301
|
Li Y, Li L, Hölscher C. Incretin-based therapy for type 2 diabetes mellitus is promising for treating neurodegenerative diseases. Rev Neurosci 2018; 27:689-711. [PMID: 27276528 DOI: 10.1515/revneuro-2016-0018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/02/2016] [Indexed: 12/13/2022]
Abstract
Incretin hormones include glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). Due to their promising action on insulinotropic secretion and improving insulin resistance (IR), incretin-based therapies have become a new class of antidiabetic agents for the treatment of type 2 diabetes mellitus (T2DM). Recently, the links between neurodegenerative diseases and T2DM have been identified in a number of studies, which suggested that shared mechanisms, such as insulin dysregulation or IR, may underlie these conditions. Therefore, the effects of incretins in neurodegenerative diseases have been extensively investigated. Protease-resistant long-lasting GLP-1 mimetics such as lixisenatide, liraglutide, and exenatide not only have demonstrated promising effects for treating neurodegenerative diseases in preclinical studies but also have shown first positive results in Alzheimer's disease (AD) and Parkinson's disease (PD) patients in clinical trials. Furthermore, the effects of other related incretin-based therapies such as GIP agonists, dipeptidyl peptidase-IV (DPP-IV) inhibitors, oxyntomodulin (OXM), dual GLP-1/GIP, and triple GLP-1/GIP/glucagon receptor agonists on neurodegenerative diseases have been tested in preclinical studies. Incretin-based therapies are a promising approach for treating neurodegenerative diseases.
Collapse
|
302
|
Liu Y, Li Y, Gong Y, Yu N, Zhang Y, You R, Qu C, Lu G, Huang Y, Gao Y, Gao Y, Guo X. CD26 expression is down-regulated on CD8+ T cells in patients with Hashimoto's thyroiditis. Int Immunopharmacol 2018; 54:280-285. [DOI: 10.1016/j.intimp.2017.11.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 12/12/2022]
|
303
|
Upadhyay J, Polyzos SA, Perakakis N, Thakkar B, Paschou SA, Katsiki N, Underwood P, Park KH, Seufert J, Kang ES, Sternthal E, Karagiannis A, Mantzoros CS. Pharmacotherapy of type 2 diabetes: An update. Metabolism 2018; 78:13-42. [PMID: 28920861 DOI: 10.1016/j.metabol.2017.08.010] [Citation(s) in RCA: 139] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 08/24/2017] [Accepted: 08/26/2017] [Indexed: 12/22/2022]
Abstract
Type 2 diabetes (T2DM) is a leading cause of morbidity and mortality worldwide and a major economic burden. The prevalence of T2DM is rising, suggesting more effective prevention and treatment strategies are necessary. The aim of this narrative review is to summarize the pharmacologic treatment options available for patients with T2DM. Each therapeutic class is presented in detail, outlining medication effects, side effects, glycemic control, effect on weight, indications and contraindications, and use in selected populations (heart failure, renal insufficiency, obesity and the elderly). We also present representative cost for each antidiabetic category. Then, we provide an individualized guide for initiation and intensification of treatment and discuss the considerations and rationale for an individualized glycemic goal.
Collapse
Affiliation(s)
- Jagriti Upadhyay
- Section of Endocrinology, Diabetes and Metabolism, Boston VA Healthcare System, Boston, MA, USA; Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Stergios A Polyzos
- First Department of Pharmacology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Perakakis
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Divisions of Endocrinology and Diabetology, Department of Internal Medicine II, University Hospital of Freiburg, Freiburg, Germany
| | - Bindiya Thakkar
- Section of Endocrinology, Diabetes and Metabolism, Boston VA Healthcare System, Boston, MA, USA
| | - Stavroula A Paschou
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Niki Katsiki
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippocration Hospital, Thessaloniki, Greece
| | - Patricia Underwood
- Section of Endocrinology, Diabetes and Metabolism, Boston VA Healthcare System, Boston, MA, USA
| | - Kyung-Hee Park
- Department of Family Medicine, Hallym University Sacred Heart Hospital, Gyeonggi-do, Republic of Korea
| | - Jochen Seufert
- Divisions of Endocrinology and Diabetology, Department of Internal Medicine II, University Hospital of Freiburg, Freiburg, Germany
| | - Eun Seok Kang
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Elliot Sternthal
- Section of Endocrinology, Diabetes and Metabolism, Boston VA Healthcare System, Boston, MA, USA
| | - Asterios Karagiannis
- First Department of Pharmacology, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christos S Mantzoros
- Section of Endocrinology, Diabetes and Metabolism, Boston VA Healthcare System, Boston, MA, USA; Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
304
|
Dietert K, Gutbier B, Wienhold SM, Reppe K, Jiang X, Yao L, Chaput C, Naujoks J, Brack M, Kupke A, Peteranderl C, Becker S, von Lachner C, Baal N, Slevogt H, Hocke AC, Witzenrath M, Opitz B, Herold S, Hackstein H, Sander LE, Suttorp N, Gruber AD. Spectrum of pathogen- and model-specific histopathologies in mouse models of acute pneumonia. PLoS One 2017; 12:e0188251. [PMID: 29155867 PMCID: PMC5695780 DOI: 10.1371/journal.pone.0188251] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/05/2017] [Indexed: 01/03/2023] Open
Abstract
Pneumonia may be caused by a wide range of pathogens and is considered the most common infectious cause of death in humans. Murine acute lung infection models mirror human pathologies in many aspects and contribute to our understanding of the disease and the development of novel treatment strategies. Despite progress in other fields of tissue imaging, histopathology remains the most conclusive and practical read out tool for the descriptive and semiquantitative evaluation of mouse pneumonia and therapeutic interventions. Here, we systematically describe and compare the distinctive histopathological features of established models of acute pneumonia in mice induced by Streptococcus (S.) pneumoniae, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Legionella pneumophila, Escherichia coli, Middle East respiratory syndrome (MERS) coronavirus, influenza A virus (IAV) and superinfection of IAV-incuced pneumonia with S. pneumoniae. Systematic comparisons of the models revealed striking differences in the distribution of lesions, the characteristics of pneumonia induced, principal inflammatory cell types, lesions in adjacent tissues, and the detectability of the pathogens in histological sections. We therefore identified core criteria for each model suitable for practical semiquantitative scoring systems that take into account the pathogen- and model-specific patterns of pneumonia. Other critical factors that affect experimental pathologies are discussed, including infectious dose, time kinetics, and the genetic background of the mouse strain. The substantial differences between the model-specific pathologies underscore the necessity of pathogen- and model-adapted criteria for the comparative quantification of experimental outcomes. These criteria also allow for the standardized validation and comparison of treatment strategies in preclinical models.
Collapse
MESH Headings
- Acinetobacter baumannii/pathogenicity
- Acinetobacter baumannii/physiology
- Animals
- Disease Models, Animal
- Escherichia coli/pathogenicity
- Escherichia coli/physiology
- Female
- Host Specificity
- Humans
- Immunohistochemistry
- Influenza A virus/pathogenicity
- Influenza A virus/physiology
- Klebsiella pneumoniae/pathogenicity
- Klebsiella pneumoniae/physiology
- Legionella pneumophila/pathogenicity
- Legionella pneumophila/physiology
- Lung/microbiology
- Lung/pathology
- Lung/virology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Middle East Respiratory Syndrome Coronavirus/pathogenicity
- Middle East Respiratory Syndrome Coronavirus/physiology
- Pneumonia, Bacterial/genetics
- Pneumonia, Bacterial/microbiology
- Pneumonia, Bacterial/pathology
- Pneumonia, Bacterial/physiopathology
- Pneumonia, Viral/genetics
- Pneumonia, Viral/pathology
- Pneumonia, Viral/physiopathology
- Pneumonia, Viral/virology
- Species Specificity
- Staphylococcus aureus/pathogenicity
- Staphylococcus aureus/physiology
- Streptococcus pneumoniae/pathogenicity
- Streptococcus pneumoniae/physiology
Collapse
Affiliation(s)
- Kristina Dietert
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Birgitt Gutbier
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sandra M. Wienhold
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Katrin Reppe
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Xiaohui Jiang
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ling Yao
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Catherine Chaput
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jan Naujoks
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Markus Brack
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Alexandra Kupke
- Department of Internal Medicine II, Section for Infectious Diseases, Universities Giessen & Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) Giessen, Germany
- Institute of Virology, Philipps University of Marburg, German Center for Infection Research (DZIF), TTU Emerging Infections, Marburg, Germany
| | - Christin Peteranderl
- Department of Internal Medicine II, Section for Infectious Diseases, Universities Giessen & Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) Giessen, Germany
| | - Stephan Becker
- Department of Internal Medicine II, Section for Infectious Diseases, Universities Giessen & Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) Giessen, Germany
- Institute of Virology, Philipps University of Marburg, German Center for Infection Research (DZIF), TTU Emerging Infections, Marburg, Germany
| | | | - Nelli Baal
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Giessen, Germany
| | - Hortense Slevogt
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Andreas C. Hocke
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Martin Witzenrath
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Bastian Opitz
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Susanne Herold
- Department of Internal Medicine II, Section for Infectious Diseases, Universities Giessen & Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL) Giessen, Germany
| | - Holger Hackstein
- Institute for Clinical Immunology and Transfusion Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), University Hospital Giessen und Marburg, Justus-Liebig-University Giessen, Giessen, Germany
| | - Leif E. Sander
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases and Pulmonary Medicine, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Achim D. Gruber
- Department of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
305
|
Esposito G, Cappetta D, Russo R, Rivellino A, Ciuffreda LP, Roviezzo F, Piegari E, Berrino L, Rossi F, De Angelis A, Urbanek K. Sitagliptin reduces inflammation, fibrosis and preserves diastolic function in a rat model of heart failure with preserved ejection fraction. Br J Pharmacol 2017; 174:4070-4086. [PMID: 27922176 PMCID: PMC5659996 DOI: 10.1111/bph.13686] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 11/07/2016] [Accepted: 11/29/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Heart failure with preserved ejection fraction (HFpEF) is a systemic syndrome driven by co-morbidities, and its pathophysiology is poorly understood. Several studies suggesting that dipeptidyl peptidase 4 (DPP4) might be involved in the pathophysiology of heart failure have prompted experimental and clinical investigations of DPP4 inhibitors in the cardiovascular system. Here we have investigated whether the DPP4 inhibitor sitagliptin affected the progression of HFpEF independently of its effects on glycaemia. EXPERIMENTAL APPROACH Seven-week-old Dahl salt-sensitive rats were fed a high-salt diet for 5 weeks to induce hypertension. Then the rats continued with the high-salt diet and were treated with either sitagliptin (10 mg·kg-1 ) or vehicle for the following 8 weeks. Blood pressure and cardiac function were measured in vivo. Histochemical and molecular biology analyses of myocardium were used to assay cytokines, fibrotic markers, DPP4 and glucagon-like peptide-1 (GLP-1)/GLP-1 receptor. KEY RESULTS Treatment with sitagliptin attenuated diastolic dysfunction, reduced mortality and reduced cardiac DPP4 activity, along with increased circulating GLP-1 and myocardial expression of GLP-1 receptors. Myocardial levels of pro-inflammatory cytokines (TNF-α, IL-6 and CCL2) were reduced. Sitagliptin treatment decreased the levels of endothelial NOS monomer, responsible for generation of ROS, while the amount of NO-producing dimeric form increased. Markers of oxidative and nitrosative stress were decreased. Moreover, increased collagen deposition and activation of pro-fibrotic signalling, inducing elevated myocardial stiffness, were attenuated by sitagliptin treatment. CONCLUSIONS AND IMPLICATIONS Sitagliptin positively modulated active relaxation and passive diastolic compliance by decreasing inflammation-related endothelial dysfunction and fibrosis, associated with HFpEF. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Grazia Esposito
- Department of Experimental Medicine, Section of PharmacologyUnivesity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Donato Cappetta
- Department of Experimental Medicine, Section of PharmacologyUnivesity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Rosa Russo
- Department of Experimental Medicine, Section of PharmacologyUnivesity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Alessia Rivellino
- Department of Experimental Medicine, Section of PharmacologyUnivesity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Loreta Pia Ciuffreda
- Department of Experimental Medicine, Section of PharmacologyUnivesity of Campania “Luigi Vanvitelli”NaplesItaly
| | | | - Elena Piegari
- Department of Experimental Medicine, Section of PharmacologyUnivesity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Liberato Berrino
- Department of Experimental Medicine, Section of PharmacologyUnivesity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Francesco Rossi
- Department of Experimental Medicine, Section of PharmacologyUnivesity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of PharmacologyUnivesity of Campania “Luigi Vanvitelli”NaplesItaly
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of PharmacologyUnivesity of Campania “Luigi Vanvitelli”NaplesItaly
| |
Collapse
|
306
|
Cordero OJ, Varela-Calviño R, López-González T, Grujic M, Juranic Z, Mouriño C, Hernández-Rodríguez Í, Rodríguez-López M, de la Iglesia BA, Pego-Reigosa JM. Anti-CD26 autoantibodies are involved in rheumatoid arthritis and show potential clinical interest. Clin Biochem 2017; 50:903-910. [DOI: 10.1016/j.clinbiochem.2017.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/19/2017] [Accepted: 06/05/2017] [Indexed: 11/25/2022]
|
307
|
Deng X, Han L, Zhou J, Zhang H, Li Q. Discovery of triazole-based uracil derivatives bearing amide moieties as novel dipeptidyl peptidase-IV inhibitors. Bioorg Chem 2017; 75:357-367. [PMID: 29096096 DOI: 10.1016/j.bioorg.2017.10.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/12/2017] [Accepted: 10/22/2017] [Indexed: 01/02/2023]
Abstract
Dipeptidyl peptidase-IV (DPP-4) is a validated target for T2DM treatment. We previously reported a novel series of triazole-based uracil derivatives bearing aliphatic carboxylic acids with potent DPP-4 inhibitory activities in vitro, but these compounds showed poor hypoglycemic effects in vivo. Herein we further optimized the triazole moiety by amidation of the carboxylic acid to improve in vivo activities. Two series of compounds 3a-f and 4a-g were designed and synthesized. By screening in DPP-4, compound 4c was identified as a potent DPP-4 inhibitor with the IC50 value of 28.62 nM. Docking study revealed compound 4c has a favorable binding mode and interpreted the SAR of these analogs. DPP-8 and DPP-9 tests indicated compound 4c had excellent selectivity over DPP-8 and DPP-9. Further in vivo evaluations revealed that compound 4c showed more potent hypoglycemic activity than its corresponding carboxylic acid in ICR mice and dose-dependently reduced glucose levels in type 2 diabetic C57BL/6 mice. The overall results have shown that compound 4c could be a promising lead for further development of novel DPP-4 agents treating T2DM.
