51
|
Abstract
PURPOSE OF REVIEW In addition to their effects on glycemic control, two specific classes of relatively new anti-diabetic drugs, namely the sodium glucose co-transporter-2 inhibitors (SGLT2i) and glucagon-like peptide-1 receptor agonists (GLP-1RA) have demonstrated reduced rates of major adverse cardiovascular events (MACE) in subjects with type 2 diabetes (T2D) at high risk for cardiovascular disease (CVD). This review summarizes recent experimental results that inform putative molecular mechanisms underlying these benefits. RECENT FINDINGS SGLT2i and GLP-1RA exert cardiovascular effects by targeting in both common and distinctive ways (A) several mediators of macro- and microvascular pathophysiology: namely (A1) inflammation and atherogenesis, (A2) oxidative stress-induced endothelial dysfunction, (A3) vascular smooth muscle cell reactive oxygen species (ROS) production and proliferation, and (A4) thrombosis. These agents also exhibit (B) hemodynamic effects through modulation of (B1) natriuresis/diuresis and (B2) the renin-angiotensin-aldosterone system. This review highlights that while GLP-1RA exert direct effects on vascular (endothelial and smooth muscle) cells, the effects of SGLT2i appear to include the activation of signaling pathways that prevent adverse vascular remodeling. Both SGLT2i and GLP-1RA confer hemodynamic effects that counter adverse cardiac remodeling.
Collapse
Affiliation(s)
- Dorrin Zarrin Khat
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Mansoor Husain
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- Department of Medicine, University of Toronto, Toronto, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.
- Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, Canada.
- Ted Rogers Centre for Heart Research, University Health Network, Toronto, Canada.
- Peter Munk Cardiac Centre, University Health Network, Toronto, Canada.
| |
Collapse
|
52
|
Hussien NI, Sorour SM, El-Kerdasy HI, Abdelrahman BA. The glucagon-like peptide-1 receptor agonist Exendin-4, ameliorates contrast-induced nephropathy through suppression of oxidative stress, vascular dysfunction and apoptosis independent of glycaemia. Clin Exp Pharmacol Physiol 2018; 45:808-818. [PMID: 29637584 DOI: 10.1111/1440-1681.12944] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/22/2018] [Accepted: 04/03/2018] [Indexed: 12/22/2022]
Abstract
Contrast-induced nephropathy (CIN) is a leading cause of hospital-acquired acute kidney injury, particularly in diabetic patients. Previous studies have shown renoprotective effects of glucagon-like peptide-1 (GLP-1) signalling; however, its role in CIN remains unexplored. This study investigates the prophylactic effect of exendin-4, a GLP-1R agonist, against CIN in a rat model mimicking both healthy and diabetic conditions. Animals were randomly divided into 7 groups: a control sham group (n = 8), and 2 identical sets of 3 disease groups, one received exendin-4 before exposure to contrast medium (CM), while the other served as untreated control. The 3 disease groups represented diabetes (n = 8), CIN (n = 8), or diabetes and CIN combined (n = 8). Untreated groups showed deteriorating renal function as indicated by significantly higher levels of serum creatinine and blood urea nitrogen, malondialdehyde, and endothelin-1 and caspase-3 expression compared to the sham control group. This was accompanied by a significant decrease in tissue reserves of reduced glutathione, superoxide dismutase, nitrate and endothelin nitric oxide synthase as well as deteriorating renal histology. The CM-induced changes in diabetic rats indicate impaired renal function, oxidative stress, vascular dysfunction, and apoptosis, and were significance higher in intensity compared to non-diabetic rats. Pretreatment with exendin-4 ameliorated all the aforementioned CM-induced nephropathic effects independent of the glycemic state. To our knowledge, this is the first study describing the prophylactic renoprotective effects of exendin-4 against CIN. With the current pharmaceutical use of exendin-4 as a hypoglycaemic agent, the GLP-1R agonist becomes an interesting candidate for human clinical trials on CIN prevention.
Collapse
Affiliation(s)
- Noha I Hussien
- Department of Physiology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Safwa M Sorour
- Department of Pharmacology, Faculty of Medicine, Benha University, Benha, Egypt
| | - Hanan I El-Kerdasy
- Department of Anatomy, Faculty of Medicine, Benha University, Benha, Egypt
| | - Bakr A Abdelrahman
- Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
53
|
Yu M, Benjamin MM, Srinivasan S, Morin EE, Shishatskaya EI, Schwendeman SP, Schwendeman A. Battle of GLP-1 delivery technologies. Adv Drug Deliv Rev 2018; 130:113-130. [PMID: 30009885 PMCID: PMC6843995 DOI: 10.1016/j.addr.2018.07.009] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/26/2018] [Accepted: 07/09/2018] [Indexed: 12/25/2022]
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) belong to an important therapeutic class for treatment of type 2 diabetes. Six GLP-1 RAs, each utilizing a unique drug delivery strategy, are now approved by the Food and Drug Administration (FDA) and additional, novel GLP-1 RAs are still under development, making for a crowded marketplace and fierce competition among the manufacturers of these products. As rapid elimination is a major challenge for clinical application of GLP-1 RAs, various half-life extension strategies have been successfully employed including sequential modification, attachment of fatty-acid to peptide, fusion with human serum albumin, fusion with the fragment crystallizable (Fc) region of a monoclonal antibody, sustained drug delivery systems, and PEGylation. In this review, we discuss the scientific rationale of the various half-life extension strategies used for GLP-1 RA development. By analyzing and comparing different approved GLP-1 RAs and those in development, we focus on assessing how half-life extending strategies impact the pharmacokinetics, pharmacodynamics, safety, patient usability and ultimately, the commercial success of GLP-1 RA products. We also anticipate future GLP-1 RA development trends. Since similar drug delivery strategies are also applied for developing other therapeutic peptides, we expect this case study of GLP-1 RAs will provide generalizable concepts for the rational design of therapeutic peptides products with extended duration of action.
Collapse
Affiliation(s)
- Minzhi Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States of America
| | - Mason M Benjamin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States of America
| | | | - Emily E Morin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States of America
| | - Ekaterina I Shishatskaya
- Siberian Federal University, 79 Svobodnuy Ave, Krasnoyarsk 660041, Russian Federation; Institute of Biophysics SBRAS, 50 Akademgorodok, 660036, Russian Federation
| | - Steven P Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States of America; Biointerfaces Institute, NCRC, 2800 Plymouth Rd, Ann Arbor, MI 48109, United States of America; Department of Biomedical Engineering, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, United States of America.
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States of America; Biointerfaces Institute, NCRC, 2800 Plymouth Rd, Ann Arbor, MI 48109, United States of America.
| |
Collapse
|
54
|
Kapodistria K, Tsilibary EP, Kotsopoulou E, Moustardas P, Kitsiou P. Liraglutide, a human glucagon-like peptide-1 analogue, stimulates AKT-dependent survival signalling and inhibits pancreatic β-cell apoptosis. J Cell Mol Med 2018. [PMID: 29524296 PMCID: PMC5980190 DOI: 10.1111/jcmm.13259] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Liraglutide, a human long‐lasting GLP‐1 analogue, is currently regarded as a powerful treatment option for type 2 diabetes. Apart from glucoregulatory and insulinotropic actions, liraglutide increases β‐cell mass through stimulation of β‐cell proliferation and islet neogenesis, as well as inhibition of β‐cell apoptosis. However, the underline molecular mechanisms have not been fully characterized. In this study, we investigated the mechanism by which liraglutide preserves islet β‐cells in an animal model of overt diabetes, the obese db/db mice, and protects a mouse pancreatic β‐cell line (βTC‐6 cells) against apoptosis. Treatment of 12‐week‐old diabetic mice with liraglutide for 2 weeks had no appreciable effects on blood non‐fasting glucose concentration, islet insulin content and body weight. However, morphological and biochemical examination of diabetic mouse pancreatic islets demonstrated that liraglutide restores islet size, reduces islet β‐cell apoptosis and improves nephrin expression, a protein involved in β‐cell survival signalling. Our results indicated that liraglutide protects βTC‐6 cells from serum withdrawal‐induced apoptosis through inhibition of caspase‐3 activation. The molecular mechanism of the anti‐apoptotic action of liraglutide in βTC‐6‐cells comprises stimulation of PI3‐kinase‐dependent AKT phosphorylation leading to the phosphorylation, hence inactivation of the pro‐apoptotic protein BAD and inhibition of FoxO1 transcription factor. In conclusion, we provided evidence that the GLP‐1 analogue liraglutide exerts important beneficial effects on pancreatic islet architecture and β‐cell survival by protecting cells against apoptosis. These findings extend our understanding of the actions of liraglutide and further support the use of GLP‐1R agonists in the treatment of patients with type 2 diabetes.
Collapse
Affiliation(s)
- Katerina Kapodistria
- Institute of Biosciences and Applications, National Centre for Scientific Research, N.C.S.R. "Demokritos", Terma Patriarchou Grigoriou & Neapoleos, Attiki, Greece
| | - Effie-Photini Tsilibary
- Institute of Biosciences and Applications, National Centre for Scientific Research, N.C.S.R. "Demokritos", Terma Patriarchou Grigoriou & Neapoleos, Attiki, Greece
| | - Eleni Kotsopoulou
- Institute of Biosciences and Applications, National Centre for Scientific Research, N.C.S.R. "Demokritos", Terma Patriarchou Grigoriou & Neapoleos, Attiki, Greece
| | - Petros Moustardas
- Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens (BRFAA), Athens, Greece
| | - Paraskevi Kitsiou
- Institute of Biosciences and Applications, National Centre for Scientific Research, N.C.S.R. "Demokritos", Terma Patriarchou Grigoriou & Neapoleos, Attiki, Greece
| |
Collapse
|
55
|
Jo S, Chen J, Xu G, Grayson TB, Thielen LA, Shalev A. miR-204 Controls Glucagon-Like Peptide 1 Receptor Expression and Agonist Function. Diabetes 2018; 67:256-264. [PMID: 29101219 PMCID: PMC5780066 DOI: 10.2337/db17-0506] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 10/30/2017] [Indexed: 12/18/2022]
Abstract
Glucagon-like peptide 1 receptor (GLP1R) agonists are widely used to treat diabetes. However, their function is dependent on adequate GLP1R expression, which is downregulated in diabetes. GLP1R is highly expressed on pancreatic β-cells, and activation by endogenous incretin or GLP1R agonists increases cAMP generation, which stimulates glucose-induced β-cell insulin secretion and helps maintain glucose homeostasis. We now have discovered that the highly β-cell-enriched microRNA, miR-204, directly targets the 3' UTR of GLP1R and thereby downregulates its expression in the β-cell-derived rat INS-1 cell line and primary mouse and human islets. Furthermore, in vivo deletion of miR-204 promoted islet GLP1R expression and enhanced responsiveness to GLP1R agonists, resulting in improved glucose tolerance, cAMP production, and insulin secretion as well as protection against diabetes. Since we recently identified thioredoxin-interacting protein (TXNIP) as an upstream regulator of miR-204, we also assessed whether in vivo deletion of TXNIP could mimic that of miR-204. Indeed, it also enhanced islet GLP1R expression and GLP1R agonist-induced insulin secretion and glucose tolerance. Thus, the present studies show for the first time that GLP1R is under the control of a microRNA, miR-204, and uncover a previously unappreciated link between TXNIP and incretin action.
Collapse
Affiliation(s)
- SeongHo Jo
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL
| | - Junqin Chen
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL
| | - Guanlan Xu
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL
| | - Truman B Grayson
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL
| | - Lance A Thielen
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL
| | - Anath Shalev
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
56
|
Drucker DJ, Habener JF, Holst JJ. Discovery, characterization, and clinical development of the glucagon-like peptides. J Clin Invest 2017; 127:4217-4227. [PMID: 29202475 DOI: 10.1172/jci97233] [Citation(s) in RCA: 243] [Impact Index Per Article: 34.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The discovery, characterization, and clinical development of glucagon-like-peptide-1 (GLP-1) spans more than 30 years and includes contributions from multiple investigators, science recognized by the 2017 Harrington Award Prize for Innovation in Medicine. Herein, we provide perspectives on the historical events and key experimental findings establishing the biology of GLP-1 as an insulin-stimulating glucoregulatory hormone. Important attributes of GLP-1 action and enteroendocrine science are reviewed, with emphasis on mechanistic advances and clinical proof-of-concept studies. The discovery that GLP-2 promotes mucosal growth in the intestine is described, and key findings from both preclinical studies and the GLP-2 clinical development program for short bowel syndrome (SBS) are reviewed. Finally, we summarize recent progress in GLP biology, highlighting emerging concepts and scientific insights with translational relevance.
