1
|
Huang XD, Jiang DS, Feng X, Fang ZM. The benefits of oral glucose-lowering agents: GLP-1 receptor agonists, DPP-4 and SGLT-2 inhibitors on myocardial ischaemia/reperfusion injury. Eur J Pharmacol 2024; 976:176698. [PMID: 38821168 DOI: 10.1016/j.ejphar.2024.176698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Myocardial infarction (MI) is a life-threatening cardiovascular disease that, on average, results in 8.5 million deaths worldwide each year. Timely revascularization of occluded vessels is a critical method of myocardial salvage. However, reperfusion paradoxically leads to the worsening of myocardial damage known as myocardial ischaemia/reperfusion injury (MI/RI). Therefore, reducing the size of myocardial infarction after reperfusion is critical and remains an important therapeutic goal. The susceptibility of the myocardium to MI/RI may be increased by diabetes. Currently, some traditional antidiabetic agents such as metformin reduce MI/RI by decreasing inflammation, inhibiting oxidative stress, and improving vascular endothelial function. This appears to be a new direction for the treatment of MI/RI. Recent cardiovascular outcome trials have shown that several oral antidiabetic agents, including glucagon-like peptide-1 receptor agonists (GLP-1RAs), dipeptidyl peptidase-4 inhibitors (DPP-4is), and sodium-glucose-linked transporter-2 inhibitors (SGLT-2is), not only have good antidiabetic effects but also have a protective effect on myocardial protection. This article aims to discuss the mechanisms and effects of oral antidiabetic agents, including GLP-1RAs, DPP-4is, and SGLT-2is, on MI/RI to facilitate their clinical application.
Collapse
Affiliation(s)
- Xu-Dong Huang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Cardiothoracic Surgery, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Ding-Sheng Jiang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei, China
| | - Xin Feng
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Ze-Min Fang
- Division of Cardiovascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Cardiothoracic Surgery, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
2
|
Koueik J, Wesley UV, Dempsey RJ. Pathophysiology, cellular and molecular mechanisms of large and small vessel diseases. Neurochem Int 2023; 164:105499. [PMID: 36746322 DOI: 10.1016/j.neuint.2023.105499] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/25/2023] [Accepted: 01/29/2023] [Indexed: 02/07/2023]
Abstract
Cerebrovascular disease (CVD) is the second most common cause of cognitive impairment and dementia in aged population. CVD presents in a myriad number of clinical ways based on the functional location of pathology. While primary clinical emphasis has been placed on motor, speech and visual deficits, vascular cognitive decline is a vastly under recognized and devastating condition afflicting millions of Americans. CVD, a disease of the blood vessels that supply blood to brain involves an integration between small and large vessels. Cerebral large vessel diseases (LVD) are associated with atherosclerosis, artery-to-artery embolism, intracardiac embolism and a large vessel stroke leading to substantial functional disability. Cerebral small vessel disease (SVD) is critically involved in stroke, brain hemorrhages, cognitive decline and functional loss in elderly patients. An evolving understanding of cellular and molecular mechanisms emphasizes that inflammatory vascular changes contribute to systemic pathologic conditions of the central nervous systems (CNS), with specific clinical presentations including, cognitive decline. Advances in an understanding of pathophysiology of disease processes and therapeutic interventions may help improve outcomes. This review will focus on large and small vessels diseases and their relationship to vascular cognitive decline, atherosclerosis, stroke, and inflammatory neurodegeneration. We will also emphasize the molecular and cellular mechanisms, as well as genetic and epigenetic factors associated with LVD and SVD.
Collapse
Affiliation(s)
- Joyce Koueik
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA
| | - Umadevi V Wesley
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA
| | - Robert J Dempsey
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA.
| |
Collapse
|
3
|
Pharaboz A, Kimmoun A, Gunst J, Duarte K, Merkling T, Gayat E, Mebazaa A, Glenn-Chousterman B. Association between type II diabetes mellitus and 90-day mortality in a large multicenter prospectively collected cohort. A FROG ICU post-hoc study. J Crit Care 2023; 73:154195. [PMID: 36368176 DOI: 10.1016/j.jcrc.2022.154195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 09/01/2022] [Accepted: 10/17/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE Factors associated with adverse outcomes in ICU patients with type II (T2DM) are poorly defined. The main goal of this study is to determine the impact of pre-existing T2DM on 90-day mortality post ICU admission. MATERIAL Post-hoc analysis from the FROG-ICU cohort. All patients admitted to ICU who were ventilated and/or treated by a vasoactive agent for >24 h were included. Association between T2DM and 90-day mortality was analyzed in unmatched, and populations matched by propensity score (PS) method to balance confounders recorded before ICU admission. Analysis was performed in non-imputed and imputed datasets. RESULTS 2002 patients were included, and 16% had a history of T2DM. The latter were at inclusion more severely ill (SAPSII score 51(39-67) vs 48(35-61), p < 0.0001; Charlson score 2(1-3) vs 0(0-2), p < 0.0001). In the unmatched cohort, T2DM patients had a higher 90-day risk of death compared to no-DM patients (HR 1.35(1.1-1.65)). The 90-day risk of death was not significantly different T2DM and no T2DM patients after PS matching (HR: 0.81 (0.56-1.18). Results were similar with the analysis performed on imputed datasets (pooled HR: 0.95 (0.69-1.30)). CONCLUSIONS In the present study, T2DM was not associated with 90-day mortality post ICU admission.