Collapse
Affiliation(s)
- Xiaoyan Deng
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, PR China
| | - Li Han
- Center of Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing 210009, PR China
| | - Jinpei Zhou
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjia Xiang, Nanjing 210009, PR China
| | - Huibin Zhang
- Center of Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing 210009, PR China
| | - Qing Li
- Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi, PR China; Center of Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing 210009, PR China.
| |
Collapse
|
308
|
Baumeier C, Schlüter L, Saussenthaler S, Laeger T, Rödiger M, Alaze SA, Fritsche L, Häring HU, Stefan N, Fritsche A, Schwenk RW, Schürmann A. Elevated hepatic DPP4 activity promotes insulin resistance and non-alcoholic fatty liver disease. Mol Metab 2017; 6:1254-1263. [PMID: 29031724 PMCID: PMC5641684 DOI: 10.1016/j.molmet.2017.07.016] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 07/26/2017] [Accepted: 07/31/2017] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE Increased hepatic expression of dipeptidyl peptidase 4 (DPP4) is associated with non-alcoholic fatty liver disease (NAFLD). Whether this is causative for the development of NAFLD is not yet clarified. Here we investigate the effect of hepatic DPP4 overexpression on the development of liver steatosis in a mouse model of diet-induced obesity. METHODS Plasma DPP4 activity of subjects with or without NAFLD was analyzed. Wild-type (WT) and liver-specific Dpp4 transgenic mice (Dpp4-Liv-Tg) were fed a high-fat diet and characterized for body weight, body composition, hepatic fat content and insulin sensitivity. In vitro experiments on HepG2 cells and primary mouse hepatocytes were conducted to validate cell autonomous effects of DPP4 on lipid storage and insulin sensitivity. RESULTS Subjects suffering from insulin resistance and NAFLD show an increased plasma DPP4 activity when compared to healthy controls. Analysis of Dpp4-Liv-Tg mice revealed elevated systemic DPP4 activity and diminished active GLP-1 levels. They furthermore show increased body weight, fat mass, adipose tissue inflammation, hepatic steatosis, liver damage and hypercholesterolemia. These effects were accompanied by increased expression of PPARγ and CD36 as well as severe insulin resistance in the liver. In agreement, treatment of HepG2 cells and primary hepatocytes with physiological concentrations of DPP4 resulted in impaired insulin sensitivity independent of lipid content. CONCLUSIONS Our results give evidence that elevated expression of DPP4 in the liver promotes NAFLD and insulin resistance. This is linked to reduced levels of active GLP-1, but also to auto- and paracrine effects of DPP4 on hepatic insulin signaling.
Collapse
Affiliation(s)
- Christian Baumeier
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, Potsdam-Rehbruecke, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Luisa Schlüter
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, Potsdam-Rehbruecke, Germany
| | - Sophie Saussenthaler
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, Potsdam-Rehbruecke, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Thomas Laeger
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, Potsdam-Rehbruecke, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Maria Rödiger
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, Potsdam-Rehbruecke, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Stella Amelie Alaze
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, Potsdam-Rehbruecke, Germany
| | - Louise Fritsche
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Hans-Ulrich Häring
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Norbert Stefan
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Andreas Fritsche
- Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Robert Wolfgang Schwenk
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, Potsdam-Rehbruecke, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Annette Schürmann
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, Potsdam-Rehbruecke, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany.
| |
Collapse
|
309
|
Li Q, Wu X, Liu Y, Zhang M, Bai X, Chen C. The effect of anagliptin on intimal hyperplasia of rat carotid artery after balloon injury. Mol Med Rep 2017; 16:8003-8010. [PMID: 28990108 PMCID: PMC5779884 DOI: 10.3892/mmr.2017.7667] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 08/08/2017] [Indexed: 12/30/2022] Open
Abstract
The present study evaluated the effect of anagliptin on intimal hyperplasia following carotid artery injury in Sprague-Dawley rats. Sprague-Dawley rats weighing 280–300 g were injured using a 2F Fogarty balloon embolectomy catheter. The rats were divided into injury-(saline) and anagliptin-(10 mg/kg/day) treated groups. vascular injuries were induced in the left carotid artery, followed by evaluation of neointima formation at 28 days. The right and left carotid arteries were harvested and evaluated with histological evaluation, and the plasma activity of glucagon-like peptide 1 receptor (GLP-1), stromal cell-derived factor (SDF)-1α, interleukin (IL)-6, IL-1β and tumor necrosis factor (TNF)-α were detected by ELISA analysis. Treatment with anagliptin decreased balloon injury-induced neointima formation, compared with the injury group (P<0.01). Body weight and food consumption did not alter following treatment with anagliptin. Anagliptin caused an increase in the serum active GLP-1 concentration, compared with the injury group. In addition, serum SDF-1α was significantly decreased by treatment with anagliptin (P<0.001). Anagliptin altered the serum activity of IL-6, IL-1β and TNF-α (P<0.01). The results of the present study demonstrated that anagliptin appeared to attenuate neointimal formation by inhibiting inflammatory cytokines and chemokines following balloon injury, and that treatment with a dipeptidyl peptidase 4 inhibitor may be useful for future preclinical studies and potentially for the inhibition of thrombosis formation following percutaneous coronary intervention.
Collapse
Affiliation(s)
- Qi Li
- Biotherapy Center, Tumor Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiayang Wu
- Department of Pharmacology (State‑Province Key Laboratories of Biomedicine‑Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Yanli Liu
- Department of Pharmacology (State‑Province Key Laboratories of Biomedicine‑Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Mingyu Zhang
- Department of Pharmacology (State‑Province Key Laboratories of Biomedicine‑Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Xue Bai
- Department of Pharmacology (State‑Province Key Laboratories of Biomedicine‑Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| | - Chang Chen
- Department of Pharmacology (State‑Province Key Laboratories of Biomedicine‑Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
310
|
Lee ES, Lee DS, Pandeya PR, Kim YC, Kang DG, Lee HS, Oh BC, Lee DH. Sodium butyrate has context-dependent actions on dipeptidyl peptidase-4 and other metabolic parameters. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:519-529. [PMID: 28883756 PMCID: PMC5587602 DOI: 10.4196/kjpp.2017.21.5.519] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 05/29/2017] [Accepted: 06/20/2017] [Indexed: 12/28/2022]
Abstract
Sodium butyrate (SB) has various metabolic actions. However, its effect on dipeptidyl peptidase 4 (DPP-4) needs to be studied further. We aimed to evaluate the metabolic actions of SB, considering its physiologically relevant concentration. We evaluated the effect of SB on regulation of DPP-4 and its other metabolic actions, both in vitro (HepG2 cells and mouse mesangial cells) and in vivo (high fat diet [HFD]-induced obese mice). Ten-week HFD-induced obese C57BL/6J mice were subjected to SB treatment by adding SB to HFD which was maintained for an additional 16 weeks. In HepG2 cells, SB suppressed DPP-4 activity and expression at sub-molar concentrations, whereas it increased DPP-4 activity at a concentration of 1,000 µM. In HFD-induced obese mice, SB decreased blood glucose, serum levels of insulin and IL-1β, and DPP-4 activity, and suppressed the increase in body weight. On the contrary, various tissues including liver, kidney, and peripheral blood cells showed variable responses of DPP-4 to SB. Especially in the kidney, although DPP-4 activity was decreased by SB in HFD-induced obese mice, it caused an increase in mRNA expression of TNF-α, IL-6, and IL-1β. The pro-inflammatory actions of SB in the kidney of HFD-induced obese mice were recapitulated by cultured mesangial cell experiments, in which SB stimulated the secretion of several cytokines from cells. Our results showed that SB has differential actions according to its treatment dose and the type of cells and tissues. Thus, further studies are required to evaluate its therapeutic relevance in metabolic diseases including diabetes and obesity.
Collapse
Affiliation(s)
- Eun-Sol Lee
- College of Pharmacy, Wonkwang University, Iksan 54538, Korea
| | - Dong-Sung Lee
- College of Pharmacy, Chosun University, Gwangju 61452, Korea
| | | | - Youn-Chul Kim
- College of Pharmacy, Wonkwang University, Iksan 54538, Korea
| | - Dae-Gil Kang
- Hanbang Body Fluid Research Center, Wonkwang University, Iksan 54538, Korea.,College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, Korea
| | - Ho-Sub Lee
- Hanbang Body Fluid Research Center, Wonkwang University, Iksan 54538, Korea.,College of Oriental Medicine and Professional Graduate School of Oriental Medicine, Wonkwang University, Iksan 54538, Korea
| | - Byung-Chul Oh
- Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Korea.,Department of physiology, College of Medicine, Gachon University, Incheon 21999, Korea
| | - Dae Ho Lee
- Department of Internal Medicine, Gachon University Gil Medical Center, Incheon 21565, Korea.,Department of Internal Medicine, Gachon University College of Medicine, Incheon 21999, Korea
| |
Collapse
|
311
|
Bioactive constituents from Vitex negundo var. heterophylla and their antioxidant and α-glucosidase inhibitory activities. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.05.047] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
312
|
Wilson JR, Shuey MM, Brown NJ, Devin JK. Hypertension and Type 2 Diabetes Are Associated With Decreased Inhibition of Dipeptidyl Peptidase-4 by Sitagliptin. J Endocr Soc 2017; 1:1168-1178. [PMID: 29264572 PMCID: PMC5686657 DOI: 10.1210/js.2017-00312] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 07/27/2017] [Indexed: 12/19/2022] Open
Abstract
Context Patients with diabetes often have comorbidities such as hypertension. It is not known how individual characteristics influence response to dipeptidyl peptidase-4 (DPP4) inhibitors. Objective We tested the hypothesis that individual characteristics, sitagliptin dose, and genetic variability in DPP4 influence DPP4 activity during sitagliptin. Design and Setting Post hoc analysis of clinical and laboratory data from individuals randomized to sitagliptin versus placebo in crossover studies. Patients and Interventions Sixty-five subjects [27 with type 2 diabetes mellitus (T2DM) and hypertension, 38 healthy controls] were randomized to 100 mg/d sitagliptin or 200 mg sitagliptin and matching placebo in double-blind, crossover fashion. Fasting blood was obtained at baseline and 60 to 180 minutes after sitagliptin or placebo. Main Outcome Measures DPP4 activity and antigen during placebo and sitagliptin and DPP4 inhibition during sitagliptin. Results Sitagliptin 100 mg/d was less effective at inhibiting DPP4 activity in individuals with T2DM and hypertension than in healthy controls (P = 0.001, percent inhibition). In healthy controls, 100 mg/d sitagliptin was not as effective as single-dose 200 mg sitagliptin (P = 0.001, percent inhibition). DPP4 genotypes rs2909451 TT (P = 0.02) and rs759717 CC (P = 0.02) were associated with DPP4 activity during sitagliptin. In multivariable analyses, T2DM with hypertension, sitagliptin dose, age, systolic blood pressure, DPP4 activity during placebo, and rs2909451 genotype were significantly associated with DPP4 activity during sitagliptin. Conclusions Sitagliptin is less effective in inhibiting DPP4 in individuals with T2DM and hypertension than in healthy controls. Higher doses of DPP4 inhibitors may be required in patients with the metabolic syndrome.