Collapse
Affiliation(s)
- Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Joel F Habener
- Laboratory of Molecular Endocrinology, Massachusetts General Hospital, Harvard University, Boston, Massachusetts, USA
| | - Jens Juul Holst
- Novo Nordisk Foundation Center for Basic Metabolic Research, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
57
|
Chien CT, Fan SC, Lin SC, Kuo CC, Yang CH, Yu TY, Lee SP, Cheng DY, Li PC. Glucagon-like peptide-1 receptor agonist activation ameliorates venous thrombosis-induced arteriovenous fistula failure in chronic kidneyd isease. Thromb Haemost 2017; 112:1051-64. [DOI: 10.1160/th14-03-0258] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 05/08/2014] [Indexed: 12/18/2022]
Abstract
SummaryHigh shear stress that develops in the arteriovenous fistula of chronic kidney diseases (CKD) may increase H2O2 and thromboxane A2 (TXA2) release, thereby exacerbating endothelial dysfunction, thrombosis, and neointimal hyperplasia. We investigated whether glucagon-like peptide-1 receptor agonist/exendin-4, a potentially cardiovascular protective agent, could improve TXA2-induced arteriovenous fistula injury in CKD. TXA2 administration to H2O2-exposed human umbilical vein endothelial cells increased apoptosis, senescence, and detachment; these phenotypes were associated with the downregulation of phosphorylated endothelial nitric oxide synthase/heme oxygenase-1 (eNOS/HO-1) signalling. Exendin-4 reduced H2O2/TXA2-induced endothelial injury via inhibition of apoptosis-related mechanisms and restoration of phosphorylated eNOS/HO-1 signalling. Male Wistar rats subjected to right common carotid artery-external jugular vein anastomosis were treated with exendin-4 via cervical implant osmotic pumps for 16–42 days. High shear stress induced by the arteriovenous fistula significantly increased venous haemodynamics, blood and tissue H2O2 and TXB2 levels, macrophage/monocyte infiltration, fibrosis, proliferation, and adhesion molecule-1 expression. Apoptosis was also increased due to NADPH oxidase gp91 activation and mitochondrial Bax translocation in the proximal end of the jugular vein of CKD rats. Exendin-4-treatment of rats with CKD led to the restoration of normal endothelial morphology and correction of arteriovenous fistula function. Exendin-4 treatment or thromboxane synthase gene deletion in CKD mice markedly reduced ADP-stimulated platelet adhesion to venous endothelium, and prevented venous occlusion in FeCl3-injured vessels by upregulation of HO-1. Together, these data reveal that the use of glucagon-like peptide-1 receptor agonists is an effective strategy for treatment of CKD-induced arteriovenous fistula failure.
Collapse
|
58
|
Huang XL, He Y, Ji LL, Wang KY, Wang YL, Chen DF, Geng Y, OuYang P, Lai WM. Hepatoprotective potential of isoquercitrin against type 2 diabetes-induced hepatic injury in rats. Oncotarget 2017; 8:101545-101559. [PMID: 29254185 PMCID: PMC5731895 DOI: 10.18632/oncotarget.21074] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 06/04/2017] [Indexed: 12/16/2022] Open
Abstract
Non-alcoholic fatty liver disease is a main complication of type 2 diabetes. Isoquercitrin are employed for antidiabetic therapies, but the effects on liver function and the hepatocytes are unclear. The aim of this study was to investigate the effects of isoquercitrin on the T2DM-induced hepatic injury in rats. Isoquercitrin (10 mg/kg/d, 30 mg/kg/d), sitagliptin phosphate (10 mg/kg/d) was given orally for 21 days. The administration of isoquercitrin at 10 mg/kg/d and 30 mg/kg/d showed a dose dependent. Compare to the negative control (treated with saline), rats medicated with isoquercitrin (30 mg/kg/d) and sitagliptin phosphate (10 mg/kg/d) improved the clinical symptoms, FBG and glucose tolerance, reduced serum ALT, AST and IR, but increased TP, Alb, SOD, GSH, MDA, HDL-C, INS and GLP-1. On histology, Rats of these to groups presented nearly normal liver tissue and Langerhans, degeneration, necrosis and apoptosis were markedly reduced. Instead, hepatocytes showed regenerate. These two groups also showed significant increase in mRNA expression of PKA, AKT, PKCa, InsR and PI3K, and a decrease in DPP-IV mRNA level. These results indicated that treatment with isoquercitrin protects against hepatic injury by T2DM.
Collapse
Affiliation(s)
- Xiao-Li Huang
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Wenjiang, Sichuan, P. R. China
| | - Yang He
- Department of Basic Veterinary, College of Veterinary Medicine, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, P.R. China
| | - Li-Li Ji
- Meat Processing Key Laboratory of Sichuan Province, Chengdu University, Chengdu, Sichuan, P.R. China
| | - Kai-Yu Wang
- Department of Basic Veterinary, College of Veterinary Medicine, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, P.R. China.,Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Wenjiang, Sichuan, P. R. China
| | - Yi-Li Wang
- Sichuan Tiantian Biotechnology Application Ltd, Chengdu, Sichuan, P.R. China
| | - De-Fang Chen
- Department of Aquaculture, College of Animal Science & Technology, Sichuan Agricultural University, Wenjiang, Sichuan, P. R. China
| | - Yi Geng
- Department of Basic Veterinary, College of Veterinary Medicine, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, P.R. China
| | - Ping OuYang
- Department of Basic Veterinary, College of Veterinary Medicine, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, P.R. China
| | - Wei-Min Lai
- Department of Basic Veterinary, College of Veterinary Medicine, Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Sichuan, P.R. China
| |
Collapse
|
59
|
Abbas NAT, Kabil SL. Liraglutide ameliorates cardiotoxicity induced by doxorubicin in rats through the Akt/GSK-3β signaling pathway. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:1145-1153. [PMID: 28780599 DOI: 10.1007/s00210-017-1414-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/27/2017] [Indexed: 12/21/2022]
Abstract
Doxorubicin (Dox)-induced cardiotoxicity constitutes the major adverse effect that limited its use. We investigated the possible protective effects of liraglutide on Dox-induced cardiotoxicity in rats. Rats were divided into the following groups: control group rats received normal saline [1 ml/kg, intraperitoneal (i.p.)]; doxorubicin group rats received doxorubicin (1.25 mg/kg, i.p.), four times per week for 4 weeks; and liraglutide group rats received doxorubicin (1.25 mg/kg, i.p.) four times per week for 4 weeks then received liraglutide (100 μg/kg, i.p) daily for 4 weeks. At the end of the study, animals were sacrificed and serum creatine kinase-MB (CK-MB) and troponin I levels were determined. Malondialdehyde (MDA), superoxide dismutase (SOD), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and caspase-3 levels of the heart were determined. Cardiac AMPK, phosphorylated-Akt, tissue growth factor-β1 (TGF-β1), and GSK3-β levels of the heart were determined. Hematoxylin and eosin (H&E) stained sections form the heart were examined as well as immunohistochemical sections for detection of Bcl-2 expression. Dox treatment increased serum level of troponin I and CK-MB while decreased SOD activity, decreased AMPK, and p-Akt cardiac levels with increased in MDA, IL-6, TNF-α,GSK-3b, TGFB1, and caspase-3 levels in the heart with inflammation and necrosis in cardiac histopathology with decreased Bcl-2. Treatment with liraglutide decreased troponin I and CK-MB while increased SOD activity, AMPK, p-Akt with decrements in MDA, IL-6, TNF-α, GSK-3β, TGF-β1, and caspase-3 levels with attenuation of inflammation and necrosis while increased Bcl-2 expression. Liraglutide may thus represent a new clinical tool for the treatment of Dox-induced cardiotoxicity.
Collapse
Affiliation(s)
- Noha A T Abbas
- Department of Pharmacology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Soad L Kabil
- Department of Pharmacology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
60
|
Caporarello N, Parrino C, Trischitta V, Frittitta L. Insulin receptor signaling and glucagon-like peptide 1 effects on pancreatic beta cells. PLoS One 2017; 12:e0181190. [PMID: 28767692 PMCID: PMC5540605 DOI: 10.1371/journal.pone.0181190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 06/26/2017] [Indexed: 12/03/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is a potent gluco-incretin hormone, which plays a central role on pancreatic beta cell proliferation, survival and insulin secreting activity and whose analogs are used for treating hyperglycemia in type 2 diabetes mellitus. Notably, abnormal insulin signaling affects all the above-mentioned aspects on pancreatic beta cells. The aim of our study was to investigate whether the protective effects of GLP1-1 on beta cells are affected by altered insulin receptor signaling. To this end, several effects of GLP-1 were studied in INS-1E rat beta cells transfected either with an inhibitor of insulin receptor function (i.e., the Ectonucleotide Pyrophosphatase Phosphodiesterase 1, ENPP1), or with insulin receptor small interfering RNA, as well as in control cells. Crucial experiments were carried out also in a second cell line, namely the βTC-1 mouse beta cells. Our data indicate that in insulin secreting beta cells in which either ENPP1 was up-regulated or insulin receptor was down-regulated, GLP-1 positive effects on several pancreatic beta cell activities, including glucose-induced insulin secretion, cell proliferation and cell survival, were strongly reduced. Further studies are needed to understand whether such a scenario occurs also in humans and, if so, if it plays a role of clinical relevance in diabetic patients with poor responsiveness to GLP-1 related treatments.
Collapse
Affiliation(s)
- Nunzia Caporarello
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Cristina Parrino
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Vincenzo Trischitta
- IRCCS Casa Sollievo della Sofferenza, Research Unit of Diabetes and Endocrine Diseases, San Giovanni Rotondo, Italy
- Department of Experimental Medicine “Sapienza” University, Rome, Italy
| | - Lucia Frittitta
- Endocrine Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
- * E-mail:
| |
Collapse
|
61
|
Guida C, Stephen S, Guitton R, Ramracheya RD. The Role of PYY in Pancreatic Islet Physiology and Surgical Control of Diabetes. Trends Endocrinol Metab 2017; 28:626-636. [PMID: 28533020 DOI: 10.1016/j.tem.2017.04.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/27/2017] [Indexed: 12/30/2022]
Abstract
Bariatric surgery in obese individuals leads to rapid and lasting remission of type 2 diabetes (T2D). This phenomenon occurs independently of weight loss possibly via a combination of factors. The incretin hormone GLP-1 has so far been recognised as a critical factor. However, recent data have indicated that elevation in another gut hormone, peptide tyrosine tyrosine (PYY), may drive the beneficial effects of surgery. Here we discuss recent findings on PYY-mediated control of glucose homeostasis and its role in diabetes, in the context of what is known for GLP-1. Identification of factors that increase the expression of PYY following bariatric surgery and elucidation of its role in diabetes reversal may have clinical relevance as a nonsurgical therapy for T2D.
Collapse
Affiliation(s)
- Claudia Guida
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, OX37LJ Oxford, UK
| | - Sam Stephen
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, OX37LJ Oxford, UK
| | - Romain Guitton
- Angers University Hospital, 18 Avenue du Général Patton, 49000 Angers, France
| | - Reshma D Ramracheya
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, OX37LJ Oxford, UK.
| |
Collapse
|
62
|
Abstract
AIM Glucagon-like peptide-1 (GLP-1) receptor agonists are a kind of very popular antidiabetes drugs. They promote cell proliferation and survival through activation of signaling pathways in human islet cells involving phosphate idylinositol 3 kinase (PI3K) and extracellular regulated kinases 1 and 2 (ERK1/2), which are frequently activated in human colon cancer cells. Then, it is possible that taking GLP-1 receptor (GLP-1R) agonists persistently would induce proliferation of β cells as well as colon cancer cells. So, clarifying the effects and mechanisms of GLP-1R agonists on colon cancer cells has important clinical implications. MATERIALS AND METHODS We investigated GLP-1R expression in human colon cancer tissue samples with immunohistochemisty analysis and explored the effects of exendin-4, a GLP-1 receptor agonist, on colon cancer cells in vitro and in vivo. RESULTS The results showed lack of GLP-1R expression in both human colon cancer tissues and colon cancer cell lines. Exendin-4 did not enhance the proliferation and migration of colon cancer cell lines in vitro, and nor did it inhibit apoptosis induced by cytotoxic agents such as 5-fluorouracil (5-FU) or irinotecan. In addition, exendin-4 did not promote the propagation of colon cancer cells in vivo. CONCLUSION Our study suggests that GLP-1R agonists do not modify the growth or survival of human colon cancer cells and may be safe for diabetic patients with colon cancer.
Collapse
Affiliation(s)
- He Wenjing
- a Department of Endocrinology , The First Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
- b Institute of Urology, The First Affiliated Hospital of Nanchang University , Nanchang , China
| | - Yu Shuang
- a Department of Endocrinology , The First Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| | - Li Weisong
- c Department of Pathology , The First Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| | - Xiao Haipeng
- a Department of Endocrinology , The First Affiliated Hospital of Sun Yat-sen University , Guangzhou , China
| |
Collapse
|
63
|
Muscogiuri G, Balercia G, Barrea L, Cignarelli A, Giorgino F, Holst JJ, Laudisio D, Orio F, Tirabassi G, Colao A. Gut: A key player in the pathogenesis of type 2 diabetes? Crit Rev Food Sci Nutr 2017; 58:1294-1309. [PMID: 27892685 DOI: 10.1080/10408398.2016.1252712] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The gut regulates glucose and energy homeostasis; thus, the presence of ingested nutrients into the gut activates sensing mechanisms that affect both glucose homeostasis and regulate food intake. Increasing evidence suggest that gut may also play a key role in the pathogenesis of type 2 diabetes which may be related to both the intestinal microbiological profile and patterns of gut hormones secretion. Intestinal microbiota includes trillions of microorganisms but its composition and function may be adversely affected in type 2 diabetes. The intestinal microbiota may be responsible of the secretion of molecules that may impair insulin secretion/action. At the same time, intestinal milieu regulates the secretion of hormones such as GLP-1, GIP, ghrelin, gastrin, somatostatin, CCK, serotonin, peptide YY, GLP-2, all of which importantly influence metabolism in general and in particular glucose metabolism. Thus, the aim of this paper is to review the current evidence on the role of the gut in the pathogenesis of type 2 diabetes, taking into account both hormonal and microbiological aspects.
