 (GLP-1) is reduced in type 2 diabetes and the insulin-resistant obese. We have tested the hypothesis that this response is the consequence of a reduced pancreatic GLP-1 receptor (GLP-1r) density in insulin-resistant obese animals. RESEARCH DESIGN AND METHODS GLP-1r density was measured in lean and insulin-resistant adult miniature pigs after the administration of a 68Ga-labeled GLP-1r agonist. The effect of hyperinsulinemia on GLP-1r was assessed using sequential positron emission tomography (PET), both in the fasted state and during a clamp. The impact of tissue perfusion, which could account for changes in GLP-1r agonist uptake, was also investigated using 68Ga-DOTA imaging. RESULTS GLP-1r binding potential in the obese pancreas was reduced by 75% compared with lean animals. Similar reductions were evident for fat tissue, but not for the duodenum. In the lean group, induced hyperinsulinemia reduced pancreatic GLP-1r density to a level comparable with that of the obese group. The reduction in blood to tissue transfer of the GLP-1r ligand paralleled that of tissue perfusion estimated using 68Ga-DOTA. CONCLUSIONS These observations establish that a reduction in abdominal tissue perfusion and a lower GLP-1r density account for the diminished insulinotropic effect of GLP-1 agonists in type 2 diabetes.
Collapse
Affiliation(s)
| | - Alain Chauvin
- UEPR Unit, Department of Animal Physiology, INRAE, Saint-Gilles, France
| | - Michael Horowitz
- Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| | - Karen L Jones
- Center of Research Excellence in Translating Nutritional Science to Good Health, The University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
41
|
Diz-Chaves Y, Herrera-Pérez S, González-Matías LC, Lamas JA, Mallo F. Glucagon-Like Peptide-1 (GLP-1) in the Integration of Neural and Endocrine Responses to Stress. Nutrients 2020; 12:nu12113304. [PMID: 33126672 PMCID: PMC7692797 DOI: 10.3390/nu12113304] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/14/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022] Open
Abstract
Glucagon like-peptide 1 (GLP-1) within the brain is produced by a population of preproglucagon neurons located in the caudal nucleus of the solitary tract. These neurons project to the hypothalamus and another forebrain, hindbrain, and mesolimbic brain areas control the autonomic function, feeding, and the motivation to feed or regulate the stress response and the hypothalamic-pituitary-adrenal axis. GLP-1 receptor (GLP-1R) controls both food intake and feeding behavior (hunger-driven feeding, the hedonic value of food, and food motivation). The activation of GLP-1 receptors involves second messenger pathways and ionic events in the autonomic nervous system, which are very relevant to explain the essential central actions of GLP-1 as neuromodulator coordinating food intake in response to a physiological and stress-related stimulus to maintain homeostasis. Alterations in GLP-1 signaling associated with obesity or chronic stress induce the dysregulation of eating behavior. This review summarized the experimental shreds of evidence from studies using GLP-1R agonists to describe the neural and endocrine integration of stress responses and feeding behavior.
Collapse
Affiliation(s)
- Yolanda Diz-Chaves
- CINBIO, Universidade de Vigo, Grupo FB3A, Laboratorio de Endocrinología, 36310 Vigo, Spain;
- Correspondence: (Y.D.-C.); (F.M.); Tel.: +34-(986)-130226 (Y.D.-C.); +34-(986)-812393 (F.M.)
| | - Salvador Herrera-Pérez
- CINBIO, Universidade de Vigo, Grupo FB3B, Laboratorio de Neurociencia, 36310 Vigo, Spain; (S.H.-P.); (J.A.L.)
| | | | - José Antonio Lamas
- CINBIO, Universidade de Vigo, Grupo FB3B, Laboratorio de Neurociencia, 36310 Vigo, Spain; (S.H.-P.); (J.A.L.)
| | - Federico Mallo
- CINBIO, Universidade de Vigo, Grupo FB3A, Laboratorio de Endocrinología, 36310 Vigo, Spain;
- Correspondence: (Y.D.-C.); (F.M.); Tel.: +34-(986)-130226 (Y.D.-C.); +34-(986)-812393 (F.M.)