Collapse
Affiliation(s)
- Jessica R Wilson
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Nashville, Tennessee 37232
| | | | - Nancy J Brown
- Vanderbilt University Medical Center, Department of Medicine, Division of Clinical Pharmacology, Nashville, Tennessee 37232
| | - Jessica K Devin
- Vanderbilt University Medical Center, Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Nashville, Tennessee 37232
| |
Collapse
|
313
|
Böhm A, Wagner R, Machicao F, Holst JJ, Gallwitz B, Stefan N, Fritsche A, Häring HU, Staiger H. DPP4 gene variation affects GLP-1 secretion, insulin secretion, and glucose tolerance in humans with high body adiposity. PLoS One 2017; 12:e0181880. [PMID: 28750074 PMCID: PMC5531535 DOI: 10.1371/journal.pone.0181880] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/08/2017] [Indexed: 11/23/2022] Open
Abstract
Objective Dipeptidyl-peptidase 4 (DPP-4) cleaves and inactivates the insulinotropic hormones glucagon-like peptide 1 (GLP-1) and gastric inhibitory polypeptide, collectively termed incretins. DPP-4 inhibitors entered clinical practice as approved therapeutics for type-2 diabetes in 2006. However, inter-individual variance in the responsiveness to DPP-4 inhibitors was reported. Thus, we asked whether genetic variation in the DPP4 gene affects incretin levels, insulin secretion, and glucose tolerance in participants of the TÜbingen Family study for type-2 diabetes (TÜF). Research design and methods Fourteen common (minor allele frequencies ≥0.05) DPP4 tagging single nucleotide polymorphisms (SNPs) were genotyped in 1,976 non-diabetic TÜF participants characterized by oral glucose tolerance tests and bioimpedance measurements. In a subgroup of 168 subjects, plasma incretin levels were determined. Results We identified a variant, i.e., SNP rs6741949, in intron 2 of the DPP4 gene that, after correction for multiple comparisons and appropriate adjustment, revealed a significant genotype-body fat interaction effect on glucose-stimulated plasma GLP-1 levels (p = 0.0021). Notably, no genotype-BMI interaction effects were detected (p = 0.8). After stratification for body fat content, the SNP negatively affected glucose-stimulated GLP-1 levels (p = 0.0229), insulin secretion (p = 0.0061), and glucose tolerance (p = 0.0208) in subjects with high body fat content only. Conclusions A common variant, i.e., SNP rs6741949, in the DPP4 gene interacts with body adiposity and negatively affects glucose-stimulated GLP-1 levels, insulin secretion, and glucose tolerance. Whether this SNP underlies the reported inter-individual variance in responsiveness to DPP-4 inhibitors, at least in subjects with high body fat content, remains to be shown.
Collapse
Affiliation(s)
- Anja Böhm
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
| | - Robert Wagner
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
| | - Fausto Machicao
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
| | - Jens Juul Holst
- Section of Translational Metabolic Physiology, Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Baptist Gallwitz
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Norbert Stefan
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
- Department of Internal Medicine, Division of Nutritional and Preventive Medicine, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Hans-Ulrich Häring
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
- Interfaculty Center for Pharmacogenomics and Pharma Research at the Eberhard Karls University Tübingen, Tübingen, Germany
| | - Harald Staiger
- German Center for Diabetes Research (DZD), Tübingen, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, Tübingen, Germany
- Interfaculty Center for Pharmacogenomics and Pharma Research at the Eberhard Karls University Tübingen, Tübingen, Germany
- Institute of Pharmaceutical Sciences, Department of Pharmacy and Biochemistry, Eberhard Karls University Tübingen, Tübingen, Germany
- * E-mail:
| |
Collapse
|
314
|
Glycosaminoglycans Regulate CXCR3 Ligands at Distinct Levels: Protection against Processing by Dipeptidyl Peptidase IV/CD26 and Interference with Receptor Signaling. Int J Mol Sci 2017; 18:ijms18071513. [PMID: 28703769 PMCID: PMC5536003 DOI: 10.3390/ijms18071513] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 07/03/2017] [Accepted: 07/06/2017] [Indexed: 12/22/2022] Open
Abstract
CXC chemokine ligand (CXCL)9, CXCL10 and CXCL11 direct chemotaxis of mainly T cells and NK cells through activation of their common CXC chemokine receptor (CXCR)3. They are inactivated upon NH2-terminal cleavage by dipeptidyl peptidase IV/CD26. In the present study, we found that different glycosaminoglycans (GAGs) protect the CXCR3 ligands against proteolytic processing by CD26 without directly affecting the enzymatic activity of CD26. In addition, GAGs were shown to interfere with chemokine-induced CXCR3 signaling. The observation that heparan sulfate did not, and heparin only moderately, altered CXCL10-induced T cell chemotaxis in vitro may be explained by a combination of protection against proteolytic inactivation and altered receptor interaction as observed in calcium assays. No effect of CD26 inhibition was found on CXCL10-induced chemotaxis in vitro. However, treatment of mice with the CD26 inhibitor sitagliptin resulted in an enhanced CXCL10-induced lymphocyte influx into the joint. This study reveals a dual role for GAGs in modulating the biological activity of CXCR3 ligands. GAGs protect the chemokines from proteolytic cleavage but also directly interfere with chemokine–CXCR3 signaling. These data support the hypothesis that both GAGs and CD26 affect the in vivo chemokine function.
Collapse
|
315
|
Akoumianakis I, Antoniades C. Dipeptidyl peptidase IV inhibitors as novel regulators of vascular disease. Vascul Pharmacol 2017; 96-98:1-4. [PMID: 28697993 DOI: 10.1016/j.vph.2017.07.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 07/01/2017] [Indexed: 12/21/2022]
Abstract
Dipeptidyl peptidase IV (DPP-IV) has been revealed as an adipokine with potential relevance in cardiovascular disease (CVD), while clinically used DPP-IV inhibitors have demonstrated beneficial cardiovascular effects in several experimental studies. Perivascular adipose tissue (PVAT) is a unique adipose tissue depot in close anatomical proximity and bidirectional functional interaction with the vascular wall, which is a source of DPP-IV and its biology may be influenced by DPP-IV inhibition. Recently, DPP-IV inhibition has been associated with decreased local inflammation and oxidative stress both in the vascular wall and the PVAT, potentially regulating atherogenesis progression in vivo. DPP-IV inhibition may thus be a promising target in cardiovascular disease. However, the exact pleiotropic mechanisms that underlie the cardiovascular effects of DPP-IV inhibition need to be clarified, while the in vivo benefit of DPP-IV inhibition in humans remains unclear.
Collapse
|
316
|
Proost P, Struyf S, Van Damme J, Fiten P, Ugarte-Berzal E, Opdenakker G. Chemokine isoforms and processing in inflammation and immunity. J Autoimmun 2017; 85:45-57. [PMID: 28684129 DOI: 10.1016/j.jaut.2017.06.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 06/21/2017] [Indexed: 12/16/2022]
Abstract
The first dimension of chemokine heterogeneity is reflected by their discovery and purification as natural proteins. Each of those chemokines attracted a specific inflammatory leukocyte type. With the introduction of genomic technologies, a second wave of chemokine heterogeneity was established by the discovery of putative chemokine-like sequences and by demonstrating chemotactic activity of the gene products in physiological leukocyte homing. In the postgenomic era, the third dimension of chemokine heterogeneity is the description of posttranslational modifications on most chemokines. Proteolysis of chemokines, for instance by dipeptidyl peptidase IV (DPP IV/CD26) and by matrix metalloproteinases (MMPs) is already well established as a biological control mechanism to activate, potentiate, dampen or abrogate chemokine activities. Other posttranslational modifications are less known. Theoretical N-linked and O-linked attachment sites for chemokine glycosylation were searched with bio-informatic tools and it was found that most chemokines are not glycosylated. These findings are corroborated with a low number of experimental studies demonstrating N- or O-glycosylation of natural chemokine ligands. Because attached oligosaccharides protect proteins against proteolytic degradation, their absence may explain the fast turnover of chemokines in the protease-rich environments of infection and inflammation. All chemokines interact with G protein-coupled receptors (GPCRs) and glycosaminoglycans (GAGs). Whether lectin-like GAG-binding induces cellular signaling is not clear, but these interactions are important for leukocyte migration and have already been exploited to reduce inflammation. In addition to selective proteolysis, citrullination and nitration/nitrosylation are being added as biologically relevant modifications contributing to functional chemokine heterogeneity. Resulting chemokine isoforms with reduced affinity for GPCRs reduce leukocyte migration in various models of inflammation. Here, these third dimension modifications are compared, with reflections on the biological and pathological contexts in which these posttranslational modifications take place and contribute to the repertoire of chemokine functions and with an emphasis on autoimmune diseases.
Collapse
Affiliation(s)
- Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| | - Sofie Struyf
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| | - Pierre Fiten
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| | - Estefania Ugarte-Berzal
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Herestraat 49, B-3000, Leuven, Belgium.
| |
Collapse
|
317
|
Gupta A, Jelinek HF, Al-Aubaidy H. Glucagon like peptide-1 and its receptor agonists: Their roles in management of Type 2 diabetes mellitus. Diabetes Metab Syndr 2017; 11:225-230. [PMID: 27884496 DOI: 10.1016/j.dsx.2016.09.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 09/03/2016] [Indexed: 12/25/2022]
Abstract
This study summarizes major work which investigated the roles of glucagon like peptide-1 (GLP-1) and its receptor (GLP-1R); the use of GLP-1-R agonists and dipeptidyl peptidase 4 inhibitor in the management of type 2 diabetes mellitus. It focuses on the recent therapeutic development which has occurred in this field, and also discusses the potential treatments which can be discovered and implemented in the near future to design an effective therapy for type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Ankit Gupta
- School of Medicine, University of Tasmania, Hobart, Australia
| | - Herbert F Jelinek
- School of Community Health, Charles Sturt University, Albury, NSW, Australia
| | | |
Collapse
|
318
|
Xie MJ, Zhu MR, Lu CM, Jin Y, Gao LH, Li L, Zhou J, Li FF, Zhao QH, Liu HK, Sadler PJ, Sanchez-Cano C. Synthesis and characterization of oxidovanadium complexes as enzyme inhibitors targeting dipeptidyl peptidase IV. J Inorg Biochem 2017; 175:29-35. [PMID: 28692886 DOI: 10.1016/j.jinorgbio.2017.06.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/28/2022]
Abstract
Two oxidovanadium(IV) complexes carrying Schiff base ligands obtained from the condensation of 4,5-dichlorobenzene-1,2-diamine and salicylaldehyde derivatives were synthesised and characterised, including their X-ray crystallographic structures. They were evaluated as dipeptidyl peptidase IV (DPP-IV) inhibitors for the treatment of type 2 diabetes. These compounds were moderate inhibitors of DPP-IV, with IC50 values of ca. 40μM. In vivo tests showed that complexes 1 and 2 could lower significantly the level of glucose in the blood of alloxan-diabetic mice at doses of 22.5mgV·kg-1 and 29.6mgV·kg-1, respectively. Moreover, molecular modeling studies suggested that the oxidovanadium complexes 1 and 2 could fit well into the active-site cleft of the kinase domain of DPP-IV. To the best of our knowledge, this is the first report of vanadium complexes capable of inhibiting DPP-IV.
Collapse
Affiliation(s)
- Ming-Jin Xie
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, Yunnan, China.
| | - Ming-Rong Zhu
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, Yunnan, China
| | - Chun-Mei Lu
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, Yunnan, China
| | - Yi Jin
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, Yunnan, China
| | - Li-Hui Gao
- Biomedical Engineering Research Center, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Ling Li
- Biomedical Engineering Research Center, Kunming Medical University, Kunming 650500, Yunnan, China
| | - Jie Zhou
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, Yunnan, China
| | - Fan-Fang Li
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, Yunnan, China
| | - Qi Hua Zhao
- School of Chemical Science and Technology, Yunnan University, Kunming 650091, Yunnan, China
| | - Hong-Ke Liu
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Wenyuan Road 1, Nanjing 210023, China
| | - Peter J Sadler
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK.
| | - Carlos Sanchez-Cano
- Department of Chemistry, University of Warwick, Gibbet Hill Road, Coventry CV4 7AL, UK.
| |
Collapse
|
319
|
Tang Z, Wang Z, Hu Z, Zhang M, Li L, Li B. The role of bradykinin in lung ischemia-reperfusion injury in a rat lung transplantation model. Acta Cir Bras 2017; 31:807-812. [PMID: 28076504 DOI: 10.1590/s0102-865020160120000005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 11/14/2016] [Indexed: 01/27/2023] Open
Abstract
PURPOSE: To investigate the role of bradykinin in a rat lung transplantation (LTx) model and preliminarily discuss the relationship between bradykinin and CD26/DPP-4. METHODS: Rats were randomly divided into four groups: Control (CON), Sham, low potassium dextranglucose (LPD), and AB192 (n=15/group). Orthotopic single LTx was performed in the LPD and AB192 groups. The donor lungs were flush-perfused and preserved with low potassium dextranglucose (LPD) or LPD+CD26/DPP-4 catalytic inhibitor (AB192). LTx was performed after 18 h cold ischemia time and harvested two days post-LTx. Blood gas analysis (PO2), wet/dry weight ratio (W/D), myeloperoxidase activity (MPO), and lipid peroxidation (MDA) were analyzed at 48 hr after transplantation. Immunohistochemical (IHC) analysis was performed in the same sample and validated by Western-Blot. RESULTS: Compared to the LPD group, the AB192 group showed higher PO2, lower W/D ratio, and decreased MPO and MDA. IHC studies showed strong bradykinin β2 receptor (B2R) staining in the LPD group, especially in inflammatory cells, alveolar macrophages, and respiratory epithelial cells. Expression of B2R by Western-Blot was significantly different between the AB192 and LPD groups. CONCLUSION: Bradykinin may be a competitive substrate of DPP-4, and decreased bradykinin levels may enhance protective effects against ischemia/reperfusion injury during LTx.