Collapse
Affiliation(s)
- Alexandre Pharaboz
- Université de Paris, AP-HP, CHU Lariboisière, Department of Anesthesiology and Critical Care, FHU PROMICE, INSERM U942, F-CRIN-INI CRCT, Paris, France
| | - Antoine Kimmoun
- Université de Lorraine, CHRU de Nancy, Médecine Intensive et Réanimation Brabois, INSERM INSERM U942 and U1116, F-CRIN-INIC RCT, Vandœuvre-lès-Nancy, France
| | - Jan Gunst
- Clinical Division and Laboratory of Intensive Care Medicine, Department of Cellular and Molecular Medicine, KU Leuven Herestraat 49, B-3000 Leuven, Belgium
| | - Kevin Duarte
- Université de Lorraine, CHRU de Nancy, INSERM CIC-P 1433, INSERM, F-CRIN-INI CRCT, Vandœuvre-lès-Nancy, France
| | - Thomas Merkling
- Université de Lorraine, CHRU de Nancy, INSERM CIC-P 1433, INSERM, F-CRIN-INI CRCT, Vandœuvre-lès-Nancy, France
| | - Etienne Gayat
- Université de Paris, AP-HP, CHU Lariboisière, Department of Anesthesiology and Critical Care, FHU PROMICE, INSERM U942, F-CRIN-INI CRCT, Paris, France
| | - Alexandre Mebazaa
- Université de Paris, AP-HP, CHU Lariboisière, Department of Anesthesiology and Critical Care, FHU PROMICE, INSERM U942, F-CRIN-INI CRCT, Paris, France
| | - Benjamin Glenn-Chousterman
- Université de Paris, AP-HP, CHU Lariboisière, Department of Anesthesiology and Critical Care, FHU PROMICE, INSERM U942, F-CRIN-INI CRCT, Paris, France.
| |
Collapse
|
4
|
Fan C, Wu Y, Rui X, Yang Y, Ling C, Liu S, Liu S, Wang Y. Animal models for COVID-19: advances, gaps and perspectives. Signal Transduct Target Ther 2022; 7:220. [PMID: 35798699 PMCID: PMC9261903 DOI: 10.1038/s41392-022-01087-8] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 01/08/2023] Open
Abstract
COVID-19, caused by SARS-CoV-2, is the most consequential pandemic of this century. Since the outbreak in late 2019, animal models have been playing crucial roles in aiding the rapid development of vaccines/drugs for prevention and therapy, as well as understanding the pathogenesis of SARS-CoV-2 infection and immune responses of hosts. However, the current animal models have some deficits and there is an urgent need for novel models to evaluate the virulence of variants of concerns (VOC), antibody-dependent enhancement (ADE), and various comorbidities of COVID-19. This review summarizes the clinical features of COVID-19 in different populations, and the characteristics of the major animal models of SARS-CoV-2, including those naturally susceptible animals, such as non-human primates, Syrian hamster, ferret, minks, poultry, livestock, and mouse models sensitized by genetically modified, AAV/adenoviral transduced, mouse-adapted strain of SARS-CoV-2, and by engraftment of human tissues or cells. Since understanding the host receptors and proteases is essential for designing advanced genetically modified animal models, successful studies on receptors and proteases are also reviewed. Several improved alternatives for future mouse models are proposed, including the reselection of alternative receptor genes or multiple gene combinations, the use of transgenic or knock-in method, and different strains for establishing the next generation of genetically modified mice.
Collapse
Affiliation(s)
- Changfa Fan
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), National Rodent Laboratory Animal Resources Center, Beijing, 102629, China
| | - Yong Wu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), National Rodent Laboratory Animal Resources Center, Beijing, 102629, China
| | - Xiong Rui
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), National Rodent Laboratory Animal Resources Center, Beijing, 102629, China
- Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100083, China
| | - Yuansong Yang
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), National Rodent Laboratory Animal Resources Center, Beijing, 102629, China
| | - Chen Ling
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), National Rodent Laboratory Animal Resources Center, Beijing, 102629, China
- College of Life Sciences, Northwest University; Provincial Key Laboratory of Biotechnology of Shaanxi Province, Northwest University, Xi'an, 710069, China
| | - Susu Liu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), National Rodent Laboratory Animal Resources Center, Beijing, 102629, China
| | - Shunan Liu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control (NIFDC), National Rodent Laboratory Animal Resources Center, Beijing, 102629, China
| | - Youchun Wang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC), Beijing, China.
| |
Collapse
|
5
|
Verma P, Kaur L, Aswal P, Singh A, Ojha H, Rahman AJ, Singhal R, Tiwari AK, Pathak M. Luminescence studies of binding affinity of vildagliptin with bovine serum albumin. J Biomol Struct Dyn 2022; 41:3002-3013. [PMID: 35220922 DOI: 10.1080/07391102.2022.2043939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Vildagliptin (VDG)is a frontier drug for diabetes mellitus. It is prescribed both in the monotherapy as well as in an amalgamation with other antidiabetic drugs. Drug-serum protein binding is an essential parameter which influences ADME properties of the drug. In current study, binding of VDG with serum protein (bovine serum albumin: BSA) was investigated using multi-spectroscopic techniques. A computational approach was also employed to identify the binding affinity of VDG with BSA at both Sudlow I and II sites. An enzyme activity assay specific for esterase was also investigated to know the post-binding consequences of VDG with BSA. Fluorescence spectra of BSA samples treated with VDG shows static quenching with binding parameters for VDG-BSA complex show single class of equivalent binding stoichiometry(n = 1.331) and binding constant 1.1 x 104M-1 at 298.15 K. The binding constant indicates important role of non-polar interactions in the binding process. Fluorescence resonance energy transfer (FRET) analysis of VDG absorption spectra and emission spectrum of BSA confirmed no significant resonance in energy transfer. Synchronous fluorescence of BSA after binding with VDG show maximum changes in emission intensity at tryptophan (Trp) residues. Post binding with VDG, BSA conformation changes as suggested by circular dichorism (CD) spectra of BSA and this lead to enhanced protein stability as indicated by a thermal melting curve of BSA.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Piyush Verma
- CBRN Protection and Decontamination Research Group, Division of Radiological Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences, Defence R&D Organisation, Timarpur, Delhi, India
| | - Lajpreet Kaur
- CBRN Protection and Decontamination Research Group, Division of Radiological Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences, Defence R&D Organisation, Timarpur, Delhi, India