Collapse
Affiliation(s)
| | - Giancarlo Balercia
- b Division of Endocrinology, Department of Clinical and Molecular Sciences , Umberto I Hospital, Polytechnic University of Marche , Ancona , Italy
| | | | - Angelo Cignarelli
- c Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases , University of Bari Aldo Moro , Bari , Italy
| | - Francesco Giorgino
- c Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases , University of Bari Aldo Moro , Bari , Italy
| | - Jens J Holst
- d NNF Center for Basic Metabolic Research and Department of Biomedical Sciences , Panum Institute, University of Copenhagen, Copenhagen , Denmark
| | | | - Francesco Orio
- e Endocrinology, Department of Sports Science and Wellness , "Parthenope" University Naples , Naples , Italy
| | - Giacomo Tirabassi
- b Division of Endocrinology, Department of Clinical and Molecular Sciences , Umberto I Hospital, Polytechnic University of Marche , Ancona , Italy
| | - Annamaria Colao
- f Department of Clinical Medicine and Surgery , "Federico II" University of Naples , Naples , Italy
| |
Collapse
|
64
|
Barakat G, Moustafa ME, Khalifeh I, Hodroj MH, Bikhazi A, Rizk S. Effects of exendin-4 and selenium on the expression of GLP-1R, IRS-1, and preproinsulin in the pancreas of diabetic rats. J Physiol Biochem 2017; 73:387-394. [PMID: 28589533 DOI: 10.1007/s13105-017-0565-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 05/23/2017] [Indexed: 03/23/2024]
Abstract
The mechanisms by which exendin-4 and selenium exert their antidiabetic actions are still unclear. Here, we investigated the effects of exendin-4 or selenium administration on the expression of glucagon-like peptide-1 receptor (GLP-1R), insulin receptor substrate-1 (IRS-1), and preproinsulin in the pancreas of diabetic rats. Diabetes was induced by streptozotocin administration. Diabetic rats were injected intraperitoneally with 0.03 μg exendin-4/kg body weight/daily or treated with 5 ppm selenium in drinking water for a period of 4 weeks. GLP-1R and IRS-1 levels were decreased while the level of preproinsulin messenger RNA (mRNA) was increased in the pancreas of diabetic untreated rats, as compared to that in control rats. Treatment of diabetic rats with exendin-4 increased protein and mRNA levels of GLP-1R, and IRS-1, and the mRNA level of preproinsulin in the pancreas, as compared to their levels in diabetic untreated rats. Selenium treatment of diabetic rats increased the pancreatic mRNA levels of GLP-1R, IRS-1, and preproinsulin. Exendin-4 or selenium treatment of diabetic rats also increased the numbers of pancreatic islets and GLP-1R molecules in the pancreas. Therefore, exendin-4 and selenium may exert their antidiabetic effects by increasing GLP-1R, IRS-1, and preproinsulin expression in the pancreas and by increasing the number of pancreatic islets.
Collapse
Affiliation(s)
- Ghinwa Barakat
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut, Lebanon
| | - Mohamed E Moustafa
- Department of Biological Sciences, Faculty of Science, Beirut Arab University, Beirut, Lebanon.,Department of Biochemistry, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Ibrahim Khalifeh
- Department of Pathology Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohammad H Hodroj
- Department of Natural Sciences, Lebanese American University, Beirut, 1102-2801, Lebanon
| | - Anwar Bikhazi
- Department of Physiology Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Sandra Rizk
- Department of Natural Sciences, Lebanese American University, Beirut, 1102-2801, Lebanon.
| |
Collapse
|
65
|
Panaro BL, Flock GB, Campbell JE, Beaudry JL, Cao X, Drucker DJ. β-Cell Inactivation of Gpr119 Unmasks Incretin Dependence of GPR119-Mediated Glucoregulation. Diabetes 2017; 66:1626-1635. [PMID: 28254842 PMCID: PMC5860191 DOI: 10.2337/db17-0017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 02/28/2017] [Indexed: 01/06/2023]
Abstract
GPR119 was originally identified as an orphan β-cell receptor; however, subsequent studies demonstrated that GPR119 also regulates β-cell function indirectly through incretin hormone secretion. We assessed the importance of GPR119 for β-cell function in Gpr119-/- mice and in newly generated Gpr119βcell-/- mice. Gpr119-/- mice displayed normal body weight and glucose tolerance on a regular chow (RC) diet. After high-fat feeding, Gpr119-/- mice exhibited reduced fat mass, decreased levels of circulating adipokines, improved insulin sensitivity, and better glucose tolerance. Unexpectedly, oral and intraperitoneal glucose tolerance and the insulin response to glycemic challenge were not perturbed in Gpr119βcell-/- mice on RC and high-fat diets. Moreover, islets from Gpr119-/- and Gpr119βcell-/- mice exhibited normal insulin responses to glucose and β-cell secretagogues. Furthermore, the selective GPR119 agonist AR231453 failed to directly enhance insulin secretion from perifused islets. In contrast, AR231453 increased plasma glucagon-like peptide 1 (GLP-1) and insulin levels and improved glucose tolerance in wild-type and Gpr119βcell-/- mice. These findings demonstrate that β-cell GPR119 expression is dispensable for the physiological control of insulin secretion and the pharmacological response to GPR119 agonism, findings that may inform the lack of robust efficacy in clinical programs assessing GPR119 agonists for the therapy of type 2 diabetes.
Collapse
Affiliation(s)
- Brandon L Panaro
- Lunenfeld-Tanenbaum Research Institute, Department of Medicine, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Grace B Flock
- Lunenfeld-Tanenbaum Research Institute, Department of Medicine, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Jonathan E Campbell
- Lunenfeld-Tanenbaum Research Institute, Department of Medicine, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Jacqueline L Beaudry
- Lunenfeld-Tanenbaum Research Institute, Department of Medicine, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Xiemin Cao
- Lunenfeld-Tanenbaum Research Institute, Department of Medicine, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Daniel J Drucker
- Lunenfeld-Tanenbaum Research Institute, Department of Medicine, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
66
|
Keshava HB, Mowla A, Heinberg LJ, Schauer PR, Brethauer SA, Aminian A. Bariatric surgery may reduce the risk of Alzheimer's diseases through GLP-1 mediated neuroprotective effects. Med Hypotheses 2017; 104:4-9. [PMID: 28673587 DOI: 10.1016/j.mehy.2017.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 04/26/2017] [Accepted: 05/01/2017] [Indexed: 12/25/2022]
Abstract
Obesity and diabetes are associated with deficits in multiple neurocognitive domains and increased risk for dementia. Over the last two decades, there has been a significant increase in bariatric and metabolic surgery worldwide, driven by rising intertwined pandemics of obesity and diabetes, along with improvement in surgical techniques. Patients undergoing bariatric surgery achieve a significant decrease in their excess weight and a multitude of sequela associated with obesity, diabetes, and metabolic syndrome. Glucagon-like peptide 1 (GLP-1) is an intestinal peptide that has been implicated as one of the weight loss-independent mechanisms in how bariatric surgery affects type 2 diabetes. GLP-1 improves insulin secretion, inhibits apoptosis and induce pancreatic islet neogenesis, promotes satiety, and can regulate heart rate and blood pressure. Moreover, numerous studies have demonstrated potential neuroprotective and neurotrophic effects of GLP-1. Increased GLP-1 activity has been shown to increase cortical activity, promote neuronal growth, and inhibit neuronal degeneration. Specifically, in experimental studies on Alzheimer's disease, GLP-1 decreases amyloid deposition and neurofibrillary tangles. Furthermore, recent studies have also suggested that GLP-1 based therapies, new class of antidiabetic drugs, have favorable effects on neurodegenerative disorders such as Alzheimer's disease. We present a hypothesis that bariatric surgery can help delay or even prevent the onset of Alzheimer's disease in long-term by increasing the levels of GLP-1. This hypothesis has a potential for many studies from basic science projects to large population studies to fully understand the neurological and cognitive consequences of bariatric surgery and associated rise in GLP-1.
Collapse
Affiliation(s)
- Hari B Keshava
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, OH, United States
| | - Ashkan Mowla
- Department of Neurology, Gates Vascular Institute, State University of New York (SUNY) at Buffalo, Buffalo, NY, United States
| | - Leslie J Heinberg
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, OH, United States
| | - Philip R Schauer
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, OH, United States
| | - Stacy A Brethauer
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, OH, United States
| | - Ali Aminian
- Bariatric and Metabolic Institute, Department of General Surgery, Cleveland Clinic, Cleveland, OH, United States.
| |
Collapse
|
67
|
Siba IP, Bortolanza M, Frazão Vital MAB, Andreatini R, da Cunha JM, Del Bel EA, Zanoveli JM. Fish oil prevents rodent anxious states comorbid with diabetes: A putative involvement of nitric oxide modulation. Behav Brain Res 2017; 326:173-186. [DOI: 10.1016/j.bbr.2017.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/02/2017] [Accepted: 03/04/2017] [Indexed: 01/17/2023]
|
68
|
Tang W, Yuan Q, Xu B, Osei K, Wang J. Exenatide substantially improves proinsulin conversion and cell survival that augment Ins2 +/Akita beta cell function. Mol Cell Endocrinol 2017; 439:297-307. [PMID: 27658750 DOI: 10.1016/j.mce.2016.09.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 09/16/2016] [Accepted: 09/16/2016] [Indexed: 02/06/2023]
Abstract
Proinsulin folding imperfections cause extensive beta-cell defects known in diabetes. Here, we investigated whether exenatide can alleviate such defects in proinsulin conversion, beta-cell survival, and insulin secretion, in the Ins2+/Akita beta-cells that have a spontaneous mutation (Cys 96 Tyr) in the insulin 2 gene caused dominant negative misfolding problem. 15 or 120 min exenatide administration substantially improves glucose-stimulated insulin secretion, marked in the secreted insulin levels and proinsulin/insulin ratio. This improvement is mainly due to enhanced conversion of proinsulin to insulin, having nothing to do with the prohormone convertase PC1/3 and PC2 levels. The 15 min improvement is calcium-independent. The 120 min improvement is linked to calcium and/or cAMP dependent mechanisms. This efficacy is validated during longer treatment and in Akita islets. Exenatide improves Ins2+/Akita beta-cell survival and Akita mouse's glucose tolerance. The results suggest a potential of incretin mimetics in alleviating defective proinsulin conversion and other proinsulin misfolding consequences.
Collapse
Affiliation(s)
- Wei Tang
- Department of Endocrinology, Jiangsu Province Geriatric Institute Islet Cell Senescence and Function Research Laboratory, Jiangsu Province Official Hospital, 65 Jiangsu Road, Nanjing 210024, China.
| | - Qingxin Yuan
- Department of Endocrinology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Bo Xu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China
| | - Kwame Osei
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Jie Wang
- Division of Endocrinology, Diabetes, and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA; Division of Endocrinology, Jiangsu Province Hospital on Integration of Chinese and Western Medicine, Nanjing University of Chinese and Western Medicine, Nanjing, Jiangsu 210028, China.
| |
Collapse
|
69
|
Liraglutide attenuates partial warm ischemia-reperfusion injury in rat livers. Naunyn Schmiedebergs Arch Pharmacol 2016; 390:311-319. [DOI: 10.1007/s00210-016-1330-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 12/07/2016] [Indexed: 12/27/2022]
|
70
|
Bodnaruc AM, Prud’homme D, Blanchet R, Giroux I. Nutritional modulation of endogenous glucagon-like peptide-1 secretion: a review. Nutr Metab (Lond) 2016; 13:92. [PMID: 27990172 PMCID: PMC5148911 DOI: 10.1186/s12986-016-0153-3] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 11/30/2016] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The positive influences of glucagon-like peptide-1 (GLP-1) on blood glucose homeostasis, appetite sensations, and food intake provide a strong rationale for its therapeutic potential in the nutritional management of obesity and type 2 diabetes. AIM To summarize GLP-1 physiology and the nutritional modulation of its secretion in the context of obesity and type 2 diabetes management. FINDINGS GLP-1 is mainly synthesized and secreted by enteroendocrine L-cells of the gastrointestinal tract. Its secretion is partly mediated by the direct nutrient sensing by G-protein coupled receptors which specifically bind to monosaccharides, peptides and amino-acids, monounsaturated and polyunsaturated fatty acids as well as to short chain fatty acids. Foods rich in these nutrients, such as high-fiber grain products, nuts, avocados and eggs also seem to influence GLP-1 secretion and may thus promote associated beneficial outcomes in healthy individuals as well as individuals with type 2 diabetes or with other metabolic disturbances. CONCLUSION The stimulation of endogenous GLP-1 secretion by manipulating the composition of the diet may be a relevant strategy for obesity and type 2 diabetes management. A better understanding of the dose-dependent effects as well as the synergistic effects of nutrients and whole foods is needed in order to develop recommendations to appropriately modify the diet to enhance GLP-1 beneficial effects.