Collapse
Affiliation(s)
- Zheng Tang
- PhD, Department of Cardiovascular Surgery, Renmin Hospital, Wuhan University, China. Conception and design of the study, acquisition and interpretation of data, manuscript writing
| | - Zhiwei Wang
- PhD, Full Professor, Department of Cardiovascular Surgery, Renmin Hospital, Wuhan University, China. Design and supervised all phases of the study, critical revision
| | - Zhipeng Hu
- PhD, Department of Cardiovascular Surgery, Renmin Hospital, Wuhan University, China. Acquisition of data, critical revision
| | - Min Zhang
- PhD, Department of Cardiovascular Surgery, Renmin Hospital, Wuhan University, China. Acquisition of data, critical revision
| | - Luocheng Li
- PhD, Department of Cardiovascular Surgery, Renmin Hospital, Wuhan University, China. Acquisition of data, critical revision
| | - Bowen Li
- Master, Department of Cardiovascular Surgery, Renmin Hospital, Wuhan University, China. Acquisition of data
| |
Collapse
|
320
|
Abstract
Proteases target many substrates, triggering changes in distinct biological processes correlated with cell migration, EMT/EndMT and fibrosis. Extracellular protease activity, demonstrated by secreted and membrane-bound protease forms, leads to ECM degradation, activation of other proteases (i.e., proteolysis of nonactive zymogens), decomposition of cell-cell junctions, release of sequestered growth factors (TGF-β and VEGF), activation of signal proteins and receptors, degradation of inflammatory inhibitors or inflammation-related proteins, and changes in cell mechanosensing and motility. Intracellular proteases, mainly caspases and cathepsins, modulate lysosome activity and signal transduction pathways. Herein, we discuss the current knowledge on the multidimensional impact of proteases on the development of fibrosis.
Collapse
|
321
|
Lorenzini J, Scott Fites J, Nett J, Klein BS. Blastomyces dermatitidis serine protease dipeptidyl peptidase IVA (DppIVA) cleaves ELR + CXC chemokines altering their effects on neutrophils. Cell Microbiol 2017; 19. [PMID: 28346820 DOI: 10.1111/cmi.12741] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 03/13/2017] [Accepted: 03/15/2017] [Indexed: 12/14/2022]
Abstract
Blastomycosis elicits a pyogranulomatous inflammatory response that involves a prominent recruitment of neutrophils to the site of infection. Although neutrophils are efficiently recruited to the site of infection, this event is paradoxically coupled with the host's inability to control infection by Blastomyces dermatitidis, the causative agent. The mechanisms underlying this characteristic pyogranulomatous response and inability of neutrophils to kill the yeast are poorly understood. We recently reported that the fungal protease dipeptidyl peptidase IVA (DppIVA) promotes B. dermatitidis virulence by cleaving a dipeptide from the N-terminus of C-C chemokines and granulocyte/macrophage-colony stimulating factor, thereby inactivating them. Herein, we present evidence that DppIVA can also truncate the N-terminus of members of the ELR+ CXC chemokine family, which are known to modulate neutrophil function. We show that the DppIVA cleaved form of human (h) CXCL-2, for example, hCXCL-2 (3-73), is a more potent neutrophil chemoattractant than its intact counterpart, but hCXCL-2 (3-73) is conversely impaired in its ability to prime the reactive oxygen species response of neutrophils. Thus, DppIVA action on ELR+ CXC chemokines may promote the pyogranulomatous response that is typical of blastomycosis, while also explaining the inability of neutrophils to control infection.
Collapse
Affiliation(s)
- Jenna Lorenzini
- Departments of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - J Scott Fites
- Departments of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jeniel Nett
- Department of Internal Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Bruce S Klein
- Departments of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Internal Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Department of Medical Microbiology and Immunology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
322
|
Aliyari Serej Z, Ebrahimi Kalan A, Mehdipour A, Nozad Charoudeh H. Regulation and roles of CD26/DPPIV in hematopoiesis and diseases. Biomed Pharmacother 2017; 91:88-94. [PMID: 28448874 DOI: 10.1016/j.biopha.2017.04.074] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 04/15/2017] [Accepted: 04/17/2017] [Indexed: 01/15/2023] Open
Abstract
Dipeptidyl peptidase IV (DPPIV),1 on the surface of certain cells, where it is also referred to as CD26, is involved in a vast majority of biological and pathological processes. CD26/DPPIV function contributes to cancer and tumor metastasis as well as inhibition of its expression which alters the expression of immune response-related genes. CD26/DPPIV is a widely distributed multifunctional integral membrane and secreted protein that is defined as early predictive biomarker in HIV, cancer and autoimmunity diseases like diabetes and multiple sclerosis. CD26/DPPIV-chemokine interaction may have a functional role in T-cells and overall immune function. It is expressed at low density on resting T cells, but is upregulated with T cell activation. In this review, we summarize valuable information about detailed biological aspects and pharmacokinetic characteristics of CD26/DPPIV and its clinical efficacy, focusing particularly on the role of CD26/DPPIV in immunological and non-immunological diseases. We also describe our recent work about umbilical cord blood (UCB)2 hematopoietic stem cell transplantation strategies in which identified CD26+ cells can be differentiated to immune cells under certain culture condition.
Collapse
Affiliation(s)
- Zeynab Aliyari Serej
- School of Advanced Medical Sciences, Applied Cell Sciences Department, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Ebrahimi Kalan
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Neuroscience Department, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Mehdipour
- School of Advanced Medical Sciences, Tissue Engineering Department, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hojjatollah Nozad Charoudeh
- School of Advanced Medical Sciences, Applied Cell Sciences Department, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
323
|
Wang Z, Yang L, Fan H, Wu P, Zhang F, Zhang C, Liu W, Li M. Screening of a natural compound library identifies emodin, a natural compound from Rheum palmatum Linn that inhibits DPP4. PeerJ 2017; 5:e3283. [PMID: 28507818 PMCID: PMC5428354 DOI: 10.7717/peerj.3283] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 04/07/2017] [Indexed: 12/11/2022] Open
Abstract
Historically, Chinese herbal medicines have been widely used in the treatment of hyperglycemia, but the mechanisms underlying their effectiveness remain largely unknown. Here, we screened a compound library primarily comprised of natural compounds extracted from herbs and marine organisms. The results showed that emodin, a natural compound from Rheum palmatum Linn, inhibited DPP4 activity with an in vitro IC50 of 5.76 µM without inhibiting either DPP8 or DPP9. A docking model revealed that emodin binds to DPP4 protein through Glu205 and Glu206, although with low affinity. Moreover, emodin treatment (3, 10 and 30 mg/kg, P.O.) in mice decreased plasma DPP4 activity in a dose-dependent manner. Our study suggests that emodin inhibits DPP4 activity and may represent a novel therapeutic for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Zhaokai Wang
- College of Life Sciences, Fujian Normal University, Fuzhou, P. R. China.,Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen, P. R. China
| | - Longhe Yang
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen, P. R. China
| | - Hu Fan
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen, P. R. China
| | - Peng Wu
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen, P. R. China
| | - Fang Zhang
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen, P. R. China
| | - Chao Zhang
- Engineering Research Center of Marine Biological Resource Comprehensive Utilization, Third Institute of Oceanography, State Oceanic Administration, Xiamen, P. R. China
| | - Wenjie Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen, P. R. China
| | - Min Li
- College of Life Sciences, Fujian Normal University, Fuzhou, P. R. China
| |
Collapse
|
324
|
Sokar SS, El-Sayad MES, Ghoneim MES, Shebl AM. Combination of Sitagliptin and Silymarin ameliorates liver fibrosis induced by carbon tetrachloride in rats. Biomed Pharmacother 2017; 89:98-107. [DOI: 10.1016/j.biopha.2017.02.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 01/19/2017] [Accepted: 02/07/2017] [Indexed: 12/30/2022] Open
|
325
|
Carbone LD, Bůžková P, Fink HA, Robbins JA, Bethel M, Isales CM, Hill WD. Association of DPP-4 activity with BMD, body composition, and incident hip fracture: the Cardiovascular Health Study. Osteoporos Int 2017; 28:1631-1640. [PMID: 28150034 PMCID: PMC5653373 DOI: 10.1007/s00198-017-3916-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 01/06/2017] [Indexed: 12/25/2022]
Abstract
There was no association of plasma DPP-4 activity levels with bone mineral density (BMD), body composition, or incident hip fractures in a cohort of elderly community-dwelling adults. INTRODUCTION Dipeptidyl peptidase IV (DPP-4) inactivates several key hormones including those that stimulate postprandial insulin secretion, and DPP-4 inhibitors (gliptins) are approved to treat diabetes. While DPP-4 is known to modulate osteogenesis, the relationship between DPP-4 activity and skeletal health is uncertain. The purpose of the present study was to examine possible associations between DPP-4 activity in elderly subjects enrolled in the Cardiovascular Health Study (CHS) and BMD, body composition measurements, and incident hip fractures. METHODS All 1536 male and female CHS participants who had evaluable DXA scans and plasma for DPP-4 activity were included in the analyses. The association between (1) BMD of the total hip, femoral neck, lumbar spine, and total body; (2) body composition measurements (% lean, % fat, and total body mass); and (3) incident hip fractures and plasma levels of DPP-4 activity were determined. RESULTS Mean plasma levels of DPP-4 activity were significantly higher in blacks (227 ± 78) compared with whites (216 ± 89) (p = 0.04). However, there was no significant association of DPP-4 activity with age or gender (p ≥ 0.14 for both). In multivariable adjusted models, there was no association of plasma DPP-4 activity with BMD overall (p ≥ 0.55 for all) or in gender stratified analyses (p ≥ 0.23). There was also no association of DPP-4 levels and incident hip fractures overall (p ≥ 0.24) or in gender stratified analyses (p ≥ 0.39). CONCLUSION Plasma DPP-4 activity, within the endogenous physiological range, was significantly associated with race, but not with BMD, body composition, or incident hip fractures in elderly community-dwelling subjects.
Collapse
Affiliation(s)
- L D Carbone
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, USA
- Department of Medicine, Medical College of Georgia, Augusta University (Formerly Georgia Regents University and Georgia Health Sciences University), Augusta, GA, USA
| | - P Bůžková
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - H A Fink
- Geriatric Research Education & Clinical Center, Veterans Affairs Health Care System, Minneapolis, MN, USA
- Center for Chronic Disease Outcomes Research, Veterans Affairs Health Care System, Minneapolis, MN, USA
- Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - J A Robbins
- Department of Medicine, University of California-Davis, Sacramento, CA, USA
| | - M Bethel
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, USA
- Department of Medicine, Medical College of Georgia, Augusta University (Formerly Georgia Regents University and Georgia Health Sciences University), Augusta, GA, USA
| | - C M Isales
- Department of Medicine, Medical College of Georgia, Augusta University (Formerly Georgia Regents University and Georgia Health Sciences University), Augusta, GA, USA
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University (Formerly Georgia Regents University and Georgia Health Sciences University), Augusta, GA, USA
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University (Formerly Georgia Regents University and Georgia Health Sciences University), Augusta, GA, USA
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University (Formerly Georgia Regents University and Georgia Health Sciences University), Augusta, GA, USA
| | - W D Hill
- Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, USA.
- Institute for Regenerative and Reparative Medicine, Medical College of Georgia, Augusta University (Formerly Georgia Regents University and Georgia Health Sciences University), Augusta, GA, USA.
- Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University (Formerly Georgia Regents University and Georgia Health Sciences University), Augusta, GA, USA.
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University (Formerly Georgia Regents University and Georgia Health Sciences University), Sanders Research Building, CB1119, b1459 Laney-Walker Blvd., Augusta, GA, 30912-2000, USA.
| |
Collapse
|
326
|
Sitagliptin exerts anti-apoptotic effect in nephrotoxicity induced by cisplatin in rats. Naunyn Schmiedebergs Arch Pharmacol 2017; 390:721-731. [DOI: 10.1007/s00210-017-1367-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/15/2017] [Indexed: 12/16/2022]
|
327
|
Aldewachi HS, Woodroofe N, Turega S, Gardiner PHE. Optimization of gold nanoparticle-based real-time colorimetric assay of dipeptidyl peptidase IV activity. Talanta 2017; 169:13-19. [PMID: 28411801 DOI: 10.1016/j.talanta.2017.03.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 03/11/2017] [Accepted: 03/15/2017] [Indexed: 12/16/2022]
Abstract
Dipeptidyl peptidase IV (DPP-IV also referred to as CD-26) is a serine protease enzyme with remarkable diagnostic and prognostic value in a variety of health and disease conditions. Herein, we describe a simple and real-time colorimetric assay for DPP-IV/CD-26 activity based on the aggregation of gold nanoparticles (AuNPs) functionalized with the peptide substrates: Gly-Pro-Asp-Cys (GPDC) or Val-Pro-ethylene diamine-Asp-Cys (VP-ED-DC). Cleavage of the substrates by DPP-IV resulted in aggregation of the AuNPs with accompanying color change in the solution from red to blue that was monitored using either a UV-visible spectrophotometer or by the naked eye. Factors, such as time course of the reaction, stability of the functionalized AuNPs and the structure of the substrate that influence the cleavage reaction in solution were investigated. The effects of potential interference from serum proteins (lysozyme, thrombin and trypsin) on the analytical response were negligible. The detection limits when GPDC or VP-EN-DC functionalized AuNPs were used for DPP-IV assay were 1.2U/L and 1.5U/L, respectively. The VP-EN-DC method was preferred for the quantitative determination of DPP-IV activity in serum because of its wide linear range 0-30U/L compared to 0-12U/L for the GPDC assay. Recoveries from serum samples spiked with DPP-IV activity, between 5 and 25U/L, and using the VP-EN-DC modified AuNPs method ranged between 83.6% and 114.9%. The two colorimetric biosensors described here are superior to other conventional methods because of their simplicity, stability, selectivity and reliability.