| | - Priyanka Aswal
- Department of Pharmaceutics, Veer Madho Singh Bhandari Uttarakhand Technical University, Dehradun, Uttarakhand, India
| | - Anju Singh
- Department of Chemistry, Ramjas College, University of Delhi, Delhi, India
| | - Himanshu Ojha
- CBRN Protection and Decontamination Research Group, Division of Radiological Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences, Defence R&D Organisation, Timarpur, Delhi, India
| | - Afreen Jahan Rahman
- CBRN Protection and Decontamination Research Group, Division of Radiological Nuclear and Imaging Sciences (RNAIS), Institute of Nuclear Medicine and Allied Sciences, Defence R&D Organisation, Timarpur, Delhi, India
| | - Rahul Singhal
- Department of Chemistry, Shivaji College, University of Delhi, Delhi, India
| | - Anjani K Tiwari
- Department of Chemistry, Baba Bhim Rao Ambedkar University, Lucknow, India
| | - Mallika Pathak
- Department of Chemistry, Miranda House, University of Delhi, Delhi, India
| |
Collapse
|
6
|
Pascual Alonso I, Valiente PA, Valdés-Tresanco ME, Arrebola Y, Almeida García F, Díaz L, García G, Guirola O, Pastor D, Bergado G, Sánchez B, Charli JL. Discovery of tight-binding competitive inhibitors of dipeptidyl peptidase IV. Int J Biol Macromol 2022; 196:120-130. [PMID: 34920066 DOI: 10.1016/j.ijbiomac.2021.12.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 11/24/2022]
Abstract
Dipeptidyl peptidase IV (DPP-IV, EC 3.4.14.5) is an abundant serine aminopeptidase that preferentially cleaves N-terminal Xaa-Pro or Xaa-Ala dipeptides from oligopeptides. Inhibitors of DPP-IV activity are used for treating type 2 diabetes mellitus and other diseases. DPP-IV is also involved in tumor progression. We identified four new non-peptide tight-binding competitive inhibitors of porcine DPP-IV by virtual screening and enzymatic assays. Molecular docking simulations supported the competitive behavior, and the selectivity of one of the compounds in the DPP-IV family. Since three of these inhibitors are also aminopeptidase N (APN) inhibitors, we tested their impact on APN+/DPP-IV+ and DPP-IV+ human tumor cells' viability. Using kinetic assays, we determined that HL-60 tumor cells express both APN and DPP-IV activities and that MDA-MB-231 tumor cells express DPP-IV activity. The inhibitors had a slight inhibitory effect on human HEK-293 cell viability but reduced the viability of APN+/DPP-IV+ and DPP-IV+ human tumor cells more potently. Remarkably, the intraperitoneal injection of these compounds inhibited DPP-IV activity in rat brain, liver, and pancreas. In silico studies suggested inhibitors binding to serum albumin contribute to blood-brain barrier crossing. The spectrum of action of some of these compounds may be useful for niche applications.
Collapse
Affiliation(s)
| | - Pedro A Valiente
- Center for Protein Studies, Faculty of Biology, University of Havana, Cuba; Donnelly Center for Cellular and Biomolecular Research, University of Toronto, Canada.
| | - Mario E Valdés-Tresanco
- Center for Protein Studies, Faculty of Biology, University of Havana, Cuba; Department of Biological Sciences, University of Calgary, Calgary, Canada
| | - Yarini Arrebola
- Center for Protein Studies, Faculty of Biology, University of Havana, Cuba
| | | | - Lisset Díaz
- Center for Protein Studies, Faculty of Biology, University of Havana, Cuba
| | - Gabriela García
- Center for Protein Studies, Faculty of Biology, University of Havana, Cuba
| | - Osmany Guirola
- Centro de Ingeniería Genética y Biotecnología, BioCubafarma, Cuba
| | - Daniel Pastor
- Center for Protein Studies, Faculty of Biology, University of Havana, Cuba
| | | | | | - Jean-Louis Charli
- Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Mexico
| |
Collapse
|
7
|
Therapeutic Potential of Seaweed-Derived Bioactive Compounds for Cardiovascular Disease Treatment. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12031025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Cardiovascular diseases are closely related to hypertension, type 2 diabetes mellitus, obesity, and hyperlipidemia. Many studies have reported that an unhealthy diet and sedentary lifestyle are critical factors that enhance these diseases. Recently, many bioactive compounds isolated from marine seaweeds have been studied for their benefits in improving human health. In particular, several unique bioactive metabolites such as polyphenols, polysaccharides, peptides, carotene, and sterol are the most effective components responsible for these activities. This review summarizes the current in vitro, in vivo, and clinical studies related to the protective effects of bioactive compounds isolated from seaweeds against cardiovascular disorders, including anti-diabetic, anti-hypertensive, anti-hyperlipidemia, and anti-obesity effects. Therefore, this present review summarizes these concepts and provides a basis for further in-depth research.
Collapse
|
8
|
Construction of transplantable artificial vascular tissue based on adipose tissue-derived mesenchymal stromal cells by a cell coating and cryopreservation technique. Sci Rep 2021; 11:17989. [PMID: 34504254 PMCID: PMC8429436 DOI: 10.1038/s41598-021-97547-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 08/26/2021] [Indexed: 02/07/2023] Open
Abstract
Prevascularized artificial three-dimensional (3D) tissues are effective biomaterials for regenerative medicine. We have previously established a scaffold-free 3D artificial vascular tissue from normal human dermal fibroblasts (NHDFs) and umbilical vein-derived endothelial cells (HUVECs) by layer-by-layer cell coating technique. In this study, we constructed an artificial vascular tissue constructed by human adipose tissue-derived stromal cells (hASCs) and HUVECs (ASCVT) by a modified technique with cryopreservation. ASCVT showed a higher thickness with more dense vascular networks than the 3D tissue based on NHDFs. Correspondingly, 3D-cultured ASCs showed higher expression of several angiogenesis-related factors, including vascular endothelial growth factor-A and hepatic growth factor, compared to that of NHDFs. Moreover, perivascular cells in ASCVT were detected by pericyte markers, suggesting the differentiation of hASCs into pericyte-like cells. Subcutaneous transplantation of ASCVTs to nude mice resulted in an engraftment with anastomosis of host's vascular structures at 2 weeks after operation. In the engrafted tissue, the vascular network was surrounded by mural-like structure-forming hASCs, in which some parts developed to form vein-like structures at 4 weeks, suggesting the generation of functional vessel networks. These results demonstrated that cryopreserved human cells, including hASCs, could be used directly to construct the artificial transplantable tissue for regenerative medicine.