Collapse
Affiliation(s)
- Alexandra M. Bodnaruc
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, 35, University Private, Room 050F, K1N 6N5 Ottawa, ON Canada
- Institut de Recherche de l’Hôpital Montfort, Institut du savoir, 745 Montreal Road, Room 202, K1K 0T2 Ottawa, ON Canada
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, 35 University Private, Room 050F, K1N 6N5 Ottawa, ON Canada
| | - Denis Prud’homme
- School of Human Kinetics, Faculty of Health Sciences, University of Ottawa, 35, University Private, Room 050F, K1N 6N5 Ottawa, ON Canada
- Institut de Recherche de l’Hôpital Montfort, Institut du savoir, 745 Montreal Road, Room 202, K1K 0T2 Ottawa, ON Canada
| | - Rosanne Blanchet
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, 35 University Private, Room 050F, K1N 6N5 Ottawa, ON Canada
| | - Isabelle Giroux
- Institut de Recherche de l’Hôpital Montfort, Institut du savoir, 745 Montreal Road, Room 202, K1K 0T2 Ottawa, ON Canada
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, 25 University Private, Room 116, K1N 6N5 Ottawa, ON Canada
| |
Collapse
|
71
|
EX4 stabilizes and activates Nrf2 via PKCδ, contributing to the prevention of oxidative stress-induced pancreatic beta cell damage. Toxicol Appl Pharmacol 2016; 315:60-69. [PMID: 27939242 DOI: 10.1016/j.taap.2016.12.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 11/12/2016] [Accepted: 12/06/2016] [Indexed: 12/21/2022]
Abstract
Oxidative stress in pancreatic beta cells can inhibit insulin secretion and promote apoptotic cell death. Exendin-4 (EX4), a glucagon-like peptide-1 receptor agonist, can suppress beta cell apoptosis, improve beta cell function and protect against oxidative damage. In this study, we investigated the molecular mechanisms for antioxidative effects of EX4 in pancreatic beta cells. INS-1 cells, a rat insulinoma cell line, were pretreated with EX4 and exposed to palmitate or H2O2. Reactive oxygen species (ROS) production, and glutathione and insulin secretion were measured. The mRNA and protein expression levels of antioxidant genes were examined. The level of nuclear factor erythroid 2-related factor 2 (Nrf2), its binding to antioxidant response element (ARE), and its ubiquination in the presence of EX4 were determined. The Nrf2 signaling pathway was determined using rottlerin (protein kinase [PK]Cδ inhibitor), H89 (PKA inhibitor) and LY294002 (phosphatidylinositide 3-kinase [PI3K] inhibitor). EX4 treatment decreased ROS production, recovered cellular glutathione levels and insulin secretion in the presence of oxidative stress in INS-1 cells. The expression levels of glutamate-cysteine ligase catalytic subunit and heme oxygenase-1 were increased by EX4 treatment. EX4 promoted Nrf2 translocation, ARE binding activity and enhanced stabilization of Nrf2 by inhibition of ubiquitination. Knockdown of Nrf2 abolished the effect of EX4 on increased insulin secretion. Inhibition of PKCδ attenuated Nrf2 translocation and antioxidative gene expression by EX4 treatment. We suggest that EX4 activates and stabilizes Nrf2 through PKCδ activation, contributing to the increase of antioxidant gene expression and consequently improving beta cell function in the presence of oxidative stress.
Collapse
|
72
|
Yoo BK, Triller DM, Yoo DJ. Formulary Forum: Exenatide: A New Option for the Treatment of Type 2 Diabetes. Ann Pharmacother 2016; 40:1777-84. [PMID: 16985091 DOI: 10.1345/aph.1h060] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Objective: To evaluate available literature characterizing the pharmacology, pharmacokinetics, drug interactions, efficacy, and safety of exenatide in patients with type 2 diabetes. Data Sources: A PubMed database search (1966–May 2006) was conducted, using exenatide as the search term. The manufacturer's prescribing information was also used. Study Selection and Data Extraction: English-language articles were selected and data were extracted with a focus on clinical outcomes associated with the treatment of patients with type 2 diabetes. Data Synthesis: Exenatide exerts a glucoregulatory effect by various mechanisms including secretion of glucose-dependent insulin, suppression of inappropriately high glucagon levels in patients with type 2 diabetes, delayed gastric emptying, and reduction of food intake. In placebo-controlled clinical studies, plasma exenatide concentrations appeared to exhibit dose-proportional kinetics, reaching peak plasma levels between 2 and 3 hours after a single subcutaneous injection. Exenatide's elimination half-life ranged from 3.3 to 4.0 hours, and the time to reach maximum concentration was about 2 hours. Interactions between exenatide and agents such as digoxin and lisinopril were not considered significant. In Phase III trials, exenatide demonstrated significant reduction of hemoglobin A1c levels from baseline and compared with placebo after 30 weeks of treatment in patients unable to achieve optimal glycemic control with metformin, a sulfonylurea, or oral combination therapy (0.4–0.9%). Patients' weight decreased with exenatide (0.9–2.8 kg), but adverse gastrointestinal (GI) events were common. Exenatide combined with thiazolidonediones, D-phenylalanine derivatives, meglitinides, or alpha glucosidase inhibitors has not been studied. Conclusions: Clinical trials have demonstrated that exenatide improves glycemic control when added to sulfonylureas and metformin, and it may be an alternative to insulin glargine in patients requiring additional therapy. Hypoglycemia has been encountered in clinical trials of exenatide, especially upon initiation of therapy with sulfonylureas (not with metformin); close patient monitoring is therefore recommended. Further studies should assess the impact of exenatide on clinical outcomes such as micro- and macrovascular disease.
Collapse
Affiliation(s)
- Bong Kyu Yoo
- Department of Pharmacy Practice, College of Pharmacy, Yeungnam University, Kyungsan, Kyungbuk, Korea.
| | | | | |
Collapse
|
73
|
Liu J, Hu Y, Zhang H, Xu Y, Wang G. Exenatide treatment increases serum irisin levels in patients with obesity and newly diagnosed type 2 diabetes. J Diabetes Complications 2016; 30:1555-1559. [PMID: 27503404 DOI: 10.1016/j.jdiacomp.2016.07.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/20/2016] [Accepted: 07/20/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Irisin is a myokine secreted by skeletal muscle during exercise. Abnormal serum irisin levels are associated with obesity and type 2 diabetes (T2D). This study investigated the changes in serum irisin in the obese patients with newly diagnosed T2D following glucagon-like peptide-1 (GLP-1) receptor agonist (exenatide) treatment. METHODS Fifty-four obese patients with T2D were treated with exenatide for 12weeks. The control group included 54 age-, sex-, and body mass index (BMI)-matched subjects with normal glucose tolerance. RESULTS Patients with T2D had lower irisin than the control group (38.06 [29.29-53.79] vs. 58.01 [43.07-87.79] ng/mL, P<0.01]. Serum irisin was negatively associated with BMI (r=-0.178, P<0.05), fasting blood glucose (FBG; r=-0.170, P<0.05), and glycosylated hemoglobin (HbA1c; r=-0.189, P<0.01) in patients with T2D. Exenatide treatment markedly increased serum irisin by 19.28ng/mL (12.59-25.98) compared to baseline (P<0.01). Increased irisin was significantly correlated with decreased FBG and HbA1c after exenatide treatment (FBG: r=-0.35; HbA1c: r=-0.37; both P<0.05). CONCLUSIONS Exenatide treatment significantly increased irisin in patients with T2D. Post-treatment changes in irisin were correlated with decreases in FBG and HbA1c. The upregulation of irisin might be a novel mechanism for the beneficial effects of exenatide in type 2 diabetic patients.
Collapse
Affiliation(s)
- Jia Liu
- Department of Endocrinology; Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yanjin Hu
- Department of Endocrinology; Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China
| | - Heng Zhang
- Department of Endocrinology; Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China
| | - Yuan Xu
- Department of Endocrinology; Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China
| | - Guang Wang
- Department of Endocrinology; Beijing Chao-yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
74
|
Abstract
INTRODUCTION Many persons with type 1 diabetes do not achieve glycemic targets, why new treatments, complementary to insulin, are of interest. Liraglutide, a long-acting glucagon-like peptide-1 receptor agonist could be a potential pharmacological supplement to insulin. This review discusses the mechanism of actions, efficacy and safety of liraglutide as add-on to insulin in persons with type 1 diabetes. AREAS COVERED Physiological and clinical data on liraglutide in type 1 diabetes were reviewed. We searched the Cochrane library, MEDLINE and EMBASE, with the final search performed February 16, 2016. EXPERT OPINION Liraglutide as adjunct to insulin treatment reduced body weight and daily dose of insulin compared with insulin alone. The effect on HbA1c was inconsistent with mostly uncontrolled, small-scale studies reporting improvements in glycemic control. In placebo-controlled studies there was no clinically relevant effect on HbA1c. Adverse events were mostly transient gastrointestinal side effects, primarily nausea. Based on the available data, liraglutide cannot be recommended as add-on therapy to insulin in persons with type 1 diabetes with the aim to improve glycemic control. Ongoing trials in newly diagnosed patients with type 1 diabetes and in insulin pump-treated patients will help define the future role of liraglutide therapy in type 1 diabetes.
Collapse
Affiliation(s)
- Thomas Fremming Dejgaard
- a Department of Endocrinology, Hvidovre Hospital , University of Copenhagen , Hvidovre , Denmark.,b Department of Clinical Research, Steno Diabetes Center , Gentofte , Denmark
| | | | - Jens Juul Holst
- c NNF Center for Basic Metabolic Research , University of Copenhagen , Copenhagen , Denmark.,d Department of Biomedical Sciences, Faculty of Health and Medical Sciences , University of Copenhagen , Copenhagen , Denmark
| | - Sten Madsbad
- a Department of Endocrinology, Hvidovre Hospital , University of Copenhagen , Hvidovre , Denmark.,c NNF Center for Basic Metabolic Research , University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
75
|
Effect of Exendin-4 on Autophagy Clearance in Beta Cell of Rats with Tacrolimus-induced Diabetes Mellitus. Sci Rep 2016; 6:29921. [PMID: 27436514 PMCID: PMC4951772 DOI: 10.1038/srep29921] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 06/27/2016] [Indexed: 12/11/2022] Open
Abstract
Growing evidence suggests that GLP-1 protects beta cells against various cellular injuries by modulating autophagy. In this study, we examined whether exendin-4 (Ex-4), a GLP-1 analog, had preventive effects on tacrolimus (Tac)-induced beta cell injury by improving autophagy clearance. Rats with Tac-induced diabetes mellitus exhibited increased autophagy-associated protein expression, light chain 3B levels, and autophagic vacuole numbers in pancreatic beta cells. Additionally, Tac increased autophagy in a dose- and time-dependent manner in vitro, and inhibition of autophagosome using 3-methyladenine reduced Tac-induced islet cell injury by decreasing reactive oxygen species production and apoptosis. Ex-4 treatment decreased Tac-induced hyperglycaemia, oxidative stress, and apoptosis, accompanied by decreased autophagy-associated protein expression and autophagosome numbers. In vivo and in vitro studies showed that Tac treatment impaired lysosomal function and autophagosome-lysosome fusion; these processes were improve by Ex-4 treatment. Moreover, addition of bafilomycin A1, an inhibitor of lysosomal function, abolished the protective effects of Ex-4. Our findings reveal that Tac-induced diabetes mellitus was a state of excessive burden of autophagosomes and impairment of autophagy clearance and that Ex-4 protected against Tac-induced pancreatic islet injury by reducing the burden of autophagosomes via activation of autophagosome clearance. Thus, Ex-4 had therapeutic effects on Tac-induced pancreatic beta cell injury.
Collapse
|
76
|
Timper K, Dalmas E, Dror E, Rütti S, Thienel C, Sauter NS, Bouzakri K, Bédat B, Pattou F, Kerr-Conte J, Böni-Schnetzler M, Donath MY. Glucose-Dependent Insulinotropic Peptide Stimulates Glucagon-Like Peptide 1 Production by Pancreatic Islets via Interleukin 6, Produced by α Cells. Gastroenterology 2016; 151:165-79. [PMID: 26971825 DOI: 10.1053/j.gastro.2016.03.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 02/19/2016] [Accepted: 03/03/2016] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Glucose-dependent insulinotropic peptide (GIP) induces production of interleukin 6 (IL6) by adipocytes. IL6 increases production of glucagon-like peptide (GLP)-1 by L cells and α cells, leading to secretion of insulin from β cells. We investigated whether GIP regulates GLP1 and glycemia via IL6. METHODS We obtained samples of human pancreatic islets and isolated islets from mice; human α cells and β cells were sorted by flow cytometry and incubated with GIP. Islets were analyzed by quantitative polymerase chain reaction and immunohistochemistry. BKS.Cg-Dock7m+/+ Leprdb/J db/db mice (diabetic mice) and db/+ mice, as well as C57BL/6J IL6-knockout mice (IL6-KO) and C57BL/6J mice with the full-length Il6 gene (controls), were fed a chow or a high-fat diet; some mice were given injections of recombinant GIP, IL6, GLP, a neutralizing antibody against IL6 (anti-IL6), lipopolysaccharide, and/or IL1B. Mice were given a glucose challenge and blood samples were collected and analyzed. RESULTS Incubation of mouse and human pancreatic α cells with GIP induced their production of IL6, leading to production of GLP1 and insulin secretion from pancreatic islets. This did not occur in islets from IL6-KO mice or in islets incubated with anti-IL6. Incubation of islets with IL1B resulted in IL6 production but directly reduced GLP1 production. Incubation of mouse islets with the sodium glucose transporter 2 inhibitor dapagliflozin induced production of GLP1 and IL6. Injection of control mice with GIP increased plasma levels of GLP1, insulin, and glucose tolerance; these effects were amplified in mice given lipopolysaccharide but reduced in IL6-KO mice or in mice given anti-IL6. Islets from diabetic mice had increased levels of IL1B and IL6, compared with db/+ mice, but injection of GIP did not lead to production of GLP1 or reduce glycemia. CONCLUSIONS In studies of pancreatic islets from human beings and mice, we found that GIP induces production of IL6 by α cells, leading to islet production of GLP1 and insulin. This process is regulated by inflammation, via IL1B, and by sodium glucose transporter 2. In diabetic mice, increased islet levels of IL6 and IL1B might increase or reduce the production of GLP1 and affect glycemia.