| |
Collapse
|
42
|
Ghanim H, Batra M, Green K, Abuaysheh S, Hejna J, Makdissi A, Borowski R, Kuhadiya ND, Chaudhuri A, Dandona P. Liraglutide treatment in overweight and obese patients with type 1 diabetes: A 26-week randomized controlled trial; mechanisms of weight loss. Diabetes Obes Metab 2020; 22:1742-1752. [PMID: 32424935 DOI: 10.1111/dom.14090] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 01/18/2023]
Abstract
AIM To investigate the effects of liraglutide treatment on glycaemic control and adipose tissue metabolism in overweight and obese people with type 1 diabetes (T1DM). RESEARCH DESIGN AND METHODS A total of 84 adult overweight and obese patients with T1DM, with no detectable C-peptide, were randomized (1:1) to either placebo or 1.8 mg/d liraglutide for 6 months. Blood samples were collected at 0, 12 and 26 weeks. Subcutaneous adipose tissue biopsies, a high-calorie high-fat meal challenge test, continuous glucose monitoring, dual-energy X-ray absorptiometry and MRI were performed before and at the end of treatment. RESULTS In all, 37 and 27 patients who received liraglutide and placebo, respectively, completed the study. Glycated haemoglobin fell by 0.41 ± 0.18% (4.5±1.4 mmol/mol) from baseline after liraglutide treatment (P = 0.001), and by 0.29 ± 0.19% (3.1±2.0 mmol/mol) compared to placebo (P = 0.1). There was no increase in hypoglycaemia, while the time spent in normal glycaemia increased (P = 0.015) and time spent in hyperglycaemia decreased (P = 0.019). Body weight fell significantly in the liraglutide group, mostly in the form of fat mass loss (including visceral fat), with no change in lean mass. Systolic blood pressure (SBP) also fell after liraglutide treatment. Liraglutide also caused a significant increase in the expression of adipose tissue triglyceride lipase, carnitine palmitoyl transferase-1, peroxisome proliferator-activated receptor (PPAR)α, PPARδ, uncoupling protein-2 and type 2 iodothyronine deiodinase in the adipose tissue. CONCLUSIONS Liraglutide improves glycaemia, reduces adiposity and SBP. Liraglutide also stimulates mechanisms involved with an increase in lipid oxidation and thermogenesis, while conserving lean body mass.
Collapse
Affiliation(s)
- Husam Ghanim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, 14221, New york, USA
| | - Manav Batra
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, 14221, New york, USA
| | - Kelly Green
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, 14221, New york, USA
| | - Sanaa Abuaysheh
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, 14221, New york, USA
| | - Jeanne Hejna
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, 14221, New york, USA
| | - Antione Makdissi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, 14221, New york, USA
| | - Robert Borowski
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, 14221, New york, USA
| | - Nitesh D Kuhadiya
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, 14221, New york, USA
| | - Ajay Chaudhuri
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, 14221, New york, USA
| | - Paresh Dandona
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, 14221, New york, USA
| |
Collapse
|
43
|
Onogi Y, Khalil AEMM, Ussar S. Identification and characterization of adipose surface epitopes. Biochem J 2020; 477:2509-2541. [PMID: 32648930 PMCID: PMC7360119 DOI: 10.1042/bcj20190462] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022]
Abstract
Adipose tissue is a central regulator of metabolism and an important pharmacological target to treat the metabolic consequences of obesity, such as insulin resistance and dyslipidemia. Among the various cellular compartments, the adipocyte cell surface is especially appealing as a drug target as it contains various proteins that when activated or inhibited promote adipocyte health, change its endocrine function and eventually maintain or restore whole-body insulin sensitivity. In addition, cell surface proteins are readily accessible by various drug classes. However, targeting individual cell surface proteins in adipocytes has been difficult due to important functions of these proteins outside adipose tissue, raising various safety concerns. Thus, one of the biggest challenges is the lack of adipose selective surface proteins and/or targeting reagents. Here, we discuss several receptor families with an important function in adipogenesis and mature adipocytes to highlight the complexity at the cell surface and illustrate the problems with identifying adipose selective proteins. We then discuss that, while no unique adipocyte surface protein might exist, how splicing, posttranslational modifications as well as protein/protein interactions can create enormous diversity at the cell surface that vastly expands the space of potentially unique epitopes and how these selective epitopes can be identified and targeted.