Collapse
|
9
|
Zhang S, Li P, Xin M, Jin X, Zhao L, Nan Y, Cheng XW. Dipeptidyl peptidase-4 inhibition prevents lung injury in mice under chronic stress via the modulation of oxidative stress and inflammation. Exp Anim 2021; 70:541-552. [PMID: 34219073 PMCID: PMC8614009 DOI: 10.1538/expanim.21-0067] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Exposure to chronic psychosocial stress is a risk factor for various pulmonary diseases. In view of the essential role of dipeptidyl peptidase 4 (DPP4) in animal and human lung pathobiology, we investigated the role of DPP4 in stress-related lung injury in mice. Eight-week-old male mice were randomly divided into a non-stress group and a 2-week immobilization stress group. Non-stress control mice were left undisturbed. The mice subjected to immobilized stress were randomly assigned to the vehicle or the DPP4 inhibitor anagliptin for 2 weeks. Chronic stress reduced subcutaneous and inguinal adipose volumes and increased blood DPP4 levels. The stressed mice showed increased levels in the lungs of genes and/or proteins related to oxidative stress (p67phox, p47phox, p22phox and gp91phox), inflammation (monocyte chemoattractant protein-1, vascular cell adhesion molecule-1, and intracellular adhesion molecule-1), apoptosis (caspase-3, -8, -9), senescence (p16INK4A, p21, and p53) and proteolysis (matrix metalloproteinase-2 to -9, cathepsin S/K, and tissue inhibitor of matrix metalloproteinase-1 and -2), and reduced levels of eNOS, Sirt1, and Bcl-2 proteins; and these effects were reversed by genetic and pharmacological inhibitions of DPP4. We then exposed human umbilical vein endothelial cells in vitro to hydrogen peroxide; anagliptin treatment was also observed to mitigate oxidative and inflammatory molecules in this setting. Anagliptin can improve lung injury in stressed mice, possibly by mitigating vascular inflammation, oxidative stress production, and proteolysis. DPP4 may become a new therapeutic target for chronic psychological stress-related lung disease in humans and animals.
Collapse
Affiliation(s)
- Shengming Zhang
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Ping Li
- State Key Laboratory of Cardiovascular Disease, Fu Wai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union, Medical College
| | - Minglong Xin
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Xianglan Jin
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Longguo Zhao
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Yongshan Nan
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| | - Xian Wu Cheng
- Department of Anesthesiology and Cardiology, Yanbian University Hospital
| |
Collapse
|
10
|
Increased dipeptidyl peptidase-4 accelerates chronic stress-related thrombosis in a mouse carotid artery model. J Hypertens 2021; 38:1504-1513. [PMID: 32205561 DOI: 10.1097/hjh.0000000000002418] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Exposure to chronic psychosocial stress is a risk factor for metabolic cardiovascular disorders. Given that dipeptidyl peptidase-4 (DPP-4) has an important role in human pathobiology, we investigated the role of DPP-4 in stress-related thrombosis in mice, focusing on oxidative stress and the von Willebrand factor (vWF)-cleaving protease ADAMTS13 (a disintegrin and metalloproteinase with thrombospondin type 1 motif, member 13). METHODS AND RESULTS Male mice randomly assigned to nonstress and 2-week immobilized-stress groups underwent iron chloride3 (FeCl3)-induced carotid artery thrombosis surgery for morphological and biochemical studies at specific times. On day 14 post-stress/surgery, stress had enhanced the lengths and weights of arterial thrombi, with alterations of plasma DPP-4, plasminogen activation inhibitor-1 and ADAMTS13. The stressed mice had increased levels of vascular cell adhesion molecule-1, intracellular adhesion molecule-1, monocyte chemoattractant protein-1, gp91phox, p22phox, matrix metalloproteinase-2 (MMP-2), MMP-9, cathepsins S and K mRNAs and/or proteins, and reduced levels of endothelial nitric oxide synthase, catalase and superoxide dismutase-1 mRNAs and/or proteins. Stress also accelerated arterial endothelial cell damage. The DPP-4 inhibitor anagliptin ameliorated the stress-induced targeted molecular and morphological changes and thrombosis. In vitro, DPP-4 inhibition also mitigated the alterations in the targeted ADAMTS13 and other oxidative and inflammatory molecules in human umbilical vein endothelial cells in response to H2O2. CONCLUSION DPP-4 inhibition appeared to improve the FeCl3-induced thrombosis in mice that received stress, possibly via the improvement of ADAMTS13 and oxidative stress, suggesting that DPP-4 could become a novel therapeutic target for chronic psychological stress-related thrombotic events in metabolic cardiovascular disorders.
Collapse
|
11
|
Piao L, Li Y, Narisawa M, Shen X, Cheng XW. Role of Dipeptidyl Peptidase-4 in Atherosclerotic Cardiovascular Disease in Humans and Animals with Chronic Stress. Int Heart J 2021; 62:470-478. [PMID: 33994495 DOI: 10.1536/ihj.20-181] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Exposure to psychosocial stress is a risk factor for cardiovascular disease, including vascular atherosclerosis-based cardiovascular disease (ACVD). Dipeptidyl peptidase-4 (DPP-4) is a complex enzyme that acts as a membrane-anchored cell surface exopeptidase. DPP-4 is upregulated in metabolic and inflammatory cardiovascular disorders. DPP-4 exhibits many physiological and pharmacological functions by regulating its extremely abundant substrates, such as glucagon-like peptide-1 (GLP-1). Over the last 10 years, emerging data have demonstrated unexpected roles of DPP-4 in extracellular and intracellular signaling, immune activation, inflammation, oxidative stress production, cell apoptosis, insulin resistance, and lipid metabolism. This mini-review focuses on recent novel findings in this field, highlighting a DPP-4-mediated regulation of GLP-1-dependent and -independent signaling pathways as a potential therapeutic molecular target in treatments of chronic psychological stress-related ACVD in humans and animals.