Collapse
Affiliation(s)
- Hasan Saad Aldewachi
- Biomolecular Research Centre, Sheffield Hallam University, City Campus, Sheffield S1 1WB, UK; Pharmacy College, Mosul University, Mosul, Iraq
| | - Nicola Woodroofe
- Biomolecular Research Centre, Sheffield Hallam University, City Campus, Sheffield S1 1WB, UK
| | - Simon Turega
- Biomolecular Research Centre, Sheffield Hallam University, City Campus, Sheffield S1 1WB, UK
| | - Philip H E Gardiner
- Biomolecular Research Centre, Sheffield Hallam University, City Campus, Sheffield S1 1WB, UK.
| |
Collapse
|
328
|
Lourenço P, Silva S, Cunha F, Pereira J, Ribeiro A, Silva N, Guimarães JT, Araújo JP, Bettencourt P. Dipeptidyl peptidase-IV in chronic heart failure with reduced ejection fraction. Int J Cardiol 2017; 241:249-254. [PMID: 28318663 DOI: 10.1016/j.ijcard.2017.03.042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/14/2017] [Accepted: 03/09/2017] [Indexed: 01/31/2023]
Abstract
BACKGROUND An association between dipeptidyl peptidase-IV (DPP-IV) inhibitors with worse prognosis in HF has been suggested. We aimed to assess the serum DPP-IV levels in chronic stable HF patients and determine their association with prognosis. METHODS AND RESULTS Chronic stable HF patients with optimized prognostic-modifying therapy were prospectively recruited. EXCLUSION CRITERIA 1) ejection fraction>50%, 2) hospitalizations or therapeutic adjustments in the previous 2months; 3) patients on renal replacement therapy, and 4) use of DPP-IV inhibitors. A fasting venous blood sample was collected and DPP-IV was measured. Patients were followed-up for 3years and the endpoint studied was all-cause death. Patients' characteristics were compared according to DPP-IV quartiles. A Cox regression analysis was performed and multivariate models were built. The 3rd DPP-IV quartile was the reference category. We studied 264 patients. Mean age: 69 (±13)years, 70.5% were male and 33.7% diabetic. Median (IQR) serum DPP-IV levels were 455.6 (350.0-625.5)ng/mL. DPP-IV had an inverse relationship with age. Patients in 3rd DPP-IV quartile were in lower NYHA classes and had the lowest 3years all-cause mortality. Patients in the 1st DPP-IV quartile had a multivariate adjusted HR of 3-year mortality of 2.62 (95%CI: 1.15-5.95) when compared with reference category and the HR for the 4th quartile was of 3.79 (95%CI: 1.68-8.54). CONCLUSIONS There is a U-shaped association of serum DPP-IV with mortality in chronic systolic HF patients. Patients in the 3rd DPP-IV quartile have the best multivariate adjusted 3-year survival. DPP-IV inhibition might be harmful in patients with low DPP-IV.
Collapse
Affiliation(s)
| | - Sérgio Silva
- Serviço de Medicina Interna,Centro Hospitalar São João, Portugal
| | - Filipe Cunha
- Serviço de Endocrinologia, Centro Hospitalar São João, Portugal
| | - Joana Pereira
- Serviço de Medicina Interna,Centro Hospitalar São João, Portugal
| | - Ana Ribeiro
- Serviço de Medicina Interna,Centro Hospitalar São João, Portugal
| | - Nuno Silva
- Serviço de Patologia Clínica, Centro Hospitalar São João, Portugal
| | - João Tiago Guimarães
- Faculdade de Medicina da Universidade do Porto, Unidade I&D Cardiovascular do Porto, Portugal; Serviço de Patologia Clínica, Centro Hospitalar São João, Portugal
| | - José Paulo Araújo
- Serviço de Medicina Interna,Centro Hospitalar São João, Portugal; Faculdade de Medicina da Universidade do Porto, Unidade I&D Cardiovascular do Porto, Portugal
| | - Paulo Bettencourt
- Serviço de Medicina Interna,Centro Hospitalar São João, Portugal; Faculdade de Medicina da Universidade do Porto, Unidade I&D Cardiovascular do Porto, Portugal
| |
Collapse
|
329
|
Miglio G, Vitarelli G, Klein T, Benetti E. Effects of linagliptin on human immortalized podocytes: a cellular system to study dipeptidyl-peptidase 4 inhibition. Br J Pharmacol 2017; 174:809-821. [PMID: 28177527 DOI: 10.1111/bph.13739] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 01/19/2017] [Accepted: 01/22/2017] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Dipeptidyl-peptidase 4 (DPP4) is expressed by resident renal cells, including glomerular cells. DPP4 inhibitors (gliptins) exert albuminuria lowering effects, but the role of renal DPP4 as a pharmacological target has not been elucidated. To better understand the actions of gliptins, the effects of linagliptin on the behaviour of immortalized human podocytes and mesangial cells were evaluated. EXPERIMENTAL APPROACH The expression of DPP4 was measured at both the mRNA and protein levels. The effects of linagliptin on DPP4 activity, cell growth and cell cycle progression were determined. The contribution of the stromal cell-derived factor-1- CXCR4/CXCR7 signalling pathways was evaluated by studying the effects of AMD3100 (a CXCR4 antagonist and CXCR7 agonist) alone and in combination with linagliptin. The contribution of ERK1/2 activation was analysed by studying the effects of the MAPK kinase 1/2 inhibitor AZD6244. KEY RESULTS DPP4 was highly expressed in podocytes. The activity of DPP4 and podocyte growth were reduced by linagliptin. The effects of sitagliptin on podocyte growth were similar to those of linagliptin, were associated with inhibition of cell proliferation and mimicked by AMD3100. Moreover, linagliptin and AMD3100 were found to have a synergistic interaction, whereas no interaction was seen between linagliptin and AZD6244. CONCLUSIONS AND IMPLICATIONS Our cultures of human glomerular cells represent a reliable system for investigating the actions of gliptins. Moreover, DPP4 contributes to the regulation of podocyte behaviour. Inhibition of DPP4 in podocytes could underlie the effects of linagliptin on glomerular cells.
Collapse
Affiliation(s)
- Gianluca Miglio
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Turin, Italy
| | - Giovanna Vitarelli
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Turin, Italy
| | - Thomas Klein
- Department of Cardio Metabolic Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Elisa Benetti
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Turin, Italy
| |
Collapse
|
330
|
Beckers PAJ, Gielis JF, Van Schil PE, Adriaensen D. Lung ischemia reperfusion injury: the therapeutic role of dipeptidyl peptidase 4 inhibition. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:129. [PMID: 28462209 DOI: 10.21037/atm.2017.01.41] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Dipeptidyl peptidase 4 (DPP4) is a cell surface protease that has been reported to play a role in glucose homeostasis, cancer, HIV, autoimmunity, immunology and inflammation. A role for DPP4 in ischemia-reperfusion injury (IRI) in the heart has been established. Dipeptidyl peptidase 4 inhibition (DPP4i) appeared to decrease infarct size, improves cardiac function and promotes myocardial regeneration. Lung ischemia reperfusion injury is caused by a complex mechanism in which macrophages and neutrophils play an important role. Generation of reactive oxygen species (ROS), uncoupling of nitric oxide synthase (NOS), activation of nuclear factor-κB (NF-κB), activation of nicotinamide adenine dinucleotide phosphate metabolism, and generation of pro-inflammatory cytokines lead to acute lung injury (ALI). In this review we present the current knowledge on DPP4 as a target to treat IRI in the lung. We also provide evidence of the roles of the DPP4 substrates glucagon-like peptide 1 (GLP-1), vasoactive intestinal peptide (VIP) and stromal cell-derived factor-1α (SDF-1α) in protection against oxidative stress through activation of the mitogen-activated protein kinase (MAPK) 1/2 and phosphatidylinositol 3'-kinase (PI3K)/Akt signal transduction pathways.
Collapse
Affiliation(s)
- Paul A J Beckers
- Antwerp Surgical Training, Anatomy & Research Center, Department of Medicine, University of Antwerp, Wilrijk, Belgium
| | - Jan F Gielis
- Antwerp Surgical Training, Anatomy & Research Center, Department of Medicine, University of Antwerp, Wilrijk, Belgium
| | - Paul E Van Schil
- Antwerp Surgical Training, Anatomy & Research Center, Department of Medicine, University of Antwerp, Wilrijk, Belgium
| | - Dirk Adriaensen
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
331
|
Vliegen G, Raju TK, Adriaensen D, Lambeir AM, De Meester I. The expression of proline-specific enzymes in the human lung. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:130. [PMID: 28462210 DOI: 10.21037/atm.2017.03.36] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The pathophysiology of lung diseases is very complex and proteolytic enzymes may play a role or could be used as biomarkers. In this review, the literature was searched to make an overview of what is known on the expression of the proline-specific peptidases dipeptidyl peptidase (DPP) 4, 8, 9, prolyl oligopeptidase (PREP) and fibroblast activation protein α (FAP) in the healthy and diseased lung. Search terms included asthma, chronic obstructive pulmonary disease (COPD), lung cancer, fibrosis, ischemia reperfusion injury and pneumonia. Knowledge on the loss or gain of protein expression and activity during disease might tie these enzymes to certain cell types, substrates or interaction partners that are involved in the pathophysiology of the disease, ultimately leading to the elucidation of their functional roles and a potential therapeutic target. Most data could be found on DPP4, while the other enzymes are less explored. Published data however often appear to be conflicting, the applied methods divers and the specificity of the assays used questionable. In conclusion, information on the expression of the proline-specific peptidases in the healthy and diseased lung is lacking, begging for further well-designed research.
Collapse
Affiliation(s)
- Gwendolyn Vliegen
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Tom K Raju
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Dirk Adriaensen
- Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Anne-Marie Lambeir
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| | - Ingrid De Meester
- Laboratory of Medical Biochemistry, Department of Pharmaceutical Sciences, University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
332
|
Grujic M, Matic IZ, Crnogorac MD, Velickovic AD, Kolundzija B, Cordero OJ, Juranic Z, Prodanovic S, Zlatanovic M, Babic D, Damjanov N. Activity and expression of dipeptidyl peptidase IV on peripheral blood mononuclear cells in patients with early steroid and disease modifying antirheumatic drugs naïve rheumatoid arthritis. Clin Chem Lab Med 2017; 55:73-81. [PMID: 27341562 DOI: 10.1515/cclm-2015-1279] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 05/22/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Dipeptidyl peptidase IV (DPPIV/CD26) plays an important role in T cell activation and immune regulation, however the role of this enzyme in early rheumatoid arthritis (eRA) has not been clearly defined. The aim of this study was to determine the serum activity of DPPIV, its expression on peripheral blood mononuclear cells (PBMC) and to examine possible correlations with disease activity (DAS28) in untreated patients with eRA. METHODS The study included 50 patients newly diagnosed with RA, who had not received any corticosteroid or disease modifying antirheumatic drugs (DMARD) therapy and whose conventional radiographs of hands and feet showed no structural damage. The control group consisted of 40 healthy volunteers. Also, 30 patients with chronic RA (cRA) were examined. The serum activity of DPPIV was determined by the direct photometric method, while expression of CD26 on PBMC was determined using flow cytometry. RESULTS Decreased DPPIV serum activity was detected in patients with eRA and cRA compared to the control group (p=0.024, p<0.0001, respectively). Although, the percentage of overall CD26+ white blood cells (WBC) was significantly decreased in eRA patients (p<0.001), the percentage of CD26+ lymphocytes and monocytes and mean fluorescence intensity of CD26 on these cells in eRA patients showed no significant difference compared to healthy volunteers. DAS28 showed no significant correlation with CD26 expression or DPPIV serum activity, but a significant inverse correlation between the duration of symptoms and DPPIV serum activity was observed. CONCLUSIONS Our results show that a decrease in DPPIV serum activity, but not CD26 expression, is present in an early stage of rheumatoid arthritis.
Collapse
|
333
|
Baumeier C, Saussenthaler S, Kammel A, Jähnert M, Schlüter L, Hesse D, Canouil M, Lobbens S, Caiazzo R, Raverdy V, Pattou F, Nilsson E, Pihlajamäki J, Ling C, Froguel P, Schürmann A, Schwenk RW. Hepatic DPP4 DNA Methylation Associates With Fatty Liver. Diabetes 2017; 66:25-35. [PMID: 27999105 DOI: 10.2337/db15-1716] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 09/26/2016] [Indexed: 12/20/2022]
Abstract
Hepatic DPP4 expression is elevated in subjects with ectopic fat accumulation in the liver. However, whether increased dipeptidyl peptidase 4 (DPP4) is involved in the pathogenesis or is rather a consequence of metabolic disease is not known. We therefore studied the transcriptional regulation of hepatic Dpp4 in young mice prone to diet-induced obesity. Already at 6 weeks of age, expression of hepatic Dpp4 was increased in mice with high weight gain, independent of liver fat content. In the same animals, methylation of four intronic CpG sites was decreased, amplifying glucose-induced transcription of hepatic Dpp4 In older mice, hepatic triglyceride content was increased only in animals with elevated Dpp4 expression. Expression and release of DPP4 were markedly higher in the liver compared with adipose depots. Analysis of human liver biopsy specimens revealed a correlation of DPP4 expression and DNA methylation to stages of hepatosteatosis and nonalcoholic steatohepatitis. In summary, our results indicate a crucial role of the liver in participation to systemic DPP4 levels. Furthermore, the data show that glucose-induced expression of Dpp4 in the liver is facilitated by demethylation of the Dpp4 gene early in life. This might contribute to early deteriorations in hepatic function, which in turn result in metabolic disease such as hepatosteatosis later in life.