Collapse
Affiliation(s)
- Katharina Timper
- Clinic of Endocrinology, Diabetes and Metabolism, Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland.
| | - Elise Dalmas
- Clinic of Endocrinology, Diabetes and Metabolism, Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland
| | - Erez Dror
- Clinic of Endocrinology, Diabetes and Metabolism, Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland
| | - Sabine Rütti
- Department of Genetic Medicine and Development, Geneva University, Geneva, Switzerland
| | - Constanze Thienel
- Clinic of Endocrinology, Diabetes and Metabolism, Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland
| | - Nadine S Sauter
- Clinic of Endocrinology, Diabetes and Metabolism, Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland
| | - Karim Bouzakri
- Department of Genetic Medicine and Development, Geneva University, Geneva, Switzerland
| | - Benoit Bédat
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva School of Medicine, Geneva, Switzerland
| | | | | | - Marianne Böni-Schnetzler
- Clinic of Endocrinology, Diabetes and Metabolism, Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland
| | - Marc Y Donath
- Clinic of Endocrinology, Diabetes and Metabolism, Department of Biomedicine, University Hospital, University of Basel, Basel, Switzerland.
| |
Collapse
|
77
|
Iida A, Seino Y, Fukami A, Maekawa R, Yabe D, Shimizu S, Kinoshita K, Takagi Y, Izumoto T, Ogata H, Ishikawa K, Ozaki N, Tsunekawa S, Hamada Y, Oiso Y, Arima H, Hayashi Y. Endogenous GIP ameliorates impairment of insulin secretion in proglucagon-deficient mice under moderate beta cell damage induced by streptozotocin. Diabetologia 2016; 59:1533-1541. [PMID: 27053237 PMCID: PMC4901104 DOI: 10.1007/s00125-016-3935-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 03/02/2016] [Indexed: 01/06/2023]
Abstract
AIMS/HYPOTHESIS The action of incretin hormones including glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) is potentiated in animal models defective in glucagon action. It has been reported that such animal models maintain normoglycaemia under streptozotocin (STZ)-induced beta cell damage. However, the role of GIP in regulation of glucose metabolism under a combination of glucagon deficiency and STZ-induced beta cell damage has not been fully explored. METHODS In this study, we investigated glucose metabolism in mice deficient in proglucagon-derived peptides (PGDPs)-namely glucagon gene knockout (GcgKO) mice-administered with STZ. Single high-dose STZ (200 mg/kg, hSTZ) or moderate-dose STZ for five consecutive days (50 mg/kg × 5, mSTZ) was administered to GcgKO mice. The contribution of GIP to glucose metabolism in GcgKO mice was also investigated by experiments employing dipeptidyl peptidase IV (DPP4) inhibitor (DPP4i) or Gcg-Gipr double knockout (DKO) mice. RESULTS GcgKO mice developed severe diabetes by hSTZ administration despite the absence of glucagon. Administration of mSTZ decreased pancreatic insulin content to 18.8 ± 3.4 (%) in GcgKO mice, but ad libitum-fed blood glucose levels did not significantly increase. Glucose-induced insulin secretion was marginally impaired in mSTZ-treated GcgKO mice but was abolished in mSTZ-treated DKO mice. Although GcgKO mice lack GLP-1, treatment with DPP4i potentiated glucose-induced insulin secretion and ameliorated glucose intolerance in mSTZ-treated GcgKO mice, but did not increase beta cell area or significantly reduce apoptotic cells in islets. CONCLUSIONS/INTERPRETATION These results indicate that GIP has the potential to ameliorate glucose intolerance even under STZ-induced beta cell damage by increasing insulin secretion rather than by promoting beta cell survival.
Collapse
Affiliation(s)
- Atsushi Iida
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 4668550, Japan
| | - Yusuke Seino
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 4668550, Japan.
- Department of Metabolic Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Ayako Fukami
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 4668550, Japan
| | - Ryuya Maekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 4668550, Japan
| | - Daisuke Yabe
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Kobe, Japan
- Division of Molecular and Metabolic Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shinobu Shimizu
- Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Kobe, Japan
| | - Keita Kinoshita
- Research Center of Health, Physical Fitness and Sports, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Genetics, Division of Stress Adaptation and Recognition, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 4648601, Japan
| | - Yusuke Takagi
- Research Center of Health, Physical Fitness and Sports, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Genetics, Division of Stress Adaptation and Recognition, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 4648601, Japan
| | - Takako Izumoto
- Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidetada Ogata
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 4668550, Japan
| | - Kota Ishikawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 4668550, Japan
| | - Nobuaki Ozaki
- Research Center of Health, Physical Fitness and Sports, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
| | - Shin Tsunekawa
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 4668550, Japan
| | - Yoji Hamada
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 4668550, Japan
- Department of Metabolic Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yutaka Oiso
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 4668550, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 4668550, Japan
| | - Yoshitaka Hayashi
- Department of Genetics, Division of Stress Adaptation and Recognition, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 4648601, Japan.
| |
Collapse
|
78
|
Activation of GPR119 Stimulates Human β-Cell Replication and Neogenesis in Humanized Mice with Functional Human Islets. J Diabetes Res 2016; 2016:1620821. [PMID: 27413754 PMCID: PMC4927982 DOI: 10.1155/2016/1620821] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/22/2016] [Indexed: 01/09/2023] Open
Abstract
Using humanized mice with functional human islets, we investigated whether activating GPR119 by PSN632408, a small molecular agonist, can stimulate human β-cell regeneration in vivo. Human islets were transplanted under the left kidney capsule of immunodeficient mice with streptozotocin- (STZ-) induced diabetes. The recipient mice were treated with PSN632408 or vehicle and BrdU daily. Human islet graft function in the mice was evaluated by nonfasting glucose levels, oral glucose tolerance, and removal of the grafts. Immunostaining for insulin, glucagon, and BrdU or Ki67 was performed in islet grafts to evaluate α- and β-cell replication. Insulin and CK19 immunostaining was performed to evaluate β-cell neogenesis. Four weeks after human islet transplantation, 71% of PSN632408-treated mice achieved normoglycaemia compared with 24% of vehicle-treated mice. Also, oral glucose tolerance was significantly improved in the PSN632408-treated mice. PSN632408 treatment significantly increased both human α- and β-cell areas in islet grafts and stimulated α- and β-cell replication. In addition, β-cell neogenesis was induced from pancreatic duct cells in the islet grafts. Our results demonstrated that activation of GPR119 increases β-cell mass by stimulating human β-cell replication and neogenesis. Therefore, GPR119 activators may qualify as therapeutic agents to increase human β-cell mass in patients with diabetes.
Collapse
|
79
|
Cui JP, Zhang MJ, Liu BL. Research progress of mesenchymal stem cells combined with islet transplantation in treatment of type I diabetes mellitus. Shijie Huaren Xiaohua Zazhi 2016; 24:2213-2218. [DOI: 10.11569/wcjd.v24.i14.2213] [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] [Indexed: 02/06/2023] Open
Abstract
The most significant feature of type I diabetes is β-cell loss, which results in a series of complications. While β-cell loss occurs, β-cells are ultimately damaged by macrophages and T cells in the presence of inflammatory mediators. Because of this characteristic, five kinds of antibodies are commonly used in clinical practice to diagnose and evaluate β-cell loss, including islet cell antibody, insulin antibody, GAD65, IA-2 and IA-2b. In addition to the HLA gene related factors, environmental factors, such as infection, diet and physiological and psychological factors, are suspected to be causes of this disease. At present, there are many treatments for type I diabetes, and the clinical goal is to control blood glucose, prevent further damage of βcells and control patients' own immune response. In 1992, the discovery of insulin, which converts the fatal diabetes into a chronic disease, to some extent, delayed the progression of microvascular complications; however, it is not able to delay the progression of the disease. β-cell transplantation is currently the only minimally invasive means for reasonable control of blood glucose control disease related complications. Although whole pancreas transplantation can achieve a promising effect to some extent, it is accompanied by high incidence and mortality, as well as lifelong mandatory immune suppression. Bone marrow mesenchymal stem cells transplantation, lipopolysaccharideon (LPS) bone marrow mesenchymal stem cell pretreatment and islet cell exendin-4 liquid preservation reduce warm ischemia time damage and provide new avenues for islet cell transplantation.
Collapse
|
80
|
Barbetti F, Colombo C, Haataja L, Cras-Méneur C, Bernardini S, Arvan P. Hyperglucagonemia in an animal model of insulin- deficient diabetes: what therapy can improve it? Clin Diabetes Endocrinol 2016; 2:11. [PMID: 28702245 PMCID: PMC5471666 DOI: 10.1186/s40842-016-0029-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/11/2016] [Indexed: 02/06/2023] Open
Abstract
Background Intra-islet insulin contributes to alpha-cell suppression. Akita mice carry a toxic-gain-of- function Ins2 gene mutation encoding proinsulin-C(A7)Y, similar to that described in human Mutant Ins-gene induced Diabetes of Youth, which decreases intra-islet insulin. Herein, we examined Akita mice for examination of circulating insulin and circulating glucagon levels. The possibility that loss of intra-islet suppression of alpha-cells, with increased circulating glucagon, contributes to diabetes under conditions of intra-islet insulin deficiency, raises questions about effective treatments that may be available. Methods Blood glucose, plasma insulin, C-peptide I, C-peptide II, and glucagon were measured at various times during development of diabetes in Akita mice. We also used Akita- like hProC(A7)Y-CpepGFP transgenic mice in Ins2+/+, Ins2+/− and Ins2−/− genetic backgrounds (providing animals with greater or lesser defects in islet insulin production, respectively) in order to examine the relative abundance of immunostainable intra-islet glucagon-positive and insulin-positive cells. Similar measurements were made in Akita mice. Finally, the effects of treatment with insulin, exendin-4, and leptin on blood glucose were then compared in Akita mice. Results Interestingly, total insulin levels in the circulation were not frankly low in Akita mice, although they did not rise appropriately with the onset of hyperglycemia. By contrast, in severely diabetic Akita mice at 6 weeks of age, circulating glucagon levels were significantly elevated. Additionally, in Ins2+/− and Ins2−/− mice bearing the Akita-like hProC(A7)Y-CpepGFP transgene, development of diabetes correlated with an increase in the relative intra-islet abundance of immunostainable glucagon-positive cells, and a similar observation was made in Akita islets. In Akita mice, whereas a brief treatment with exendin-4 resulted in no apparent improvement in hyperglycemia, leptin treatment resulted in restoration of normoglycemia. Curiously, leptin treatment also suppressed circulating glucagon levels. Conclusions Loss of insulin-mediated intra-islet suppression of glucagon production may be a contributor to hyperglycemia in Akita mice, and leptin treatment appears beneficial in such a circumstance. This treatment might also be considered in some human diabetes patients with diminished insulin reserve.
Collapse
Affiliation(s)
- Fabrizio Barbetti
- Department of Experimental Medicine and Surgery, University of Tor Vergata, Rome, Italy.,Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.,Department of Experimental Medicine and Surgery, University of Tor Vergata. Tor Vergata University Hospital, first floor, section D, room 118, Viale Oxford 81, 00133 Rome, Italy
| | - Carlo Colombo
- Department of Experimental Medicine and Surgery, University of Tor Vergata, Rome, Italy
| | - Leena Haataja
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical Center, Ann Arbor, MI USA
| | - Corentin Cras-Méneur
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical Center, Ann Arbor, MI USA
| | - Sergio Bernardini
- Department of Experimental Medicine and Surgery, University of Tor Vergata, Rome, Italy
| | - Peter Arvan
- Division of Metabolism, Endocrinology & Diabetes, University of Michigan Medical Center, Ann Arbor, MI USA.,University of Michigan Medical Center, Brehm Tower room 5112, 1000 Wall St., Ann Arbor, MI 48105 USA
| |
Collapse
|
81
|
Exenatide reverses dysregulated microRNAs in high-fat diet-induced obese mice. Obes Res Clin Pract 2016; 10:315-26. [DOI: 10.1016/j.orcp.2015.07.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 06/28/2015] [Accepted: 07/20/2015] [Indexed: 12/24/2022]
|
82
|
Tudurí E, López M, Diéguez C, Nadal A, Nogueiras R. Glucagon-Like Peptide 1 Analogs and their Effects on Pancreatic Islets. Trends Endocrinol Metab 2016; 27:304-318. [PMID: 27062006 DOI: 10.1016/j.tem.2016.03.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/04/2016] [Accepted: 03/04/2016] [Indexed: 02/07/2023]
Abstract
Glucagon-like peptide 1 (GLP-1) exerts many actions that improve glycemic control. GLP-1 stimulates glucose-stimulated insulin secretion and protects β cells, while its extrapancreatic effects include cardioprotection, reduction of hepatic glucose production, and regulation of satiety. Although an appealing antidiabetic drug candidate, the rapid degradation of GLP-1 by dipeptidyl peptidase 4 (DPP-4) means that its therapeutic use is unfeasible, and this prompted the development of two main GLP-1 therapies: long-acting GLP-1 analogs and DPP-4 inhibitors. In this review, we focus on the pancreatic effects exerted by current GLP-1 derivatives used to treat diabetes. Based on the results from in vitro and in vivo studies in humans and animal models, we describe the specific actions of GLP-1 analogs on the synthesis, processing, and secretion of insulin, islet morphology, and β cell proliferation and apoptosis.