Collapse
Affiliation(s)
- Limei Piao
- Department of Cardiology, Yanbian University Hospital
| | - Yanglong Li
- Department of Cardiology, Yanbian University Hospital
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Xionghu Shen
- Department of Oncology, Yanbian University Hospital
| | - Xian Wu Cheng
- Department of Cardiology, Yanbian University Hospital
| |
Collapse
|
12
|
Wang M, Liu Y, Liang Y, Naruse K, Takahashi K. Systematic Understanding of Pathophysiological Mechanisms of Oxidative Stress-Related Conditions-Diabetes Mellitus, Cardiovascular Diseases, and Ischemia-Reperfusion Injury. Front Cardiovasc Med 2021; 8:649785. [PMID: 33928135 PMCID: PMC8076504 DOI: 10.3389/fcvm.2021.649785] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/22/2021] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species (ROS) plays a role in intracellular signal transduction under physiological conditions while also playing an essential role in diseases such as hypertension, ischemic heart disease, and diabetes, as well as in the process of aging. The influence of ROS has some influence on the frequent occurrence of cardiovascular diseases (CVD) in diabetic patients. In this review, we considered the pathophysiological relationship between diabetes and CVD from the perspective of ROS. In addition, considering organ damage due to ROS elevation during ischemia-reperfusion, we discussed heart and lung injuries. Furthermore, we have focused on the transient receptor potential (TRP) channels and L-type calcium channels as molecular targets for ROS in ROS-induced tissue damages and have discussed about the pathophysiological mechanism of the injury.
Collapse
Affiliation(s)
| | | | | | | | - Ken Takahashi
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
13
|
Couturier A, Blot G, Vignaud L, Nanteau C, Slembrouck-Brec A, Fradot V, Acar N, Sahel JA, Tadayoni R, Thuret G, Sennlaub F, Roger JE, Goureau O, Guillonneau X, Reichman S. Reproducing diabetic retinopathy features using newly developed human induced-pluripotent stem cell-derived retinal Müller glial cells. Glia 2021; 69:1679-1693. [PMID: 33683746 PMCID: PMC8252429 DOI: 10.1002/glia.23983] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 12/13/2022]
Abstract
Muller glial cells (MGCs) are responsible for the homeostatic and metabolic support of the retina. Despite the importance of MGCs in retinal disorders, reliable and accessible human cell sources to be used to model MGC-associated diseases are lacking. Although primary human MGCs (pMGCs) can be purified from post-mortem retinal tissues, the donor scarcity limits their use. To overcome this problem, we developed a protocol to generate and bank human induced pluripotent stem cell-derived MGCs (hiMGCs). Using a transcriptome analysis, we showed that the three genetically independent hiMGCs generated were homogeneous and showed phenotypic characteristics and transcriptomic profile of pMGCs. These cells expressed key MGC markers, including Vimentin, CLU, DKK3, SOX9, SOX2, S100A16, ITGB1, and CD44 and could be cultured up to passage 8. Under our culture conditions, hiMGCs and pMGCs expressed low transcript levels of RLPB1, AQP4, KCNJ1, KCJN10, and SLC1A3. Using a disease modeling approach, we showed that hiMGCs could be used to model the features of diabetic retinopathy (DR)-associated dyslipidemia. Indeed, palmitate, a major free fatty acid with elevated plasma levels in diabetic patients, induced the expression of inflammatory cytokines found in the ocular fluid of DR patients such as CXCL8 (IL-8) and ANGPTL4. Moreover, the analysis of palmitate-treated hiMGC secretome showed an upregulation of proangiogenic factors strongly related to DR, including ANG2, Endoglin, IL-1β, CXCL8, MMP-9, PDGF-AA, and VEGF. Thus, hiMGCs could be an alternative to pMGCs and an extremely valuable tool to help to understand and model glial cell involvement in retinal disorders, including DR.
Collapse
Affiliation(s)
- Aude Couturier
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France.,Department of Ophthalmology, Hôpital Lariboisière, AP-HP, Université de Paris, Paris, France
| | - Guillaume Blot
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Lucile Vignaud
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Céline Nanteau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - Valérie Fradot
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Niyazi Acar
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, Dijon, France
| | - José-Alain Sahel
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France.,CHNO des Quinze-Vingts, DHU Sight Restore, INSERM-DGOS CIC 1423, Paris, France.,Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ramin Tadayoni
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France.,Department of Ophthalmology, Hôpital Lariboisière, AP-HP, Université de Paris, Paris, France
| | - Gilles Thuret
- Biologie, Ingénierie et Imagerie de la Greffe de Cornée, EA2521, Faculté de Médecine, Université Jean Monnet, Saint-Etienne, France
| | - Florian Sennlaub
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | - Jerome E Roger
- Institut des Neurosciences Paris-Saclay, CERTO-Retina France, CNRS, Univ Paris-Saclay, Orsay, France
| | - Olivier Goureau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| | | | - Sacha Reichman
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, Paris, France
| |
Collapse
|
14
|
Cao F, Wu K, Zhu YZ, Bao ZW. Roles and Mechanisms of Dipeptidyl Peptidase 4 Inhibitors in Vascular Aging. Front Endocrinol (Lausanne) 2021; 12:731273. [PMID: 34489872 PMCID: PMC8416540 DOI: 10.3389/fendo.2021.731273] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/21/2021] [Indexed: 12/22/2022] Open
Abstract
Vascular aging is characterized by alterations in the constitutive properties and biological functions of the blood vessel wall. Endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) are indispensability elements in the inner layer and the medial layer of the blood vessel wall, respectively. Dipeptidyl peptidase-4 (DPP4) inhibitors, as a hypoglycemic agent, play a protective role in reversing vascular aging regardless of their effects in meliorating glycemic control in humans and animal models of type 2 diabetes mellitus (T2DM) through complex cellular mechanisms, including improving EC dysfunction, promoting EC proliferation and migration, alleviating EC senescence, obstructing EC apoptosis, suppressing the proliferation and migration of VSMCs, increasing circulating endothelial progenitor cell (EPC) levels, and preventing the infiltration of mononuclear macrophages. All of these showed that DPP4 inhibitors may exert a positive effect against vascular aging, thereby preventing vascular aging-related diseases. In the current review, we will summarize the cellular mechanism of DPP4 inhibitors regulating vascular aging; moreover, we also intend to compile the roles and the promising therapeutic application of DPP4 inhibitors in vascular aging-related diseases.