Collapse
Affiliation(s)
- Christian Baumeier
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrüecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Sophie Saussenthaler
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrüecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Anne Kammel
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrüecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Markus Jähnert
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrüecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Luisa Schlüter
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrüecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Deike Hesse
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrüecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Mickaël Canouil
- CNRS UMR 8199, Lille Pasteur Institute, Lille, France
- Lille 1 University, Lille, France
- European Genome Institute for Diabetes, Lille, France
| | - Stephane Lobbens
- CNRS UMR 8199, Lille Pasteur Institute, Lille, France
- Lille 1 University, Lille, France
- European Genome Institute for Diabetes, Lille, France
| | - Robert Caiazzo
- Lille 1 University, Lille, France
- European Genome Institute for Diabetes, Lille, France
- INSERM UMR 1190, CHU Lille, Lille, France
| | - Violeta Raverdy
- Lille 1 University, Lille, France
- European Genome Institute for Diabetes, Lille, France
- INSERM UMR 1190, CHU Lille, Lille, France
| | - François Pattou
- Lille 1 University, Lille, France
- European Genome Institute for Diabetes, Lille, France
- INSERM UMR 1190, CHU Lille, Lille, France
| | - Emma Nilsson
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Malmö, Sweden
| | - Jussi Pihlajamäki
- Institute of Public Health and Clinical Nutrition, Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Clinical Nutrition and Obesity Center, Kuopio University Hospital, Kuopio, Finland
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Malmö, Sweden
| | - Philippe Froguel
- CNRS UMR 8199, Lille Pasteur Institute, Lille, France
- Lille 1 University, Lille, France
- European Genome Institute for Diabetes, Lille, France
- Department of Genomics of Common Disease, School of Public Health, Hammersmith Hospital, Imperial College London, London, U.K
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrüecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Robert W Schwenk
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbrüecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| |
Collapse
|
334
|
Gschwandtner M, Paulitschke V, Mildner M, Brunner PM, Hacker S, Eisenwort G, Sperr WR, Valent P, Gerner C, Tschachler E. Proteome analysis identifies L1CAM/CD171 and DPP4/CD26 as novel markers of human skin mast cells. Allergy 2017; 72:85-97. [PMID: 27091730 DOI: 10.1111/all.12919] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND The function of skin mast cells has been well documented in IgE-mediated allergic reactions, whereas other mast cell functions are poorly defined. This study aimed at identifying novel mast cell proteins by proteome analysis of primary human skin mast cells. METHODS The proteome of skin mast cells was compared to other cell types and analyzed using bioinformatics. The expression and function of two proteins hitherto not described in skin mast cells was investigated in isolated mast cells as well as in mast cells in situ. RESULTS Within the mast cell proteome, we identified 49 highly expressed proteins previously not described in mast cells; 21 of these proteins were found to be selectively expressed in mast cells. Two proteins, the neural cell adhesion molecule L1 and dipeptidyl peptidase 4, were further studied. L1 was found to be highly expressed in mast cells in normal, psoriasis, and mastocytosis skin. Dipeptidyl peptidase 4 was found to be expressed in mast cells in normal, psoriasis, and mastocytosis skin as well as in bone marrow mast cells in patients with systemic mastocytosis. In normal skin, mast cells were identified as a major source of dipeptidyl peptidase 4 and we also found that skin mast cells and fibroblasts secrete an active form of this enzyme. CONCLUSIONS In a systematic proteomics approach we identified two novel mast cell proteins potentially relevant to skin homeostasis: neural cell adhesion molecule L1 and dipeptidyl peptidase 4.
Collapse
Affiliation(s)
- M. Gschwandtner
- Research Division of Biology and Pathobiology of the Skin; Department of Dermatology; Medical University of Vienna; Vienna Austria
| | - V. Paulitschke
- Department of Dermatology; Medical University of Vienna; Vienna Austria
| | - M. Mildner
- Research Division of Biology and Pathobiology of the Skin; Department of Dermatology; Medical University of Vienna; Vienna Austria
| | - P. M. Brunner
- Department of Dermatology; Medical University of Vienna; Vienna Austria
| | - S. Hacker
- Department of Plastic and Reconstructive Surgery; Medical University of Vienna; Vienna Austria
| | - G. Eisenwort
- Department of Internal Medicine I; Division of Hematology and Hemostaseology; Medical University of Vienna; Vienna Austria
- Ludwig Boltzmann Cluster Oncology; Medical University of Vienna; Vienna Austria
| | - W. R. Sperr
- Department of Internal Medicine I; Division of Hematology and Hemostaseology; Medical University of Vienna; Vienna Austria
- Ludwig Boltzmann Cluster Oncology; Medical University of Vienna; Vienna Austria
| | - P. Valent
- Department of Internal Medicine I; Division of Hematology and Hemostaseology; Medical University of Vienna; Vienna Austria
- Ludwig Boltzmann Cluster Oncology; Medical University of Vienna; Vienna Austria
| | - C. Gerner
- Institute of Analytical Chemistry; Faculty of Chemistry; University of Vienna; Vienna Austria
- Institute of Cancer Research; Comprehensive Cancer Center; Medical University of Vienna; Vienna Austria
| | - E. Tschachler
- Research Division of Biology and Pathobiology of the Skin; Department of Dermatology; Medical University of Vienna; Vienna Austria
| |
Collapse
|
335
|
Al-Tawfiq JA, Memish ZA. Update on therapeutic options for Middle East Respiratory Syndrome Coronavirus (MERS-CoV). Expert Rev Anti Infect Ther 2016; 15:269-275. [PMID: 27937060 PMCID: PMC7103731 DOI: 10.1080/14787210.2017.1271712] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is an important emerging respiratory pathogen. MERS-CoV resulted in multiple hospital outbreaks within and outside the Arabian Peninsula. The disease has a high case fatality rate, with the need for a therapeutic option. Areas covered: In this review, we provide an overview of the progress in the development of therapeutic strategies for MERS. We searched PubMed, Embase, Cochrane, Scopus, and Google Scholar, using the following terms: 'MERS', 'MERS-CoV', 'Middle East respiratory syndrome' in combination with 'treatment' or 'therapy'. Expert commentary: There are multiple agents tried in vitro and in vivo. None of these agents were used in large clinical studies. Available clinical studies are limited to the use of the combination of interferon and other agents. These clinical studies are based solely on case reports and case series. There are no prospective or randomized trials. There is a need to have prospective and randomized clinical trials for the therapy of MERS-CoV. However, this strategy might be hampered by the sporadic cases outside the large hospital outbreaks.
Collapse
Affiliation(s)
- Jaffar A Al-Tawfiq
- a Specialty Internal Medicine , Johns Hopkins Aramco Healthcare , Dhahran , Kingdom of Saudi Arabia.,b Department of Medicine , Indiana University School of Medicine , Indianapolis , IN , USA
| | - Ziad A Memish
- c Department of Medicine , Ministry of Health , Riyadh , Kingdom of Saudi Arabia.,d Hubert Department of Global Health, Rollins School of Public Health , Emory University , Atlanta , GA , USA
| |
Collapse
|
336
|
Angioedema Due to ACE Inhibitors. CURRENT TREATMENT OPTIONS IN ALLERGY 2016. [DOI: 10.1007/s40521-016-0099-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
337
|
Ye C, Tian X, Yue G, Yan L, Guan X, Wang S, Hao C. Suppression of CD26 inhibits growth and metastasis of pancreatic cancer. Tumour Biol 2016; 37:15677–15686. [PMID: 27718126 DOI: 10.1007/s13277-016-5315-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 09/05/2016] [Indexed: 10/20/2022] Open
Abstract
CD26/DPPIV is a glycosylated transmembrane type II protein and has a multitude of biological functions, while its impact on the malignant phenotypes of cancer cells has not been fully understood. This study aimed to investigate the effect of CD26 on growth and metastasis of pancreatic cancer cells in vitro and in vivo. We found in this study that CD26 expression was higher in cell lines that derived from the metastatic sites than those from the primary tumor sites. In specimens of pancreatic cancer patients, CD26 expression was higher in cancerous tissues than in paired normal tissues. In in vitro experiments, knockdown of CD26 expression inhibited cell growth, migration, invasion, colony formation, and increased cell apoptosis of pancreatic cancer cells. Knockdown of CD26 also decreased tumor growth and liver metastasis in vivo by using xenograft animal models. Suppression of CD26 could inhibit expression of epithelial-mesenchymal transition (EMT) regulatory genes. Our results indicated that CD26 may represent a new therapeutic target for pancreatic cancer.
Collapse
Affiliation(s)
- Chunxiang Ye
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
- Department of Gastroenterological Surgery and Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, People's Republic of China
| | - Xiuyun Tian
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Guanjun Yue
- Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Liang Yan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Xiaoya Guan
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Shan Wang
- Department of Gastroenterological Surgery and Laboratory of Surgical Oncology, Peking University People's Hospital, Beijing, People's Republic of China.
| | - Chunyi Hao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Hepato-Pancreato-Biliary Surgery, Peking University Cancer Hospital and Institute, Beijing, People's Republic of China.
| |
Collapse
|
338
|
A mouse model for MERS coronavirus-induced acute respiratory distress syndrome. Nat Microbiol 2016; 2:16226. [PMID: 27892925 PMCID: PMC5578707 DOI: 10.1038/nmicrobiol.2016.226] [Citation(s) in RCA: 168] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 10/14/2016] [Indexed: 02/07/2023]
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) is a novel virus that emerged in 2012, causing acute respiratory distress syndrome (ARDS), severe pneumonia-like symptoms and multi-organ failure, with a case fatality rate of ∼36%. Limited clinical studies indicate that humans infected with MERS-CoV exhibit pathology consistent with the late stages of ARDS, which is reminiscent of the disease observed in patients infected with severe acute respiratory syndrome coronavirus. Models of MERS-CoV-induced severe respiratory disease have been difficult to achieve, and small-animal models traditionally used to investigate viral pathogenesis (mouse, hamster, guinea-pig and ferret) are naturally resistant to MERS-CoV. Therefore, we used CRISPR–Cas9 gene editing to modify the mouse genome to encode two amino acids (positions 288 and 330) that match the human sequence in the dipeptidyl peptidase 4 receptor, making mice susceptible to MERS-CoV infection and replication. Serial MERS-CoV passage in these engineered mice was then used to generate a mouse-adapted virus that replicated efficiently within the lungs and evoked symptoms indicative of severe ARDS, including decreased survival, extreme weight loss, decreased pulmonary function, pulmonary haemorrhage and pathological signs indicative of end-stage lung disease. Importantly, therapeutic countermeasures comprising MERS-CoV neutralizing antibody treatment or a MERS-CoV spike protein vaccine protected the engineered mice against MERS-CoV-induced ARDS. Mice made susceptible to MERS-CoV, using CRISPR–Cas9 to alter the gene encoding the dipeptidyl peptidase 4 receptor, allow efficient viral replication in the lungs and display symptoms indicative of severe acute respiratory stress.
Collapse
|
339
|
Metzemaekers M, Van Damme J, Mortier A, Proost P. Regulation of Chemokine Activity - A Focus on the Role of Dipeptidyl Peptidase IV/CD26. Front Immunol 2016; 7:483. [PMID: 27891127 PMCID: PMC5104965 DOI: 10.3389/fimmu.2016.00483] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 10/21/2016] [Indexed: 12/15/2022] Open
Abstract
Chemokines are small, chemotactic proteins that play a crucial role in leukocyte migration and are, therefore, essential for proper functioning of the immune system. Chemokines exert their chemotactic effect by activation of chemokine receptors, which are G protein-coupled receptors (GPCRs), and interaction with glycosaminoglycans (GAGs). Furthermore, the exact chemokine function is modulated at the level of posttranslational modifications. Among the different types of posttranslational modifications that were found to occur in vitro and in vivo, i.e., proteolysis, citrullination, glycosylation, and nitration, NH2-terminal proteolysis of chemokines has been described most intensively. Since the NH2-terminal chemokine domain mediates receptor interaction, NH2-terminal modification by limited proteolysis or amino acid side chain modification can drastically affect their biological activity. An enzyme that has been shown to provoke NH2-terminal proteolysis of various chemokines is dipeptidyl peptidase IV or CD26. This multifunctional protein is a serine protease that preferably cleaves dipeptides from the NH2-terminal region of peptides and proteins with a proline or alanine residue in the penultimate position. Various chemokines possess such a proline or alanine residue, and CD26-truncated forms of these chemokines have been identified in cell culture supernatant as well as in body fluids. The effects of CD26-mediated proteolysis in the context of chemokines turned out to be highly complex. Depending on the chemokine ligand, loss of these two NH2-terminal amino acids can result in either an increased or a decreased biological activity, enhanced receptor specificity, inactivation of the chemokine ligand, or generation of receptor antagonists. Since chemokines direct leukocyte migration in homeostatic as well as pathophysiologic conditions, CD26-mediated proteolytic processing of these chemotactic proteins may have significant consequences for appropriate functioning of the immune system. After introducing the chemokine family together with the GPCRs and GAGs, as main interaction partners of chemokines, and discussing the different forms of posttranslational modifications, this review will focus on the intriguing relationship of chemokines with the serine protease CD26.