Collapse
Affiliation(s)
- Eva Tudurí
- Instituto de Investigaciones Sanitarias (IDIS), CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain.
| | - Miguel López
- Instituto de Investigaciones Sanitarias (IDIS), CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain; Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Carlos Diéguez
- Instituto de Investigaciones Sanitarias (IDIS), CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain; Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain
| | - Angel Nadal
- Instituto de Bioingeniería and CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Universidad Miguel Hernández, Elche, Spain
| | - Rubén Nogueiras
- Instituto de Investigaciones Sanitarias (IDIS), CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela 15706, Spain; Department of Physiology, CIMUS, University of Santiago de Compostela, Santiago de Compostela 15782, Spain.
| |
Collapse
|
83
|
Schernthaner-Reiter MH, Schernthaner G. Combination therapy of SGLT2 inhibitors with incretin-based therapies for the treatment of type 2 diabetes mellitus: Effects and mechanisms of action. Expert Rev Endocrinol Metab 2016; 11:281-296. [PMID: 30058933 DOI: 10.1586/17446651.2016.1151783] [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] [Indexed: 11/08/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is a growing health problem worldwide; its pathogenesis is multifactorial and its progressive nature often necessitates a combination therapy with multiple antihyperglycemic agents. Sodium glucose cotransporter 2 (SGLT2) inhibitors and the incretin-based therapies - dipeptidyl peptidase 4(DPP-4) inhibitors and glucagon-like peptide 1 (GLP-1) receptor agonists - were introduced for the treatment of T2DM within the last decade. Evidence of the beneficial effects of these antihyperglycemic agents on micro- and macrovascular complications have started to emerge, which will become important in individualizing different combinations of antihyperglycemic agents to different patient populations. We review here the mechanisms of action, glycemic and cardiovascular effects of SGLT2 inhibitors and incretin-based therapies and their combination in the treatment of T2DM.
Collapse
Affiliation(s)
- Marie Helene Schernthaner-Reiter
- a Clinical Division of Endocrinology and Metabolism, Department of Internal Medicine III , Medical University of Vienna , Vienna , Austria
| | | |
Collapse
|
84
|
Brill AL, Wisinski JA, Cadena MT, Thompson MF, Fenske RJ, Brar HK, Schaid MD, Pasker RL, Kimple ME. Synergy Between Gαz Deficiency and GLP-1 Analog Treatment in Preserving Functional β-Cell Mass in Experimental Diabetes. Mol Endocrinol 2016; 30:543-56. [PMID: 27049466 DOI: 10.1210/me.2015-1164] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A defining characteristic of type 1 diabetes mellitus (T1DM) pathophysiology is pancreatic β-cell death and dysfunction, resulting in insufficient insulin secretion to properly control blood glucose levels. Treatments that promote β-cell replication and survival, thus reversing the loss of β-cell mass, while also preserving β-cell function, could lead to a real cure for T1DM. The α-subunit of the heterotrimeric Gz protein, Gαz, is a tonic negative regulator of adenylate cyclase and downstream cAMP production. cAMP is one of a few identified signaling molecules that can simultaneously have a positive impact on pancreatic islet β-cell proliferation, survival, and function. The purpose of our study was to determine whether mice lacking Gαz might be protected, at least partially, from β-cell loss and dysfunction after streptozotocin treatment. We also aimed to determine whether Gαz might act in concert with an activator of the cAMP-stimulatory glucagon-like peptide 1 receptor, exendin-4 (Ex4). Without Ex4 treatment, Gαz-null mice still developed hyperglycemia, albeit delayed. The same finding held true for wild-type mice treated with Ex4. With Ex4 treatment, Gαz-null mice were protected from developing severe hyperglycemia. Immunohistological studies performed on pancreas sections and in vitro apoptosis, cytotoxicity, and survival assays demonstrated a clear effect of Gαz signaling on pancreatic β-cell replication and death; β-cell function was also improved in Gαz-null islets. These data support our hypothesis that a combination of therapies targeting both stimulatory and inhibitory pathways will be more effective than either alone at protecting, preserving, and possibly regenerating β-cell mass and function in T1DM.
Collapse
Affiliation(s)
- Allison L Brill
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Jaclyn A Wisinski
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Mark T Cadena
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Mary F Thompson
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Rachel J Fenske
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Harpreet K Brar
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Michael D Schaid
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Renee L Pasker
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| | - Michelle E Kimple
- Department of Medicine (A.L.B., J.A.W., M.T.C., M.F.T., H.K.B., R.L.P., M.E.K.), Division of Endocrinology, Diabetes, and Metabolism; Department of Cell and Regenerative Biology (M.E.K.); and Interdisciplinary Graduate Program in Nutritional Sciences (R.J.F., M.D.S., M.E.K.), University of Wisconsin-Madison, Madison; and Research Service (A.L.B., J.A.W., M.T.C., M.F.T., R.J.F., H.K.B., M.D.S., M.E.K.), William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705
| |
Collapse
|
85
|
Chang TJ, Tseng HC, Liu MW, Chang YC, Hsieh ML, Chuang LM. Glucagon-like peptide-1 prevents methylglyoxal-induced apoptosis of beta cells through improving mitochondrial function and suppressing prolonged AMPK activation. Sci Rep 2016; 6:23403. [PMID: 26997114 PMCID: PMC4800673 DOI: 10.1038/srep23403] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 03/02/2016] [Indexed: 11/09/2022] Open
Abstract
Accumulation of methylglyoxal (MG) contributes to glucotoxicity and mediates beta cell apoptosis. The molecular mechanism by which GLP-1 protects MG-induced beta cell apoptosis remains unclear. Metformin is a first-line drug for treating type 2 diabetes associated with AMPK activation. However, whether metformin prevents MG-induced beta cell apoptosis is controversial. Here, we explored the signaling pathway involved in the anti-apoptotic effect of GLP-1, and investigated whether metformin had an anti-apoptotic effect on beta cells. MG treatment induced apoptosis of beta cells, impaired mitochondrial function, and prolonged activation of AMP-dependent protein kinase (AMPK). The MG-induced pro-apoptotic effects were abolished by an AMPK inhibitor. Pretreatment of GLP-1 reversed MG-induced apoptosis, and mitochondrial dysfunction, and suppressed prolonged AMPK activation. Pretreatment of GLP-1 reversed AMPK activator 5-aminoimidazole-4-carboxamide riboside (AICAR)-induced apoptosis, and suppressed prolonged AMPK activation. However, metformin neither leads to beta cell apoptosis nor ameliorates MG-induced beta cell apoptosis. In parallel, GLP-1 also prevents MG-induced beta cell apoptosis through PKA and PI3K-dependent pathway. In conclusion, these data indicates GLP-1 but not metformin protects MG-induced beta cell apoptosis through improving mitochondrial function, and alleviating the prolonged AMPK activation. Whether adding GLP-1 to metformin provides better beta cell survival and delays disease progression remains to be validated.
Collapse
Affiliation(s)
- Tien-Jyun Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Hsing-Chi Tseng
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan.,Institute of Molecular Medicine, National Taiwan University Medical College, Taipei 10002, Taiwan
| | - Meng-Wei Liu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan
| | - Yi-Cheng Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan.,Graduate Institute of Medical Genomics and Proteomics, National Taiwan University Medical College, Taipei 10002, Taiwan.,Institute of Biomedical Science, Academia Sinica, Taipei, 11500, Taiwan
| | - Meng-Lun Hsieh
- Graduate Institute of Medical Genomics and Proteomics, National Taiwan University Medical College, Taipei 10002, Taiwan
| | - Lee-Ming Chuang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei 10002, Taiwan.,Institute of Molecular Medicine, National Taiwan University Medical College, Taipei 10002, Taiwan
| |
Collapse
|
86
|
Linnemann AK, Davis DB. Glucagon-like peptide-1 and cholecystokinin production and signaling in the pancreatic islet as an adaptive response to obesity. J Diabetes Investig 2016; 7 Suppl 1:44-9. [PMID: 27186355 PMCID: PMC4854504 DOI: 10.1111/jdi.12465] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 12/21/2015] [Indexed: 12/22/2022] Open
Abstract
Precise control of blood glucose is dependent on adequate β‐cell mass and function. Thus, reductions in β‐cell mass and function lead to insufficient insulin production to meet demand, and result in diabetes. Recent evidence suggests that paracrine signaling in the islet might be important in obesity, and disruption of this signaling could play a role in the pathogenesis of diabetes. For example, we recently discovered a novel islet incretin axis where glucagon‐like peptide‐1 regulates β‐cell production of another classic gut hormone, cholecystokinin. This axis is stimulated by obesity, and plays a role in enhancing β‐cell survival. In the present review, we place our observations in the wider context of the literature on incretin regulation in the islet, and discuss the potential for therapeutic targeting of these pathways.
Collapse
Affiliation(s)
- Amelia K Linnemann
- Department of Medicine Division of Endocrinology University of Wisconsin-Madison Madison Wisconsin USA
| | - Dawn Belt Davis
- Department of MedicineDivision of EndocrinologyUniversity of Wisconsin-MadisonMadisonWisconsinUSA; William S Middleton Memorial Veterans HospitalMadisonWisconsinUSA
| |
Collapse
|
87
|
Jiang D, Wang Y, Zang Y, Liu X, Zhao L, Wang Q, Liu C, Feng W, Yin X, Fang Y. Neuroprotective Effects of rhGLP-1 in Diabetic Rats with Cerebral Ischemia/Reperfusion Injury. Drug Dev Res 2016; 77:124-33. [PMID: 26971396 DOI: 10.1002/ddr.21297] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/05/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Daoli Jiang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy; Xuzhou Medical College; Xuzhou 221004 China
| | - Yitong Wang
- Department of Pharmacy; Peking University People's Hospital; Beijing 100044 China
- Department of Pharmacy Administration and Clinical Pharmacy; Peking University Health Science Center; Beijing 100191 China
| | - Yannan Zang
- Department of Pharmacy; Peking University People's Hospital; Beijing 100044 China
- Department of Pharmacy Administration and Clinical Pharmacy; Peking University Health Science Center; Beijing 100191 China
| | - Xiaofang Liu
- Zhongda Hospital, School of Medicine; Southeast University; Nanjing 210009 China
| | - Libo Zhao
- Department of Pharmacy; Peking University People's Hospital; Beijing 100044 China
| | - Qian Wang
- Department of Pharmacy; Peking University People's Hospital; Beijing 100044 China
| | - Chang Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy; Xuzhou Medical College; Xuzhou 221004 China
| | - Wanyu Feng
- Department of Pharmacy; Peking University People's Hospital; Beijing 100044 China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy; Xuzhou Medical College; Xuzhou 221004 China
| | - Yi Fang
- Department of Pharmacy; Peking University People's Hospital; Beijing 100044 China
| |
Collapse
|
88
|
Röder PV, Wu B, Liu Y, Han W. Pancreatic regulation of glucose homeostasis. Exp Mol Med 2016; 48:e219. [PMID: 26964835 PMCID: PMC4892884 DOI: 10.1038/emm.2016.6] [Citation(s) in RCA: 474] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 12/03/2015] [Accepted: 12/06/2015] [Indexed: 12/11/2022] Open
Abstract
In order to ensure normal body function, the human body is dependent on a tight control of its blood glucose levels. This is accomplished by a highly sophisticated network of various hormones and neuropeptides released mainly from the brain, pancreas, liver, intestine as well as adipose and muscle tissue. Within this network, the pancreas represents a key player by secreting the blood sugar-lowering hormone insulin and its opponent glucagon. However, disturbances in the interplay of the hormones and peptides involved may lead to metabolic disorders such as type 2 diabetes mellitus (T2DM) whose prevalence, comorbidities and medical costs take on a dramatic scale. Therefore, it is of utmost importance to uncover and understand the mechanisms underlying the various interactions to improve existing anti-diabetic therapies and drugs on the one hand and to develop new therapeutic approaches on the other. This review summarizes the interplay of the pancreas with various other organs and tissues that maintain glucose homeostasis. Furthermore, anti-diabetic drugs and their impact on signaling pathways underlying the network will be discussed.
Collapse
Affiliation(s)
- Pia V Röder
- Metabolism in Human Diseases Unit, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Metabolism in Human Diseases Unit, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore. E-mail: or
| | - Bingbing Wu
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, A*STAR, Singapore, Singapore
| | - Yixian Liu
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, A*STAR, Singapore, Singapore
| | - Weiping Han
- Metabolism in Human Diseases Unit, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Laboratory of Metabolic Medicine, Singapore Bioimaging Consortium, A*STAR, Singapore, Singapore
- Metabolism in Human Diseases Unit, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore 138673, Singapore. E-mail: or
| |
Collapse
|
89
|
Du J, Zhang L, Wang Z, Yano N, Zhao YT, Wei L, Dubielecka-Szczerba P, Liu PY, Zhuang S, Qin G, Zhao TC. Exendin-4 induces myocardial protection through MKK3 and Akt-1 in infarcted hearts. Am J Physiol Cell Physiol 2016; 310:C270-83. [PMID: 26739490 DOI: 10.1152/ajpcell.00194.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/18/2015] [Indexed: 11/22/2022]
Abstract
We have demonstrated that glucagon like peptide-1 (GLP-1) protects the heart against ischemic injury. However, the physiological mechanism by which GLP-1 receptor (GLP-1R) initiates cardioprotection remains to be determined. The objective of this study is to elucidate the functional roles of MAPK kinase 3 (MKK3) and Akt-1 in mediating exendin-4-elicited protection in the infarcted hearts. Adult mouse myocardial infarction (MI) was created by ligation of the left descending artery. Wild-type, MKK3(-/-), Akt-1(-/-), and Akt-1(-/-);MKK3(-/-) mice were divided into one of several groups: 1) sham: animals underwent thoracotomy without ligation; 2) MI: animals underwent MI and received a daily dose of intraperitoneal injection of vehicle (saline); 3) MI + exendin-4: infarcted mice received daily injections of exendin-4, a GLP-1R agonist (0.1 mg/kg, ip). Echocardiographic measurements indicate that exendin-4 treatment resulted in the preservation of ventricular function and increases in the survival rate, but these effects were diminished in MKK3(-/-), Akt-1(-/-), and Akt-1(-/-);MKK3(-/-) mice. Exendin-4 treatments suppressed cardiac hypotrophy and reduced scar size and cardiac interstitial fibrosis, respectively, but these beneficial effects were lost in genetic elimination of MKK3, Akt-1, or Akt-1(-/-);MKK3(-/-) mice. GLP-1R stimulation stimulated angiogenic responses, which were also mitigated by deletion of MKK3 and Akt-1. Exendin-4 treatment increased phosphorylation of MKK3, p38, and Akt-1 at Ser129 but decreased levels of active caspase-3 and cleaved poly (ADP-ribose) polymerase; these proteins were diminished in MKK3(-/-), Akt-1(-/-), and Akt-1(-/-);MKK3(-/-) mice. These results reveal that exendin-4 treatment improves cardiac function, attenuates cardiac remodeling, and promotes angiogenesis in the infarcted myocardium through MKK3 and Akt-1 pathway.