Collapse
Affiliation(s)
- Fen Cao
- Department of Cardiology, Huaihua First People’s Hospital, Huaihua, China
| | - Kun Wu
- Department of Neurology, Huaihua First People’s Hospital, Huaihua, China
| | - Yong-Zhi Zhu
- Department of Cardiology, Huaihua First People’s Hospital, Huaihua, China
| | - Zhong-Wu Bao
- Department of Cardiology, Huaihua First People’s Hospital, Huaihua, China
- *Correspondence: Zhong-Wu Bao,
| |
Collapse
|
15
|
Masre SF, Jufri NF, Ibrahim FW, Abdul Raub SH. Classical and alternative receptors for SARS-CoV-2 therapeutic strategy. Rev Med Virol 2020; 31:1-9. [PMID: 33368788 PMCID: PMC7883063 DOI: 10.1002/rmv.2207] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 01/08/2023]
Abstract
Understanding the molecules that are essential for severe acute respiratory syndrome coronavirus‐2 (SARS‐CoV‐2) entry can provide insights into viral infection and dissemination. Recently, it has been identified from several studies that angiotensin‐converting enzyme 2 receptor and transmembrane serine protease 2 are the main entry molecules for the SARS‐CoV‐2, which produced the pandemic of Covid‐19. However, additional evidence showed several other viral receptors and cellular proteases that are also important in facilitating viral entry and transmission in the target cells. In this review, we summarized the types of SARS‐CoV‐2 entry molecules and discussed their crucial roles for virus binding, protein priming and fusion to the cellular membrane important for SARS‐CoV‐2 infection.
Collapse
Affiliation(s)
- Siti Fathiah Masre
- Faculty of Health Sciences, Centre for Toxicology and Health Risk Studies, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nurul Farhana Jufri
- Faculty of Health Sciences, Centre for Toxicology and Health Risk Studies, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Farah Wahida Ibrahim
- Faculty of Health Sciences, Centre for Toxicology and Health Risk Studies, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Sayyidi Hamzi Abdul Raub
- Pantai Premier Pathology SDN BHD, Reference Specialised Laboratory, Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| |
Collapse
|
16
|
Bestatin and bacitracin inhibit porcine kidney cortex dipeptidyl peptidase IV activity and reduce human melanoma MeWo cell viability. Int J Biol Macromol 2020; 164:2944-2952. [DOI: 10.1016/j.ijbiomac.2020.08.157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 01/10/2023]
|
17
|
Paganelli F, Gaudry M, Ruf J, Guieu R. Recent advances in the role of the adenosinergic system in coronary artery disease. Cardiovasc Res 2020; 117:1284-1294. [PMID: 32991685 DOI: 10.1093/cvr/cvaa275] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/14/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022] Open
Abstract
Adenosine is an endogenous nucleoside that plays a major role in the physiology and physiopathology of the coronary artery system, mainly by activating its A2A receptors (A2AR). Adenosine is released by myocardial, endothelial, and immune cells during hypoxia, ischaemia, or inflammation, each condition being present in coronary artery disease (CAD). While activation of A2AR improves coronary blood circulation and leads to anti-inflammatory effects, down-regulation of A2AR has many deleterious effects during CAD. A decrease in the level and/or activity of A2AR leads to: (i) lack of vasodilation, which decreases blood flow, leading to a decrease in myocardial oxygenation and tissue hypoxia; (ii) an increase in the immune response, favouring inflammation; and (iii) platelet aggregation, which therefore participates, in part, in the formation of a fibrin-platelet thrombus after the rupture or erosion of the plaque, leading to the occurrence of acute coronary syndrome. Inflammation contributes to the development of atherosclerosis, leading to myocardial ischaemia, which in turn leads to tissue hypoxia. Therefore, a vicious circle is created that maintains and aggravates CAD. In some cases, studying the adenosinergic profile can help assess the severity of CAD. In fact, inducible ischaemia in CAD patients, as assessed by exercise stress test or fractional flow reserve, is associated with the presence of a reserve of A2AR called spare receptors. The purpose of this review is to present emerging experimental evidence supporting the existence of this adaptive adenosinergic response to ischaemia or inflammation in CAD. We believe that we have achieved a breakthrough in the understanding and modelling of spare A2AR, based upon a new concept allowing for a new and non-invasive CAD management.
Collapse
Affiliation(s)
- Franck Paganelli
- C2VN, INSERM, INRAE, Aix-Marseille University, Campus Santé Timone, Faculté de Pharmacie, 27 Bd Jean Moulin, F-13005 Marseille, France.,Department of Cardiology, North Hospital, Chemin des Bourrely, F-13015 Marseille, France
| | - Marine Gaudry
- Department of Vascular Surgery, Timone Hospital, 278 Rue Saint Pierre, F-13005 Marseille, France
| | - Jean Ruf
- C2VN, INSERM, INRAE, Aix-Marseille University, Campus Santé Timone, Faculté de Pharmacie, 27 Bd Jean Moulin, F-13005 Marseille, France
| | - Régis Guieu
- C2VN, INSERM, INRAE, Aix-Marseille University, Campus Santé Timone, Faculté de Pharmacie, 27 Bd Jean Moulin, F-13005 Marseille, France.,Laboratory of Biochemistry, Timone Hospital, 278 Rue Saint Pierre, F-13005 Marseille, France
| |
Collapse
|
18
|
Solerte SB, Di Sabatino A, Galli M, Fiorina P. Dipeptidyl peptidase-4 (DPP4) inhibition in COVID-19. Acta Diabetol 2020; 57:779-783. [PMID: 32506195 PMCID: PMC7275134 DOI: 10.1007/s00592-020-01539-z] [Citation(s) in RCA: 146] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 04/18/2020] [Indexed: 02/08/2023]
Abstract
AIMS SARS-CoV-2 causes severe respiratory syndrome (COVID-19) with high mortality due to a direct cytotoxic viral effect and a severe systemic inflammation. We are herein discussing a possible novel therapeutic tool for COVID-19. METHODS Virus binds to the cell surface receptor ACE2; indeed, recent evidences suggested that SARS-CoV-2 may be using as co-receptor, when entering the cells, the same one used by MERS-Co-V, namely the DPP4/CD26 receptor. The aforementioned observation underlined that mechanism of cell entry is supposedly similar among different coronavirus, that the co-expression of ACE2 and DPP4/CD26 could identify those cells targeted by different human coronaviruses and that clinical complications may be similar. RESULTS The DPP4 family/system was implicated in various physiological processes and diseases of the immune system, and DPP4/CD26 is variously expressed on epithelia and endothelia of the systemic vasculature, lung, kidney, small intestine and heart. In particular, DPP4 distribution in the human respiratory tract may facilitate the entrance of the virus into the airway tract itself and could contribute to the development of cytokine storm and immunopathology in causing fatal COVID-19 pneumonia. CONCLUSIONS The use of DPP4 inhibitors, such as gliptins, in patients with COVID-19 with, or even without, type 2 diabetes, may offer a simple way to reduce the virus entry and replication into the airways and to hamper the sustained cytokine storm and inflammation within the lung in patients diagnosed with COVID-19 infection.