Collapse
Affiliation(s)
- Mieke Metzemaekers
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven , Leuven , Belgium
| | - Jo Van Damme
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven , Leuven , Belgium
| | - Anneleen Mortier
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven , Leuven , Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology and Immunology, Rega Institute, KU Leuven , Leuven , Belgium
| |
Collapse
|
340
|
Glorie L, D'Haese PC, Verhulst A. Boning up on DPP4, DPP4 substrates, and DPP4-adipokine interactions: Logical reasoning and known facts about bone related effects of DPP4 inhibitors. Bone 2016; 92:37-49. [PMID: 27535784 DOI: 10.1016/j.bone.2016.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 07/29/2016] [Accepted: 08/11/2016] [Indexed: 12/19/2022]
Abstract
Dipeptidyl peptidase 4 (DPP4) is a conserved exopeptidase with an important function in protein regulation. The activity of DPP4, an enzyme which can either be anchored to the plasma membrane or circulate free in the extracellular compartment, affects the glucose metabolism, cellular signaling, migration and differentiation, oxidative stress and the immune system. DPP4 is also expressed on the surface of osteoblasts, osteoclasts and osteocytes, and was found to play a role in collagen metabolism. Many substrates of DPP4 have an established role in bone metabolism, among which are incretins, gastrointestinal peptides and neuropeptides. In general, their effects favor bone formation, but some effects are complex and have not been completely elucidated. DPP4 and some of its substrates are known to interact with adipokines, playing an essential role in the energy metabolism. The prolongation of the half-life of incretins through DPP4 inhibition led to the development of these inhibitors to improve glucose tolerance in diabetes. Current literature indicates that the inhibition of DPP4 activity might also result in a beneficial effect on the bone metabolism, but the long-term effect of DPP4 inhibition on fracture outcome has not been entirely established. Diabetic as well as postmenopausal osteoporosis is associated with an increased activity of DPP4, as well as a shift in the expression levels of DPP4 substrates, their receptors, and adipokines. The interactions between these factors and their relationship in bone metabolism are therefore an interesting field of study.
Collapse
Affiliation(s)
- Lorenzo Glorie
- Laboratory of Pathophysiology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium.
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| | - Anja Verhulst
- Laboratory of Pathophysiology, University of Antwerp, Universiteitsplein 1, 2610 Wilrijk, Belgium
| |
Collapse
|
341
|
Sato H, Kubota N, Kubota T, Takamoto I, Iwayama K, Tokuyama K, Moroi M, Sugi K, Nakaya K, Goto M, Jomori T, Kadowaki T. Anagliptin increases insulin-induced skeletal muscle glucose uptake via an NO-dependent mechanism in mice. Diabetologia 2016; 59:2426-2434. [PMID: 27525648 DOI: 10.1007/s00125-016-4071-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 07/04/2016] [Indexed: 10/21/2022]
Abstract
AIMS/HYPOTHESIS Recently, incretin-related agents have been reported to attenuate insulin resistance in animal models, although the underlying mechanisms remain unclear. In this study, we investigated whether anagliptin, the dipeptidyl peptidase 4 (DPP-4) inhibitor, attenuates skeletal muscle insulin resistance through endothelial nitric oxide synthase (eNOS) activation in the endothelial cells. We used endothelium-specific Irs2-knockout (ETIrs2KO) mice, which show skeletal muscle insulin resistance resulting from a reduction of insulin-induced skeletal muscle capillary recruitment as a consequence of impaired eNOS activation. METHODS In vivo, 8-week-old male ETIrs2KO mice were fed regular chow with or without 0.3% (wt/wt) DPP-4 inhibitor for 8 weeks to assess capillary recruitment and glucose uptake by the skeletal muscle. In vitro, human coronary arterial endothelial cells (HCAECs) were used to explore the effect of glucagon-like peptide 1 (GLP-1) on eNOS activity. RESULTS Treatment with anagliptin ameliorated the impaired insulin-induced increase in capillary blood volume, interstitial insulin concentration and skeletal muscle glucose uptake in ETIrs2KO mice. This improvement in insulin-induced glucose uptake was almost completely abrogated by the GLP-1 receptor (GLP-1R) antagonist exendin-(9-39). Moreover, the increase in capillary blood volume with anagliptin treatment was also completely inhibited by the NOS inhibitor. GLP-1 augmented eNOS phosphorylation in HCAECs, with the effect completely disappearing after exposure to the protein kinase A (PKA) inhibitor H89. These data suggest that anagliptin treatment enhances insulin-induced capillary recruitment and interstitial insulin concentrations, resulting in improved skeletal muscle glucose uptake by directly acting on the endothelial cells via NO- and GLP-1-dependent mechanisms in vivo. CONCLUSIONS/INTERPRETATION Anagliptin may be a promising agent to ameliorate skeletal muscle insulin resistance in obese patients with type 2 diabetes.
Collapse
Affiliation(s)
- Hiroyuki Sato
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Naoto Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
- Department of Clinical Nutrition Therapy, University of Tokyo, Tokyo, Japan.
- Clinical Nutrition Program, National Institute of Health and Nutrition, Tokyo, Japan.
- Laboratory for Metabolic Homeostasis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan.
| | - Tetsuya Kubota
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
- Clinical Nutrition Program, National Institute of Health and Nutrition, Tokyo, Japan
- Laboratory for Metabolic Homeostasis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
- Division of Cardiovascular Medicine, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Iseki Takamoto
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kaito Iwayama
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Kumpei Tokuyama
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | - Masao Moroi
- Division of Cardiovascular Medicine, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Kaoru Sugi
- Division of Cardiovascular Medicine, Toho University Ohashi Medical Center, Tokyo, Japan
| | - Keizo Nakaya
- Mie Research Laboratories, Sanwa Kagaku Kenkyusho Co. Ltd, Mie, Japan
| | - Moritaka Goto
- Mie Research Laboratories, Sanwa Kagaku Kenkyusho Co. Ltd, Mie, Japan
| | - Takahito Jomori
- Mie Research Laboratories, Sanwa Kagaku Kenkyusho Co. Ltd, Mie, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| |
Collapse
|
342
|
Duvnjak L, Blaslov K, Vučić Lovrenčić M, Knežević Ćuća J. Persons with latent autoimmune diabetes in adults express higher dipeptidyl peptidase-4 activity compared to persons with type 2 and type 1 diabetes. Diabetes Res Clin Pract 2016; 121:119-126. [PMID: 27693949 DOI: 10.1016/j.diabres.2016.09.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 09/04/2016] [Accepted: 09/13/2016] [Indexed: 12/12/2022]
Abstract
AIMS We aimed to determine serum dipeptidyl peptidase-4 (DPP-4) activity in a group of persons with latent autoimmune diabetes in adults (LADA) and to compare it with persons with type 1, type 2 diabetes and healthy controls. METHODS DPP-4 activity measurement was performed in 67 persons (21 with type 1, 26 type 2 and 19 with LADA) and 13 healthy age and gender matched controls. RESULTS Persons with LADA showed highest DPP-4 activity among the study groups (32.71±3.55 vs 25.37±2.84 vs 18.57±2.54 vs 18.57±2.61U/L p<0.001). Mean glutamic acid autoantibody in persons with LADA was 164.32±86.28IU/mL. It correlated with DPP-4 activity (r=0.484, p=0.013). Furthermore, DPP-4 activity correlated with waist circumference (r=0.279, p=0.034) and glycated haemoglobin A1c (r=0.483, p<0.001), as well as with LDL cholesterol (r=0.854, p<0.001) and total daily insulin dose (r=0.397, p=0.001). In the multinomial regression analysis DPP-4 activity remained associated with both LADA (prevalence ratio 1.058 (1.012-1.287), p=0.001) and type 1 diabetes (prevalence ratio 1.506 (1.335-1.765), p<0.001) while it did not show an association with type 2 diabetes (prevalence ratio 0.942 (0.713-1.988), p=0.564). CONCLUSIONS Persons with LADA express higher DPP-4 activity compared to persons with both type 1 and type 2 diabetes. The possible pathophysiological role of DPP-4 in the LADA pathogenesis needs to be further evaluated.
Collapse
Affiliation(s)
- L Duvnjak
- Vuk Vrhovac Clinic for Diabetes, Endocrinology and Metabolic Diseases, University Hospital Merkur, Zagreb, Croatia; School of Medicine Zagreb, Croatia
| | - K Blaslov
- Vuk Vrhovac Clinic for Diabetes, Endocrinology and Metabolic Diseases, University Hospital Merkur, Zagreb, Croatia.
| | - M Vučić Lovrenčić
- Vuk Vrhovac Clinic for Diabetes, Endocrinology and Metabolic Diseases, University Hospital Merkur, Zagreb, Croatia
| | - J Knežević Ćuća
- Vuk Vrhovac Clinic for Diabetes, Endocrinology and Metabolic Diseases, University Hospital Merkur, Zagreb, Croatia
| |
Collapse
|
343
|
Neidert LE, Mobley CB, Kephart WC, Roberts MD, Kluess HA. The serine protease, dipeptidyl peptidase IV as a myokine: dietary protein and exercise mimetics as a stimulus for transcription and release. Physiol Rep 2016; 4:4/12/e12827. [PMID: 27335432 PMCID: PMC4923230 DOI: 10.14814/phy2.12827] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 05/18/2016] [Indexed: 12/15/2022] Open
Abstract
Dipeptidyl-peptidase IV (DPP-IV) is an enzyme with numerous roles within the body, mostly related to regulating energy metabolism. DPP-IV is also a myokine, but the stimulus for its release is poorly understood. We investigated the transcription and release of DPP-IV from skeletal muscle in a three-part study using C2C12 myotube cultures, an acute rat exercise and postexercise feeding model, and human feeding or human exercise models. When myotubes were presented with leucine only, hydrolyzed whey protein, or chemicals that cause exercise-related signaling to occur in cell culture, all caused an increase in the mRNA expression of DPP-IV (1.63 to 18.56 fold change, P < 0.05), but only whey protein caused a significant increase in DPP-IV activity in the cell culture media. When rats were fed whey protein concentrate immediately following stimulated muscle contractions, DPP-IV mRNA in both the exercised and nonexercised gastrocnemius muscles significantly increased 2.5- to 3.7-fold (P < 0.05) 3-6 h following the exercise/feeding bout; of note exercise alone or postexercise leucine-only feeding had no significant effect. In humans, plasma and serum DPP-IV activities were not altered by the ingestion of whey protein up to 1 h post consumption, after a 10 min bout of vigorous running, or during the completion of three repeated lower body resistance exercise bouts. Our cell culture and rodent data suggest that whey protein increases DPP-IV mRNA expression and secretion from muscle cells. However, our human data suggest that DPP-IV is not elevated in the bloodstream following acute whey protein ingestion or exercise.
Collapse
Affiliation(s)
- Leslie E Neidert
- School of Kinesiology, Auburn University, 301 Wire Road, Auburn, Alabama, 36849
| | - C Brooks Mobley
- School of Kinesiology, Auburn University, 301 Wire Road, Auburn, Alabama, 36849
| | - Wesley C Kephart
- School of Kinesiology, Auburn University, 301 Wire Road, Auburn, Alabama, 36849
| | - Michael D Roberts
- School of Kinesiology, Auburn University, 301 Wire Road, Auburn, Alabama, 36849
| | - Heidi A Kluess
- School of Kinesiology, Auburn University, 301 Wire Road, Auburn, Alabama, 36849
| |
Collapse
|
344
|
Kawase H, Bando YK, Nishimura K, Aoyama M, Monji A, Murohara T. A dipeptidyl peptidase-4 inhibitor ameliorates hypertensive cardiac remodeling via angiotensin-II/sodium-proton pump exchanger-1 axis. J Mol Cell Cardiol 2016; 98:37-47. [DOI: 10.1016/j.yjmcc.2016.06.066] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2016] [Revised: 06/26/2016] [Accepted: 06/28/2016] [Indexed: 10/21/2022]
|
345
|
Abstract
Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are responsible for the higher insulin response after oral versus intravenous glucose administration. This effect is called the incretin effect. An impaired incretin effect in patients with type 2 diabetes focused attention on the possible importance of GIP and GLP-1 in diabetes mellitus. Metabolic control can be markedly improved by administration of exogenous GLP-1, but the native peptide is almost immediately degraded by the enzyme dipeptidyl peptidase IV (DPP IV) and, therefore, has little clinical value. Orally active inhibitors of DPP IV have now been developed and have been shown to enhance endogenous levels of GLP-1, resulting in improved glucose tolerance, lasting improvement of HbA1C and improved beta-cell function. In general the DPP IV inhibitors are weight neutral, and well tolerated. One DPP IV inhibitor, sitagliptin, was approved as a once-daily oral therapy for the treatment of type 2 diabetes mellitus in Mexico and USA in 2006, and Europe in 2007. Other DPP IV inhibitors are in late-stage clinical development.