Collapse
Affiliation(s)
- Jianfeng Du
- Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, Rhode Island
| | - Ling Zhang
- Department of Emergency Medicine, Alpert Medical School, Brown University, Providence, Rhode Island
| | - Zhengke Wang
- Department of Dermatology, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, Rhode Island
| | - Naohiro Yano
- Women and Infants Hospital, Alpert Medical School, Brown University, Providence, Rhode Island
| | - Yu Tina Zhao
- Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, Rhode Island
| | - Lei Wei
- Department of Orthopedics, Rhode Island Hospital, Alpert Medical School, Brown University, Providence, Rhode Island
| | | | - Paul Y Liu
- Department of Plastic Surgery, Alpert Medical School, Brown University, Providence, Rhode Island
| | - Shougang Zhuang
- Department of Medicine, Alpert Medical School, Brown University, Providence, Rhode Island
| | - Gangjian Qin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ting C Zhao
- Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, Rhode Island;
| |
Collapse
|
90
|
Zhang L, Song X, Zhou L, Liang G, Xu H, Wang F, Huang F, Jiang G. Accumulation of intestinal tissue 3-deoxyglucosone attenuated GLP-1 secretion and its insulinotropic effect in rats. Diabetol Metab Syndr 2016; 8:78. [PMID: 27956941 PMCID: PMC5129672 DOI: 10.1186/s13098-016-0194-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 11/24/2016] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Our recent findings support the idea that 3-deoxyglucosone (3DG), a dietary composition, has been suggested as an independent factor for the development of prediabetes. Secretion of glucagon-like peptide-1 (GLP-1) has been suggested to be impaired in T2DM and in conditions associated with hyperglycemia. Since low oral bioavailability of 3DG has been indicated in a single administration study, in the present study we examined if 3DG is capable of accumulating in intestinal tissue of rats after 2-week administration of 3DG, and the 3DG treatment affects GLP-1 secretion and glucose tolerance. METHODS Rats were administered by gastric gavage for 2 weeks. We measured 3DG contents of intestinal tissues (by HPLC), plasma levels of total GLP-1 (by ELISA), insulin and glucagon (both by radioimmunoassay) and blood glucose concentrations. The expressions of the sweet receptor subunits (TAS1R2, TAS1R3) and its downstream molecule TRPM5 in duodenum and colon tissues of rats were quantified by WB. We examined GLP-1 secretion in enteroendocrine STC-1 cells exposured to 3DG. RESULTS 3DG treatment for 2 weeks increased 3DG content of intestinal tissues, fasting blood glucose concentration, and reduced plasma concentrations of GLP-1 and insulin at fasting and 15 and 180 min after the glucose load and oral glucose tolerance in conjunction with increased plasma glucagon concentrations. The expressions of TAS1R2, TAS1R3 and TRPM5 were shown to be reduced whereas 3DG treatment did not affect plasma dipeptidyl peptidase-4 activity, indicating an impaired GLP-1 secretion in 3DG-treated rats. This idea was further supported by the fact that exposure to 3DG directly decrease GLP-1 secretion in STC-1. CONCLUSION It is the first demonstration that 3DG was capable of accumulating in intestinal tissue and thereby decreased secretion of GLP-1 and insulin in a similar manner. 3DG-treated rats developed impaired glucose regulation (IGR) with obviously pancreatic islet cell dysfunction. It is further concluded that a decrease in the biological function of GLP-1 resulting from the decreased GLP-1 secretion is the most likely mechanism for the impaired insulin secretion, which ultimately promoted the development of IGR. These results will also contribute to a better understanding of the significance for restoring physiological GLP-1 secretion.
Collapse
Affiliation(s)
- Lurong Zhang
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, No. 18, Yangsu Road, Suzhou, 215003 Jiangsu People’s Republic of China
| | - Xiudao Song
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, No. 18, Yangsu Road, Suzhou, 215003 Jiangsu People’s Republic of China
| | - Liang Zhou
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, No. 18, Yangsu Road, Suzhou, 215003 Jiangsu People’s Republic of China
| | - Guoqiang Liang
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, No. 18, Yangsu Road, Suzhou, 215003 Jiangsu People’s Republic of China
| | - Heng Xu
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, No. 18, Yangsu Road, Suzhou, 215003 Jiangsu People’s Republic of China
| | - Fei Wang
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, No. 18, Yangsu Road, Suzhou, 215003 Jiangsu People’s Republic of China
| | - Fei Huang
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, No. 18, Yangsu Road, Suzhou, 215003 Jiangsu People’s Republic of China
| | - Guorong Jiang
- Suzhou Academy of Wumen Chinese Medicine, Suzhou Hospital of Traditional Chinese Medicine, No. 18, Yangsu Road, Suzhou, 215003 Jiangsu People’s Republic of China
| |
Collapse
|
91
|
Finan B, Clemmensen C, Müller TD. Emerging opportunities for the treatment of metabolic diseases: Glucagon-like peptide-1 based multi-agonists. Mol Cell Endocrinol 2015; 418 Pt 1:42-54. [PMID: 26151488 DOI: 10.1016/j.mce.2015.07.003] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 06/24/2015] [Accepted: 07/02/2015] [Indexed: 12/18/2022]
Abstract
Obesity is a pathogenic gateway to the metabolic syndrome and the complications thereof, thus interventions aimed at preventing or reversing the metabolic derangements underlying obesity hold great therapeutic promise. However, the complexity of energy balance regulation, combined with the heterologous pathophysiology of human obesity, renders effective medicinal intervention very difficult. Indeed, the search for the silver bullet in anti-obesity medicines has been laden with drugs of underwhelming efficacy and unacceptable side effects. This can partly be the consequence that many of these drug interventions have been historically directed at single molecular targets. New multi-molecular combination therapies have shown promising clinical outcomes in terms of weight loss, yet multi-functional single molecules may offer even more advantages than adjunctive co-treatments. Single molecules with integrated activities derived from multiple hormones involved in the physiological control of metabolism have emerged as one of the more promising candidates for reversing obesity. The inclusion of glucagon-like peptide-1 (GLP-1) as one of the constituents is a unifying factor amongst the majority of these unimolecular multi-agonists. The scope of this review is to summarize the current preclinical and clinical landscape of GLP-1-based therapies, focusing on combinatorial therapies with a particular emphasis on single molecule compounds displaying multi-agonist properties.
Collapse
Affiliation(s)
- Brian Finan
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany.
| | - Christoffer Clemmensen
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| |
Collapse
|
92
|
Baden MY, Fukui K, Hosokawa Y, Iwahashi H, Imagawa A, Shimomura I. Examination of a Viral Infection Mimetic Model in Human iPS Cell-Derived Insulin-Producing Cells and the Anti-Apoptotic Effect of GLP-1 Analogue. PLoS One 2015; 10:e0144606. [PMID: 26659307 PMCID: PMC4676675 DOI: 10.1371/journal.pone.0144606] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/21/2015] [Indexed: 12/22/2022] Open
Abstract
Aims Viral infection is associated with pancreatic beta cell destruction in fulminant type 1 diabetes mellitus. The aim of this study was to investigate the acceleration and protective mechanisms of beta cell destruction by establishing a model of viral infection in pancreatic beta cells. Methods Polyinosinic:polycytidylic acid was transfected into MIN6 cells and insulin-producing cells differentiated from human induced pluripotent stem cells via small molecule applications. Gene expression was analyzed by real-time PCR, and apoptosis was evaluated by caspase-3 activity and TUNEL staining. The anti-apoptotic effect of Exendin-4 was also evaluated. Results Polyinosinic:polycytidylic acid transfection led to elevated expression of the genes encoding IFNα, IFNβ, CXCL10, Fas, viral receptors, and IFN-inducible antiviral effectors in MIN6 cells. Exendin-4 treatment suppressed the elevated gene expression levels and reduced polyinosinic:polycytidylic acid-induced apoptosis both in MIN6 cells and in insulin-producing cells from human induced pluripotent stem cells. Glucagon-like peptide-1 receptor, protein kinase A, and phosphatidylinositol-3 kinase inhibitors counteracted the anti-apoptotic effect of Exendin-4. Conclusions Polyinosinic:polycytidylic acid transfection can mimic viral infection, and Exendin-4 exerted an anti-apoptotic effect both in MIN6 and insulin-producing cells from human induced pluripotent stem cells.
Collapse
Affiliation(s)
- Megu Yamaguchi Baden
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kenji Fukui
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yoshiya Hosokawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Hiromi Iwahashi
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Akihisa Imagawa
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
- * E-mail:
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
93
|
Campbell JE, Ussher JR, Mulvihill EE, Kolic J, Baggio LL, Cao X, Liu Y, Lamont BJ, Morii T, Streutker CJ, Tamarina N, Philipson LH, Wrana JL, MacDonald PE, Drucker DJ. TCF1 links GIPR signaling to the control of beta cell function and survival. Nat Med 2015; 22:84-90. [DOI: 10.1038/nm.3997] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/26/2015] [Indexed: 12/18/2022]
|
94
|
El Batsh MM, El Batch MM, Shafik NM, Younos IH. Favorable effects of vildagliptin on metabolic and cognitive dysfunctions in streptozotocin-induced diabetic rats. Eur J Pharmacol 2015; 769:297-305. [PMID: 26607467 DOI: 10.1016/j.ejphar.2015.11.033] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 11/16/2015] [Accepted: 11/18/2015] [Indexed: 02/08/2023]
Abstract
Progression of diabetes mellitus is accompanied by metabolic disorders together with psychological deficits including cognitive dysfunctions. Herein, we used a murine streptozotocin (STZ)-induced diabetes to investigate the beneficial effects of vildagliptin not only on metabolic abnormalities, but also on diabetes-induced cognitive decline. Sixty rats were divided randomly and equally into 2 groups; one remains normal and the other serves as STZ- induced diabetic. Both groups were further divided equally into 2 groups; one received vehicle and the other received oral vildagliptin for 8 weeks. Cognitive behavior was assessed using novel object recognition test. Blood samples were collected to measure metabolic parameters and dipeptidyl peptidase (DPP)-IV activity. Brains were removed and investigated for the levels of inflammatory and oxidative stress markers malondialdehyde (MDA), superoxide dismutase (SOD) and tumor necrosis factor-α (TNF-α), in addition to brain-derived neurotrophic factor (BDNF) and relative expression of nuclear factor kappa B (NF-κB)/p65. Treatment of STZ-induced diabetic rats with vildagliptin increased their body weight and corrected diabetes-induced memory and learning impairment. Moreover, vildagliptin significantly decreased serum levels of glucose and lipids (except high density lipoprotein) together with brain MDA, TNF-α, serum DPP-IV activities and NF-κB/p65 gene expression. On the other hand, vildagliptin significantly increased brain BDNF, SOD as well as serum insulin. Results suggested that vildagliptin has a protective role in counteracting both metabolic abnormalities and memory deficits in diabetic rats, possibly via its anti-hyperglycemic, anti-inflammatory, antioxidant effects, together with reduction of brain NF-κB/p65 over expression.
Collapse
Affiliation(s)
- Maha M El Batsh
- Department of Clinical Pharmacology, Faculty of Medicine, Menoufia University, Egypt
| | - Manal M El Batch
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Egypt
| | - Noha M Shafik
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Egypt.
| | - Ibrahim H Younos
- Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Sciences, Sultan Qaboos University, Oman
| |
Collapse
|
95
|
Lavine JA, Kibbe CR, Baan M, Sirinvaravong S, Umhoefer HM, Engler KA, Meske LM, Sacotte KA, Erhardt DP, Davis DB. Cholecystokinin expression in the β-cell leads to increased β-cell area in aged mice and protects from streptozotocin-induced diabetes and apoptosis. Am J Physiol Endocrinol Metab 2015; 309:E819-28. [PMID: 26394663 PMCID: PMC4652070 DOI: 10.1152/ajpendo.00159.2015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 09/15/2015] [Indexed: 12/23/2022]
Abstract
Cholecystokinin (CCK) is a peptide hormone produced in the gut and brain with beneficial effects on digestion, satiety, and insulin secretion. CCK is also expressed in pancreatic β-cells, but only in models of obesity and insulin resistance. Whole body deletion of CCK in obese mice leads to reduced β-cell mass expansion and increased apoptosis. We hypothesized that islet-derived CCK is important in protection from β-cell apoptosis. To determine the specific role of β-cell-derived CCK in β-cell mass dynamics, we generated a transgenic mouse that expresses CCK in the β-cell in the lean state (MIP-CCK). Although this transgene contains the human growth hormone minigene, we saw no expression of human growth hormone protein in transgenic islets. We examined the ability of MIP-CCK mice to maintain β-cell mass when subjected to apoptotic stress, with advanced age, and after streptozotocin treatment. Aged MIP-CCK mice have increased β-cell area. MIP-CCK mice are resistant to streptozotocin-induced diabetes and exhibit reduced β-cell apoptosis. Directed CCK overexpression in cultured β-cells also protects from cytokine-induced apoptosis. We have identified an important new paracrine/autocrine effect of CCK in protection of β-cells from apoptotic stress. Understanding the role of β-cell CCK adds to the emerging knowledge of classic gut peptides in intraislet signaling. CCK receptor agonists are being investigated as therapeutics for obesity and diabetes. While these agonists clearly have beneficial effects on body weight and insulin sensitivity in peripheral tissues, they may also directly protect β-cells from apoptosis.