Collapse
Affiliation(s)
- Sebastiano Bruno Solerte
- Geriatric and Diabetology Unit, Department of Internal Medicine, University of Pavia, Pavia, Italy
| | - Antonio Di Sabatino
- Internal Medicine Unit, University of Pavia and IRCCS Policlinico San Matteo, Pavia, Italy
| | - Massimo Galli
- Department of Biomedical, Clinical Sciences 'Luigi Sacco', University of Milan, Milan, Italy
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Paolo Fiorina
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, DIBIC L. Sacco, Università Degli Studi di Milano, Milan, Italy.
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy.
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
19
|
Ratushnyy A, Ezdakova M, Buravkova L. Secretome of Senescent Adipose-Derived Mesenchymal Stem Cells Negatively Regulates Angiogenesis. Int J Mol Sci 2020; 21:ijms21051802. [PMID: 32151085 PMCID: PMC7084202 DOI: 10.3390/ijms21051802] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/28/2020] [Accepted: 03/02/2020] [Indexed: 12/17/2022] Open
Abstract
Nowadays, paracrine regulation is considered as a major tool of mesenchymal stem cell (MSC) involvement in tissue repair and renewal in adults. Aging results in alteration of tissue homeostasis including neovascularization. In this study, we examined the influence of replicative senescence on the angiogenic potential of adipose-derived MSCs (ASCs). Angiogenic activity of conditioned medium (CM) from senescent and “young” ASCs was evaluated in chorioallantoic membrane (CAM) assay in ovo using Japanese quail embryos. Also, the formation of capillary-like tubes by human umbilical vein endothelial cells (HUVECs) in 3D basement membrane matrix “Matrigel” and HUVEC migration capacity were analyzed. Multiplex, dot-blot and gene expression analysis were performed to characterize transcription and production of about 100 angiogenesis-associated proteins. The results point to decreased angiogenic potential of senescent ASC secretome in ovo. A number of angiogenesis-associated proteins demonstrated elevation in CM after long-term cultivation. Meanwhile, VEGF (key positive regulator of angiogenesis) did not change transcription level and concentration in CM. Increasing both pro- (FGF-2, uPA, IL-6, IL-8 etc.) and antiangiogenic (IL-4, IP-10, PF4, Activin A, DPPIV etc.) factors was observed. Some proangiogenic genes were downregulated (IGF1, MMP1, TGFB3, PDGFRB, PGF). Senescence-associated secretory phenotype (SASP) modifications after long-term cultivation lead to attenuation of angiogenic potential of ASC.
Collapse
|
20
|
Xin M, Jin X, Cui X, Jin C, Piao L, Wan Y, Xu S, Zhang S, Yue X, Wang H, Nan Y, Cheng X. Dipeptidyl peptidase-4 inhibition prevents vascular aging in mice under chronic stress: Modulation of oxidative stress and inflammation. Chem Biol Interact 2019; 314:108842. [DOI: 10.1016/j.cbi.2019.108842] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 09/29/2019] [Indexed: 12/24/2022]
|
21
|
Qi Y, Du X, Yao X, Zhao Y. Vildagliptin inhibits high free fatty acid (FFA)-induced NLRP3 inflammasome activation in endothelial cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:1067-1074. [PMID: 30945564 DOI: 10.1080/21691401.2019.1578783] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Elevated free fatty acids (FFAs) are a risk factor for type 2 diabetes. Endothelial dysfunction induced by high levels of FFAs is one of the mechanisms related to the progression of diabetes. In clinical diabetes care, DPP-4 inhibitors have been shown to be effective in reducing glucose levels. In this study, we investigated the molecular mechanism of the clinically available DPP-4 inhibitor vildagliptin in the protection of FFA-induced endothelial dysfunction. Treatment of endothelial cells with vildagliptin inhibits FFA-induced cellular LDH release and generation of ROS. Vildagliptin also reverses FFA-induced reduced levels of GSH and elevated expression of the FFA-associated NAPHD oxidase protein NOX-4. Moreover, vildagliptin ameliorates the reduction in mitochondrial potential triggered by FFAs. Mechanistically, we show that vildagliptin suppresses FFA-induced expression of proteins of the NLRP3 inflammasome complex, including NLRP3, ASC, p20 and HMGB-1, and mitigates FFA-induced inactivation of the AMPK pathway. Consequently, vildagliptin inhibits production of two cytokines that are favored by NLRP3 inflammasome machinery: IL-1β and IL-18. Finally, we demonstrate that vildagliptin ameliorates FFA-induced reduced eNOS, indicating its protective role against endothelial dysfunction. Collectively, we conclude that the protective role of vildagliptin in endothelial cells is mediated via suppression of the AMPK-NLRP3 inflammasome-HMGB-1 axis pathway. These findings imply that the anti-diabetic drug vildagliptin possesses dual therapeutic applications in lowering glucose and improving vascular function.