Collapse
Affiliation(s)
- Tina Vilsbøll
- Department of Internal Medicine F, Gentofte Hospital, University of Copenhagen, Niels Andersens Vej 65, DK-2900 Hellerup, Denmark,
| | - Filip K Knop
- Department of Internal Medicine F, Gentofte Hospital, University of Copenhagen, Niels Andersens Vej 65, DK-2900 Hellerup, Denmark
| |
Collapse
|
346
|
DPP-4 inhibitors in diabetic complications: role of DPP-4 beyond glucose control. Arch Pharm Res 2016; 39:1114-28. [PMID: 27502601 DOI: 10.1007/s12272-016-0813-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 08/04/2016] [Indexed: 12/12/2022]
Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors (gliptins) are an emerging class of antidiabetic drugs that constitutes approximately fifty percent of the market share of the oral hypoglycemic drugs. Its mechanism of action for lowering blood glucose is essentially via inhibition of the rapid degradation of incretin hormones, such as glucagon-like peptide (GLP)-1 and gastric inhibitory polypeptide (GIP), thus the plasma concentration of GLP-1 increases, which promotes insulin secretion from the pancreatic β cells and suppresses glucagon secretion from the α cells. In addition to the direct actions on the pancreas, GLP-1 exhibits diverse actions on different tissues through its action on GLP-1 receptor, which is expressed ubiquitously. Moreover, DPP-4 has multiple substrates besides GLP-1 and GIP, including cytokines, chemokines, neuropeptides, and growth factors, which are involved in many pathophysiological conditions. Recently, it was suggested that DPP-4 is a new adipokine secreted from the adipose tissue, which plays an important role in the regulation of the endocrine function in obesity-associated type 2 diabetes. Consequently, DPP-4 inhibitors have been reported to exhibit cytoprotective functions against various diabetic complications affecting the liver, heart, kidneys, retina, and neurons. This review outlines the current understanding of the effect of DPP-4 inhibitors on the complications associated with type 2 diabetes, such as liver steatosis and inflammation, dysfunction of the adipose tissue and pancreas, cardiovascular diseases, nephropathy, and neuropathy in preclinical and clinical studies.
Collapse
|
347
|
Kanasaki K. The pathological significance of dipeptidyl peptidase-4 in endothelial cell homeostasis and kidney fibrosis. Diabetol Int 2016; 7:212-220. [PMID: 30603266 DOI: 10.1007/s13340-016-0281-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/22/2016] [Indexed: 02/06/2023]
Abstract
Endothelial dysfunction and tubulointerstitial fibrosis are characteristics of diabetic kidneys. Recent evidence has suggested that the diabetic kidney is associated with dipeptidyl peptidase (DPP)-4 overexpression in endothelial cells. Several insults can induce endothelial cells to alter their phenotype into a mesenchymal-like phenotype via endothelial-mesenchymal transition (EndMT), which plays pivotal roles in tissue fibrosis. We have recently revealed the fibrogenic role of DPP-4 through the induction of EndMT in diabetic kidneys. This review mainly focuses on the biological and pathological significance of DPP-4 overexpression in endothelial cells through the mechanisms of endothelial homeostasis defects, EndMT, and kidney fibrosis.
Collapse
Affiliation(s)
- Keizo Kanasaki
- 1Department of Diabetology and Endocrinology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293 Japan.,2Division of Anticipatory Molecular Food Science and Technology, Kanazawa Medical University, Uchinada, Ishikawa 920-0293 Japan
| |
Collapse
|
348
|
Abstract
Coronaviruses (CoVs) are enveloped RNA viruses that infect birds, mammals, and humans. Infections caused by human coronaviruses (hCoVs) are mostly associated with the respiratory, enteric, and nervous systems. The hCoVs only occasionally induce lower respiratory tract disease, including bronchitis, bronchiolitis, and pneumonia. In 2002 to 2003, a global outbreak of severe acute respiratory syndrome (SARS) was the seminal detection of a novel CoV (SARS-CoV). A decade later (June 2012), another novel CoV was implicated as the cause of Middle East respiratory syndrome (MERS) in Saudi Arabia. Although bats might serve as a reservoir of MERS-CoV, it is unlikely that they are the direct source for most human cases. Severe lines of evidence suggest that dromedary camels have been the major cause of transmission to humans. The emergence of MERS-CoV has triggered serious concerns about the potential for a widespread outbreak. All MERS cases were linked directly or indirectly to the Middle East region including Saudi Arabia, Jordan, Qatar, Oman, Kuwait, and UAE. MERS cases have also been reported in the later phases in the United Kingdom, France, Germany, Italy, Spain, and Tunisia. Most of these MERS cases were linked with the Middle East. The high mortality rates in family-based and hospital-based outbreaks were reported among patients with comorbidities such as diabetes and renal failure. MERS-CoV causes an acute, highly lethal pneumonia and renal dysfunction. The major complications reported in fatal cases are hyperkalemia with associated ventricular tachycardia, disseminated intravascular coagulation, pericarditis, and multiorgan failure. The case-fatality rate seems to be higher for MERS-CoV (around 30%) than for SARS-CoV (9.6%). The combination regimen of type 1 interferon + lopinavir/ritonavir is considered as the first-line therapy for MERS. Antiviral treatment is generally recommended for 10 to 14 days in patients with MERS-CoV infection. Convalescent plasma therapy has shown some efficacy among patients refractory to antiviral drugs if administered within 2 weeks of the onset of the disease.
Collapse
Affiliation(s)
- Sunit K Singh
- Molecular Biology Unit, Institute of Medical Sciences, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh, India
| |
Collapse
|
349
|
Arruda-Junior DF, Martins FL, Dariolli R, Jensen L, Antonio EL, Dos Santos L, Tucci PJF, Girardi ACC. Dipeptidyl Peptidase IV Inhibition Exerts Renoprotective Effects in Rats with Established Heart Failure. Front Physiol 2016; 7:293. [PMID: 27462276 PMCID: PMC4941796 DOI: 10.3389/fphys.2016.00293] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 06/27/2016] [Indexed: 12/14/2022] Open
Abstract
Circulating dipeptidyl peptidase IV (DPPIV) activity is associated with worse cardiovascular outcomes in humans and experimental heart failure (HF) models, suggesting that DPPIV may play a role in the pathophysiology of this syndrome. Renal dysfunction is one of the key features of HF, but it remains to be determined whether DPPIV inhibitors are capable of improving cardiorenal function after the onset of HF. Therefore, the present study aimed to test the hypothesis that DPPIV inhibition by vildagliptin improves renal water and salt handling and exerts anti-proteinuric effects in rats with established HF. To this end, male Wistar rats were subjected to left ventricle (LV) radiofrequency ablation or sham operation. Six weeks after surgery, radiofrequency-ablated rats who developed HF were randomly divided into two groups and treated for 4 weeks with vildagliptin (120 mg/kg/day) or vehicle by oral gavage. Echocardiography was performed before (pretreatment) and at the end of treatment (post-treatment) to evaluate cardiac function. The fractional area change (FAC) increased (34 ± 5 vs. 45 ± 3%, p < 0.05), and the isovolumic relaxation time decreased (33 ± 2 vs. 27 ± 1 ms; p < 0.05) in HF rats treated with vildagliptin (post-treatment vs. pretreatment). On the other hand, cardiac dysfunction deteriorated further in vehicle-treated HF rats. Renal function was impaired in vehicle-treated HF rats as evidenced by fluid retention, low glomerular filtration rate (GFR) and high levels of urinary protein excretion. Vildagliptin treatment restored urinary flow, GFR, urinary sodium and urinary protein excretion to sham levels. Restoration of renal function in HF rats by DPPIV inhibition was associated with increased active glucagon-like peptide-1 (GLP-1) serum concentration, reduced DPPIV activity and increased activity of protein kinase A in the renal cortex. Furthermore, the anti-proteinuric effect of vildagliptin treatment in rats with established HF was associated with upregulation of the apical proximal tubule endocytic receptor megalin and of the podocyte main slit diaphragm proteins nephrin and podocin. Collectively, these findings demonstrate that DPPIV inhibition exerts renoprotective effects and ameliorates cardiorenal function in rats with established HF. Long-term studies with DPPIV inhibitors are needed to ascertain whether these effects ultimately translate into improved clinical outcomes.
Collapse
Affiliation(s)
| | - Flavia L Martins
- Heart Institute (InCor), University of São Paulo Medical School São Paulo, Brazil
| | - Rafael Dariolli
- Heart Institute (InCor), University of São Paulo Medical School São Paulo, Brazil
| | - Leonardo Jensen
- Heart Institute (InCor), University of São Paulo Medical School São Paulo, Brazil
| | - Ednei L Antonio
- Cardiology Division, Department of Medicine, Federal University of São Paulo São Paulo, Brazil
| | - Leonardo Dos Santos
- Department of Physiological Sciences, Federal University of Espírito Santo Vitória, Brazil
| | - Paulo J F Tucci
- Cardiology Division, Department of Medicine, Federal University of São Paulo São Paulo, Brazil
| | - Adriana C C Girardi
- Heart Institute (InCor), University of São Paulo Medical School São Paulo, Brazil
| |
Collapse
|
350
|
Busek P, Vanickova Z, Hrabal P, Brabec M, Fric P, Zavoral M, Skrha J, Kmochova K, Laclav M, Bunganic B, Augustyns K, Van Der Veken P, Sedo A. Increased tissue and circulating levels of dipeptidyl peptidase-IV enzymatic activity in patients with pancreatic ductal adenocarcinoma. Pancreatology 2016; 16:829-38. [PMID: 27320722 DOI: 10.1016/j.pan.2016.06.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 05/04/2016] [Accepted: 06/02/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) is frequently heralded by an impairment of glucose homeostasis. Dipeptidyl peptidase-IV (DPP-IV) and fibroblast activation protein alpha (FAP) are aminopeptidases that regulate several bioactive peptides involved in glucoregulation, and are frequently dysregulated in cancer. The present study analyzes blood plasma levels and the quantity and localization of DPP-IV and FAP in PDAC tissues. METHODS DPP-IV and FAP concentration and enzymatic activity were evaluated in the plasma from 93 PDAC, 39 type 2 diabetes mellitus (T2DM) and 29 control subjects, and in matched paired non-tumorous and tumor tissues from 48 PDAC patients. The localization of DPP-IV and FAP was determined using immunohistochemistry and catalytic histochemistry. RESULTS The enzymatic activity and concentration of DPP-IV was higher in PDAC tumor tissues compared to non-tumorous pancreas. DPP-IV was expressed in cancer cells and in the fibrotic stroma by activated (myo)fibroblasts including DPP-IV(+)FAP(+) cells. FAP was expressed in stromal cells and in some cancer cells and its expression was increased in the tumors. Plasmatic DPP-IV enzymatic activity, and in particular the ratio between DPP-IV enzymatic activity and concentration in PDAC with recent onset DM was higher compared to T2DM. In contrast, the plasmatic FAP enzymatic activity was lower in PDAC compared to T2DM and controls and rose after tumor removal. CONCLUSIONS DPP-IV-like enzymatic activity is upregulated in PDAC tissues. PDAC patients with recent onset diabetes or prediabetes have increased plasmatic DPP-IV enzymatic activity. These changes may contribute to the frequently observed association of PDAC and recent onset impairment of glucoregulation.
Collapse
Affiliation(s)
- Petr Busek
- Laboratory of Cancer Cell Biology, Institute of Biochemistry & Experimental Oncology, First Faculty of Medicine, Charles University in Prague, Czech Republic.
| | - Zdislava Vanickova
- Laboratory of Cancer Cell Biology, Institute of Biochemistry & Experimental Oncology, First Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Petr Hrabal
- Department of Pathology, Military University Hospital Prague, Czech Republic
| | - Marek Brabec
- Department of Nonlinear Modeling, Institute of Computer Science, The Czech Academy of Sciences, Prague, Czech Republic
| | - Premysl Fric
- Department of Internal Medicine of First Faculty of Medicine of Charles University and Military University Hospital Prague, Czech Republic
| | - Miroslav Zavoral
- Department of Internal Medicine of First Faculty of Medicine of Charles University and Military University Hospital Prague, Czech Republic
| | - Jan Skrha
- 3rd Department of Medicine - Department of Endocrinology and Metabolism, First Faculty of Medicine, Charles University in Prague and General University Hospital in Prague, Czech Republic
| | - Klara Kmochova
- Department of Internal Medicine of First Faculty of Medicine of Charles University and Military University Hospital Prague, Czech Republic
| | - Martin Laclav
- Department of Internal Medicine of First Faculty of Medicine of Charles University and Military University Hospital Prague, Czech Republic
| | - Bohus Bunganic
- Department of Internal Medicine of First Faculty of Medicine of Charles University and Military University Hospital Prague, Czech Republic
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, UAMC University of Antwerp, Belgium
| | - Pieter Van Der Veken
- Laboratory of Medicinal Chemistry, Department of Pharmaceutical Sciences, UAMC University of Antwerp, Belgium
| | - Aleksi Sedo
- Laboratory of Cancer Cell Biology, Institute of Biochemistry & Experimental Oncology, First Faculty of Medicine, Charles University in Prague, Czech Republic.
| |
Collapse
|