Collapse
Affiliation(s)
- Yasuhiro Onogi
- RG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Ahmed Elagamy Mohamed Mahmoud Khalil
- RG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Siegfried Ussar
- RG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Department of Medicine, Technische Universität München, Munich, Germany
| |
Collapse
|
44
|
Li Y, Liu X, Li G, Zhang P. Effect of liraglutide on epicardial adipose tissue thickness with echocardiography in patients with obese type 2 diabetes mellitus. Int J Diabetes Dev Ctries 2020. [DOI: 10.1007/s13410-020-00820-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
45
|
Iacobellis G, Villasante Fricke AC. Effects of Semaglutide Versus Dulaglutide on Epicardial Fat Thickness in Subjects with Type 2 Diabetes and Obesity. J Endocr Soc 2020; 4:bvz042. [PMID: 32190806 PMCID: PMC7069837 DOI: 10.1210/jendso/bvz042] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/11/2020] [Indexed: 12/29/2022] Open
Abstract
Background and Aims Epicardial adipose tissue (EAT), the visceral fat depot of the heart, is a modifiable cardio-metbolic risk factor and therapeutic target. Semaglutide and dulaglutide, glucagon-like peptide-1 (GLP-1) receptor agonists, are indicated for the treatment of type 2 diabetes mellitus (T2DM). GLP-1 receptor agonists have recently shown to reduce cardiovascular risk. Epicardial adipose tissue expresses GLP-1 receptors (GLP-1Rs). GLP-1 receptor agonist liraglutide is known to significantly decrease EAT thickness. However, the effects of GLP-1 receptor agonists semaglutide and dulaglutide on EAT thickness are unknown. Materials and Methods We performed a 12-week, controlled, parallel study in 80 subjects with T2DM and obesity. Patients received either semaglutide, up to 1 mg subcutaneous (sc) weekly, or dulaglutide, up to 1.5 mg sc weekly, as the standard of care in addition to their usual medication regimen. Twenty subjects with T2DM and obesity were started on metformin and a diet and served as the control group. Ultrasound-measured EAT thickness was measured at baseline and at the 12-week follow-up. Results Epicardial adipose tissue thickness significantly decreased in both semaglutide and dulaglutide groups (P < 0.001) after 12 weeks, accounting for a 20% reduction. There was no EAT reduction in the metformin group. Body mass index (BMI) and HbA1c improved in all groups without reaching statistical significance. Epicardial adipose tissue thickness reduction was significantly greater (P < 0.01) with the higher doses of semaglutide (1 mg) and dulaglutide (1.5 mg), respectively. Conclusion Weekly administration of either GLP-1 receptor agonists semaglutide or dulaglutide causes a rapid, substantial, and dose-dependent reduction in EAT thickness.
Collapse
Affiliation(s)
- Gianluca Iacobellis
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
| | - Alexandra C Villasante Fricke
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Miller School of Medicine, University of Miami, Miami, Florida
| |
Collapse
|
46
|
Ceddia RP, Collins S. A compendium of G-protein-coupled receptors and cyclic nucleotide regulation of adipose tissue metabolism and energy expenditure. Clin Sci (Lond) 2020; 134:473-512. [PMID: 32149342 PMCID: PMC9137350 DOI: 10.1042/cs20190579] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/17/2020] [Accepted: 02/24/2020] [Indexed: 12/15/2022]
Abstract
With the ever-increasing burden of obesity and Type 2 diabetes, it is generally acknowledged that there remains a need for developing new therapeutics. One potential mechanism to combat obesity is to raise energy expenditure via increasing the amount of uncoupled respiration from the mitochondria-rich brown and beige adipocytes. With the recent appreciation of thermogenic adipocytes in humans, much effort is being made to elucidate the signaling pathways that regulate the browning of adipose tissue. In this review, we focus on the ligand-receptor signaling pathways that influence the cyclic nucleotides, cAMP and cGMP, in adipocytes. We chose to focus on G-protein-coupled receptor (GPCR), guanylyl cyclase and phosphodiesterase regulation of adipocytes because they are the targets of a large proportion of all currently available therapeutics. Furthermore, there is a large overlap in their signaling pathways, as signaling events that raise cAMP or cGMP generally increase adipocyte lipolysis and cause changes that are commonly referred to as browning: increasing mitochondrial biogenesis, uncoupling protein 1 (UCP1) expression and respiration.