Collapse
Affiliation(s)
- Jeremy A Lavine
- School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Carly R Kibbe
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Mieke Baan
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Sirinart Sirinvaravong
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Heidi M Umhoefer
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kimberly A Engler
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Louise M Meske
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kaitlyn A Sacotte
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Daniel P Erhardt
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Dawn Belt Davis
- Department of Medicine, Division of Endocrinology, University of Wisconsin-Madison, Madison, Wisconsin; Geriatric Research Education and Clinical Centers, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
| |
Collapse
|
96
|
Beneficial effects of growth hormone-releasing hormone agonists on rat INS-1 cells and on streptozotocin-induced NOD/SCID mice. Proc Natl Acad Sci U S A 2015; 112:13651-6. [PMID: 26474831 DOI: 10.1073/pnas.1518540112] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Agonists of growth hormone-releasing hormone (GHRH) have been previously reported to promote growth, function, and engraftment of islet cells following transplantation. Here we evaluated recently synthesized GHRH agonists on the proliferation and biological functions of rat pancreatic β-cell line (INS-1) and islets. In vitro treatment of INS-1 cells with GHRH agonists increased cell proliferation, the expression of cellular insulin, insulin-like growth factor-1 (IGF1), and GHRH receptor, and also stimulated insulin secretion in response to glucose challenge. Exposure of INS-1 cells to GHRH agonists, MR-356 and MR-409, induced activation of ERK and AKT pathways. Agonist MR-409 also significantly increased the levels of cellular cAMP and the phosphorylation of cAMP response element binding protein (CREB) in INS-1 cells. Treatment of rat islets with agonist, MR-409 significantly increased cell proliferation, islet size, and the expression of insulin. In vivo daily s.c. administration of 10 μg MR-409 for 3 wk dramatically reduced the severity of streptozotocin (STZ)-induced diabetes in nonobese diabetic severe combined immunodeficiency (NOD/SCID) mice. The maximal therapeutic benefits with respect to the efficiency of engraftment, ability to reach normoglycemia, gain in body weight, response to high glucose challenge, and induction of higher levels of serum insulin and IGF1 were observed when diabetic mice were transplanted with rat islets preconditioned with GHRH agonist, MR-409, and received additional treatment with MR-409 posttransplantation. This study provides an improved approach to the therapeutic use of GHRH agonists in the treatment of diabetes mellitus.
Collapse
|
97
|
Carlessi R, Lemos NE, Dias AL, Oliveira FS, Brondani LA, Canani LH, Bauer AC, Leitão CB, Crispim D. Exendin-4 protects rat islets against loss of viability and function induced by brain death. Mol Cell Endocrinol 2015; 412:239-50. [PMID: 25976662 DOI: 10.1016/j.mce.2015.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 05/07/2015] [Accepted: 05/07/2015] [Indexed: 02/01/2023]
Abstract
Islet quality loss after isolation from brain-dead donors still hinders the implementation of human islet transplantation for treatment of type 1 diabetes. In this scenario, systemic inflammation elicited by donor brain death (BD) is among the main factors influencing islet viability and functional impairment. Exendin-4 is largely recognized to promote anti-inflammatory and cytoprotective effects on β-cells. Therefore, we hypothesized that administration of exendin-4 to brain-dead donors might improve islet survival and insulin secretory capabilities. Here, using a rat model of BD, we demonstrate that exendin-4 administration to the brain-dead donors increases both islet viability and glucose-stimulated insulin secretion. In this model, exendin-4 treatment produced a significant decrease in interleukin-1β expression in the pancreas. Furthermore, exendin-4 treatment increased the expression of superoxide dismutase-2 and prevented BD-induced elevation in uncoupling protein-2 expression. Such observations were accompanied by a reduction in gene expression of two genes often associated with endoplasmic reticulum (ER) stress response in freshly isolated islets from treated animals, C/EBP homologous protein and immunoglobulin heavy-chain binding protein. As ER stress response has been shown to be triggered by and to participate in cytokine-induced β-cell death, we suggest that exendin-4 might exert its beneficial effects through alleviation of pancreatic inflammation and oxidative stress, which in turn could prevent islet ER stress and β-cell death. Our findings might unveil a novel strategy to preserve islet quality from brain-dead donors. After testing in the human pancreatic islet transplantation setting, this approach might sum to the ongoing effort to achieve consistent and successful single-donor islet transplantation.
Collapse
Affiliation(s)
- Rodrigo Carlessi
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; School of Biomedical Sciences, CHIRI - Biosciences, Curtin University, Perth, Western Australia 6845, Australia
| | - Natália E Lemos
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Ana L Dias
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Fernanda S Oliveira
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Letícia A Brondani
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Luis H Canani
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andrea C Bauer
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Cristiane B Leitão
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Daisy Crispim
- Laboratory of Human Pancreatic Islet Biology, Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-Graduate Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
98
|
Nassar NN, Al-Shorbagy MY, Arab HH, Abdallah DM. Saxagliptin: a novel antiparkinsonian approach. Neuropharmacology 2015; 89:308-17. [PMID: 25446674 DOI: 10.1016/j.neuropharm.2014.10.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 09/03/2014] [Accepted: 10/06/2014] [Indexed: 01/08/2023]
Abstract
The emergence of glucagon-like peptide-1 as a crucial contender in modifying neurodegenerative diseases in the preclinical studies has instigated interest in investigating the antiparkinsonian effect of dipeptidyl peptidase (DPP)-4 inhibition. Notably, saxagliptin (SAX), the DPP-4 inhibitor, recently showed efficacy in ameliorating streptozotocin-induced Alzheimer's disease; however, its effect on Parkinson's disease (PD) has not yet been elucidated. In a rat rotenone (ROT) model, SAX prominently improved motor performance as well as muscle coordination and corrected akinesia. Moreover, SAX preserved substantia nigra pars compacta tyrosine hydroxylase (TH) immunoreactivity while halting the reduction in the striatal TH, dopamine (DA) and complex I. Meanwhile, SAX prevented the ROT-induced increment of striatal DPP-4 and the decline in cAMP, ATP/ADP and brain-derived neurotropic factor levels. Improvement in striatal energy level was associated with partial hindrance of ROT-induced body weight reduction. In addition, through its anti-inflammatory potential, SAX decreased the ROT-induced nuclear factor-κΒ, inducible nitric oxide synthase, tumor necrosis factor-α, intracellular adhesion molecule-1 and myeloperoxidase. The antiapoptotic marker B-cell lymphoma-2 was enhanced by SAX, versus reduction in caspase-3 and its intrinsic apoptotic activator cytochrome C. Furthermore, SAX amended alterations induced by ROT in the thiobarbituric acid reactive substances and the transcriptional factor Nrf-2 level. In conclusion, SAX can be introduced as a novel approach for the management of PD based on the remarkable improvement in motor functions denoting antiparkinsonian efficacy via antioxidant, anti-inflammatory, antiapoptotic, neuroprotective and neurorestorative mechanisms. These effects were linked to DPP-4 inhibition, reduced neurodegeneration and enhanced DA synthesis.
Collapse
Affiliation(s)
- Noha N Nassar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | | | | | | |
Collapse
|
99
|
Asakawa M, Mitsui H, Akihisa M, Sekine T, Niitsu Y, Kobayashi A, Miyake A, Hashimoto N, Kawamura M, Ogawa Y. Efficacy and safety of sitagliptin for the treatment of diabetes mellitus complicated by chronic liver injury. SPRINGERPLUS 2015; 4:346. [PMID: 26191473 PMCID: PMC4501339 DOI: 10.1186/s40064-015-1135-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 06/17/2015] [Indexed: 01/10/2023]
Abstract
Aim To investigate the efficacy and safety of a dipeptidyl peptidase-4 inhibitor, sitagliptin, for treating diabetes mellitus complicated by chronic liver injury. Methods Sitagliptin was administered for 13.7 ± 10.1 months to 122 patients with DM complicated by chronic liver injury (including 19 patients with liver cirrhosis), and changes in hemoglobin A1c (HbA1c) and liver enzymes (transaminases, etc.) were evaluated. Results HbA1c was reduced from 8.48 ± 1.43% to 7.87 ± 1.35% (P < 0.001). Among liver enzymes, alanine aminotransferase (ALT) levels improved from 75.1 ± 45.2 to 65.8 ± 35.8 IU/L (P = 0.012) and gamma-glut amyl-trans peptidase from 155.2 ± 161.1 to 133.2 ± 127.4 IU/L (P = 0.044). Among the causes of liver injury, non-alcoholic fatty liver disease and alcoholic liver disease both showed the reductions in HbA1c with no deterioration of liver enzymes. An analysis of 19 patients with liver cirrhosis also showed reductions in HbA1c with no deterioration of liver enzymes. Conclusion It is suggested that sitagliptin can be administered effectively and safely to patients with diabetes mellitus complicated by chronic liver injury, including liver cirrhosis.
Collapse
Affiliation(s)
- Masahiro Asakawa
- Department of Endocrinology and Metabolism, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, 102-0071 Japan
| | - Hiroshi Mitsui
- Department of Gastroenterology, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, 102-0071 Japan
| | - Momoko Akihisa
- Department of Molecular Endocrinology and Metabolism, Tokyo Medical and Dental University Hospital, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| | - Tetsuo Sekine
- Department of Endocrinology and Metabolism, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, 102-0071 Japan
| | - Yoshihiro Niitsu
- Department of Endocrinology and Metabolism, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, 102-0071 Japan
| | - Arisa Kobayashi
- Department of Endocrinology and Metabolism, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, 102-0071 Japan
| | - Atsuko Miyake
- Department of Endocrinology and Metabolism, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, 102-0071 Japan
| | - Naoaki Hashimoto
- Department of Gastroenterology, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, 102-0071 Japan
| | - Mitsunobu Kawamura
- Department of Endocrinology and Metabolism, Tokyo Teishin Hospital, 2-14-23, Fujimi, Chiyoda-ku, Tokyo, 102-0071 Japan
| | - Yoshihiro Ogawa
- Department of Molecular Endocrinology and Metabolism, Tokyo Medical and Dental University Hospital, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510 Japan
| |
Collapse
|
100
|
Kaku K, Kiyosue A, Ono Y, Shiraiwa T, Kaneko S, Nishijima K, Bosch-Traberg H, Seino Y. Liraglutide is effective and well tolerated in combination with an oral antidiabetic drug in Japanese patients with type 2 diabetes: A randomized, 52-week, open-label, parallel-group trial. J Diabetes Investig 2015; 7:76-84. [PMID: 26816604 PMCID: PMC4718097 DOI: 10.1111/jdi.12367] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 04/07/2015] [Accepted: 04/20/2015] [Indexed: 12/23/2022] Open
Abstract
Introduction The safety and efficacy of liraglutide in combination with an oral antidiabetic drug (OAD) compared with combination of two OADs were assessed in Japanese patients with type 2 diabetes. Materials and Methods This was a 52‐week, open‐label, parallel‐group trial in which patients whose type 2 diabetes was inadequately controlled with a single OAD (glinide, metformin, α‐glucosidase inhibitor or thiazolidinedione) were randomized 2:1 to either pretrial OAD in combination with liraglutide 0.9 mg/day (liraglutide group; n = 240) or pretrial OAD in combination with an additional OAD (additional OAD group; n = 120). The primary outcome measure was the incidence of adverse events (AEs). Results Overall, 86.3% of patients in the liraglutide group and 85.0% of patients in the additional OAD group experienced AEs; these were similar in nature and severity. Adverse event rates were 361 and 331 per 100 patient‐years of exposure, respectively. Confirmed hypoglycemia was rare (seven episodes in two patients on liraglutide, and two in two patients on additional OAD). There were no reported pancreatitis events, and no unexpected safety signals were identified. Mean reductions in glycosylated hemoglobin were significantly greater in the liraglutide group than the additional OAD group [estimated mean treatment difference −0.27% (95% confidence interval (CI) −0.44, −0.09; P = 0.0026)]; reductions in mean fasting plasma glucose levels were also greater with liraglutide [estimated mean difference −5.47 mg/dL (−0.30 mmol/L; 95% CI: −10.83, −0.10; P = 0.0458)]. Conclusions Liraglutide was well tolerated and effective as combination therapy with an OAD in Japanese patients with type 2 diabetes.
Collapse
Affiliation(s)
- Kohei Kaku
- Department of Internal Medicine Kawasaki Medical School Okayama Japan
| | | | | | | | | | - Keiji Nishijima
- Medical & Scientific Affairs Department Novo Nordisk Pharma Ltd Tokyo Japan
| | | | | |
Collapse
|