Collapse
Affiliation(s)
- Ryan P Ceddia
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| |
Collapse
|
47
|
Pereira MJ, Thombare K, Sarsenbayeva A, Kamble PG, Almby K, Lundqvist M, Eriksson JW. Direct effects of glucagon on glucose uptake and lipolysis in human adipocytes. Mol Cell Endocrinol 2020; 503:110696. [PMID: 31891768 DOI: 10.1016/j.mce.2019.110696] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 11/25/2019] [Accepted: 12/27/2019] [Indexed: 12/14/2022]
Abstract
We aim to investigate the expression of the glucagon receptor (GCGR) in human adipose tissue, and the impact of glucagon in glucose uptake and lipolysis in human adipocytes. GCGR gene expression in human subcutaneous and visceral adipose tissue was demonstrated, albeit at low levels and with an inter-individual variation. Furthermore, GCGR expression was not significantly different between subjects with T2D and matched controls, and we found no significant association with BMI. Glucagon only at a supra-physiological concentration (10-100 nM) significantly increased basal and insulin-stimulated glucose uptake by up to 1.5-fold. Also, glucagon (0.01 and 1 nM) dose-dependently increased basal and isoproterenol-stimulated lipolysis up to 3.7- and 1.7-fold, respectively, compared to control. In addition, glucagon did not change insulin sensitivity to stimulate glucose uptake or inhibit lipolysis. In conclusion, we show that the GCGR gene is expressed at low levels in human adipose tissue, and glucagon at high concentrations can increase both glucose uptake and lipolysis in human adipocytes. Taken together, our data suggest that glucagon at physiological levels has minor direct effects on the regulation of adipocyte metabolism, but does not antagonize the insulin effect to stimulate glucose uptake and inhibit lipolysis in human adipocytes.
Collapse
Affiliation(s)
- Maria J Pereira
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden.
| | - Ketan Thombare
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Assel Sarsenbayeva
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Prasad G Kamble
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Kristina Almby
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Martin Lundqvist
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Clinical Diabetes and Metabolism, Uppsala University, Uppsala, Sweden
| |
Collapse
|
48
|
Beaudry JL, Drucker DJ. Proglucagon-Derived Peptides, Glucose-Dependent Insulinotropic Polypeptide, and Dipeptidyl Peptidase-4-Mechanisms of Action in Adipose Tissue. Endocrinology 2020; 161:5648010. [PMID: 31782955 DOI: 10.1210/endocr/bqz029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 12/14/2022]
Abstract
Proglucagon-derived peptides (PGDPs) and related gut hormones exemplified by glucose-dependent insulinotropic polypeptide (GIP) regulate energy disposal and storage through actions on metabolically sensitive organs, including adipose tissue. The actions of glucagon, glucagon-like peptide (GLP)-1, GLP-2, GIP, and their rate-limiting enzyme dipeptidyl peptidase-4, include direct and indirect regulation of islet hormone secretion, food intake, body weight, all contributing to control of white and brown adipose tissue activity. Moreover, agents mimicking actions of these peptides are in use for the therapy of metabolic disorders with disordered energy homeostasis such as diabetes, obesity, and intestinal failure. Here we highlight current concepts and mechanisms for direct and indirect actions of these peptides on adipose tissue depots. The available data highlight the importance of indirect peptide actions for control of adipose tissue biology, consistent with the very low level of endogenous peptide receptor expression within white and brown adipose tissue depots. Finally, we discuss limitations and challenges for the interpretation of available experimental observations, coupled to identification of enduring concepts supported by more robust evidence.
Collapse
Affiliation(s)
- Jacqueline L Beaudry
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto ON, Canada
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto ON, Canada
| |
Collapse
|
49
|
Müller TD, Finan B, Bloom SR, D'Alessio D, Drucker DJ, Flatt PR, Fritsche A, Gribble F, Grill HJ, Habener JF, Holst JJ, Langhans W, Meier JJ, Nauck MA, Perez-Tilve D, Pocai A, Reimann F, Sandoval DA, Schwartz TW, Seeley RJ, Stemmer K, Tang-Christensen M, Woods SC, DiMarchi RD, Tschöp MH. Glucagon-like peptide 1 (GLP-1). Mol Metab 2019; 30:72-130. [PMID: 31767182 PMCID: PMC6812410 DOI: 10.1016/j.molmet.2019.09.010] [Citation(s) in RCA: 915] [Impact Index Per Article: 183.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/10/2019] [Accepted: 09/22/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The glucagon-like peptide-1 (GLP-1) is a multifaceted hormone with broad pharmacological potential. Among the numerous metabolic effects of GLP-1 are the glucose-dependent stimulation of insulin secretion, decrease of gastric emptying, inhibition of food intake, increase of natriuresis and diuresis, and modulation of rodent β-cell proliferation. GLP-1 also has cardio- and neuroprotective effects, decreases inflammation and apoptosis, and has implications for learning and memory, reward behavior, and palatability. Biochemically modified for enhanced potency and sustained action, GLP-1 receptor agonists are successfully in clinical use for the treatment of type-2 diabetes, and several GLP-1-based pharmacotherapies are in clinical evaluation for the treatment of obesity. SCOPE OF REVIEW In this review, we provide a detailed overview on the multifaceted nature of GLP-1 and its pharmacology and discuss its therapeutic implications on various diseases. MAJOR CONCLUSIONS Since its discovery, GLP-1 has emerged as a pleiotropic hormone with a myriad of metabolic functions that go well beyond its classical identification as an incretin hormone. The numerous beneficial effects of GLP-1 render this hormone an interesting candidate for the development of pharmacotherapies to treat obesity, diabetes, and neurodegenerative disorders.
Collapse
Affiliation(s)
- T D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, Tübingen, Germany.
| | - B Finan
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA
| | - S R Bloom
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - D D'Alessio
- Division of Endocrinology, Duke University Medical Center, Durham, NC, USA
| | - D J Drucker
- The Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, M5G1X5, Canada
| | - P R Flatt
- SAAD Centre for Pharmacy & Diabetes, Ulster University, Coleraine, Northern Ireland, UK
| | - A Fritsche
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, Department of Internal Medicine, University of Tübingen, Tübingen, Germany
| | - F Gribble
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - H J Grill
- Institute of Diabetes, Obesity and Metabolism, Department of Psychology, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - J F Habener
- Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Harvard University, Boston, MA, USA
| | - J J Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - W Langhans
- Physiology and Behavior Laboratory, ETH Zurich, Schwerzenbach, Switzerland
| | - J J Meier
- Diabetes Division, St Josef Hospital, Ruhr-University Bochum, Bochum, Germany
| | - M A Nauck
- Diabetes Center Bochum-Hattingen, St Josef Hospital (Ruhr-Universität Bochum), Bochum, Germany
| | - D Perez-Tilve
- Department of Internal Medicine, University of Cincinnati-College of Medicine, Cincinnati, OH, USA
| | - A Pocai
- Cardiovascular & ImmunoMetabolism, Janssen Research & Development, Welsh and McKean Roads, Spring House, PA, 19477, USA
| | - F Reimann
- Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Wellcome Trust-Medical Research Council, Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - D A Sandoval
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - T W Schwartz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, DL-2200, Copenhagen, Denmark; Department of Biomedical Sciences, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - R J Seeley
- Department of Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - K Stemmer
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - M Tang-Christensen
- Obesity Research, Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - S C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, Cincinnati, OH, USA
| | - R D DiMarchi
- Novo Nordisk Research Center Indianapolis, Indianapolis, IN, USA; Department of Chemistry, Indiana University, Bloomington, IN, USA
| | - M H Tschöp
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany; Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| |
Collapse
|
50
|
Epicardial adipose tissue GLP-1 receptor is associated with genes involved in fatty acid oxidation and white-to-brown fat differentiation: A target to modulate cardiovascular risk? Int J Cardiol 2019; 292:218-224. [DOI: 10.1016/j.ijcard.2019.04.039] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 04/04/2019] [Accepted: 04/10/2019] [Indexed: 12/14/2022]
|