Collapse
Affiliation(s)
- Yanyan Qi
- a Department of Anesthesiology , Henan Province People's Hospital , Zhengzhou , Henan , China
| | - Xianhui Du
- a Department of Anesthesiology , Henan Province People's Hospital , Zhengzhou , Henan , China
| | - Xiangyan Yao
- a Department of Anesthesiology , Henan Province People's Hospital , Zhengzhou , Henan , China
| | - Yuanyuan Zhao
- b Department of Cardiology , Qilu Hospital of Shandong University , Qingdao , Shandong , China
| |
Collapse
|
22
|
Kajikawa M, Maruhashi T, Hidaka T, Matsui S, Hashimoto H, Takaeko Y, Nakano Y, Kurisu S, Kihara Y, Yusoff FM, Kishimoto S, Chayama K, Goto C, Noma K, Nakashima A, Hiro T, Hirayama A, Shiina K, Tomiyama H, Yagi S, Amano R, Yamada H, Sata M, Higashi Y. Effect of Saxagliptin on Endothelial Function in Patients with Type 2 Diabetes: A Prospective Multicenter Study. Sci Rep 2019; 9:10206. [PMID: 31308448 PMCID: PMC6629702 DOI: 10.1038/s41598-019-46726-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 07/04/2019] [Indexed: 02/06/2023] Open
Abstract
The dipeptidyl peptidase-4 inhibitor saxagliptin is a widely used antihyperglycemic agent in patients with type 2 diabetes. The purpose of this study was to evaluate the effects of saxagliptin on endothelial function in patients with type 2 diabetes. This was a prospective, multicenter, interventional study. A total of 34 patients with type 2 diabetes were enrolled at four university hospitals in Japan. Treatment of patients was initially started with saxagliptin at a dose of 5 mg daily. Assessment of endothelial function assessed by flow-mediated vasodilation (FMD) and measurement of stromal cell-derived factor-1α (SDF-1α) were conducted at baseline and at 3 months after treatment with saxagliptin. A total of 31 patients with type 2 diabetes were included in the analysis. Saxagliptin significantly increased FMD from 3.1 ± 3.1% to 4.2 ± 2.4% (P = 0.032) and significantly decreased total cholesterol from 190 ± 24 mg/dL to 181 ± 25 mg/dL (P = 0.002), glucose from 160 ± 53 mg/dL to 133 ± 25 mg/dL (P < 0.001), HbA1c from 7.5 ± 0.6% to 7.0 ± 0.6% (P < 0.001), urine albumin-to-creatinine ratio from 63.8 ± 134.2 mg/g to 40.9 ± 83.0 mg/g (P = 0.043), and total SDF-1α from 2108 ± 243 pg/mL to 1284 ± 345 pg/mL (P < 0.001). These findings suggest that saxagliptin is effective for improving endothelial function.
Collapse
Affiliation(s)
- Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
| | - Tatsuya Maruhashi
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takayuki Hidaka
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shogo Matsui
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Haruki Hashimoto
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuji Takaeko
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yukiko Nakano
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Satoshi Kurisu
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yasuki Kihara
- Department of Cardiovascular Medicine, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Farina Mohamad Yusoff
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Shinji Kishimoto
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Institute of Biomedical and Health Sciences, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Chikara Goto
- Department of Physical Therapy, Hiroshima International University, Hiroshima, Japan
| | - Kensuke Noma
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Ayumu Nakashima
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Takafumi Hiro
- Division of Cardiology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Atsushi Hirayama
- Division of Cardiology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Kazuki Shiina
- Department of Cardiology, Tokyo Medical University, Tokyo, Japan
| | | | - Shusuke Yagi
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Rie Amano
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Hirotsugu Yamada
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Masataka Sata
- Department of Cardiovascular Medicine, Institute of Health Biosciences, The University of Tokushima Graduate School, Tokushima, Japan
| | - Yukihito Higashi
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima, Japan.
- Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
23
|
McHale C, Mohammed Z, Gomez G. Human Skin-Derived Mast Cells Spontaneously Secrete Several Angiogenesis-Related Factors. Front Immunol 2019; 10:1445. [PMID: 31293594 PMCID: PMC6603178 DOI: 10.3389/fimmu.2019.01445] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 06/10/2019] [Indexed: 12/12/2022] Open
Abstract
Mast cells are classically recognized as cells that cause IgE-mediated allergic reactions. However, their ability to store and secrete vascular endothelial growth factor (VEGF) suggests a role in vascular development and tumorigenesis. The current study sought to determine if other angiogenesis-related factors, in addition to VEGF, were also secreted by human tissue-derived mast cells. Using proteome array analysis and ELISA, we found that human skin-derived mast cells spontaneously secrete CXCL16, DPPIV, Endothelin-1, GM-CSF, IL-8, MCP-1, Pentraxin 3, Serpin E1, Serpin F1, TIMP-1, Thrombospondin-1, and uPA. We identified three groups based on their dependency for stem cell factor (SCF), which is required for mast cell survival: Endothelin-1, GM-CSF, IL-8, MCP-1, and VEGF (dependent); Pentraxin 3, Serpin E1, Serpin F1, TIMP-1, and Thrombospondin-1 (partly dependent); and CXCL16, DPPIV, and uPA (independent). Crosslinking of FcεRI with multivalent antigen enhanced the secretion of GM-CSF, Serpin E1, IL-8, and VEGF, and induced Amphiregulin and MMP-8 expression. Interestingly, FcεRI signals inhibited the spontaneous secretion of CXCL16, Endothelin-1, Serpin F1, Thrombospondin-1, MCP-1 and Pentraxin-3. Furthermore, IL-6, which we previously showed could induce VEGF, significantly enhanced MCP-1 secretion. Overall, this study identified several angiogenesis-related proteins that, in addition to VEGF, are spontaneously secreted at high concentrations from human skin-derived mast cells. These findings provide further evidence supporting an intrinsic role for mast cells in blood vessel formation.
Collapse
Affiliation(s)
- Cody McHale
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Zahraa Mohammed
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Gregorio Gomez
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| |
Collapse
|
24
|
Tsao A, Nakano T, Nowak AK, Popat S, Scagliotti GV, Heymach J. Targeting angiogenesis for patients with unresectable malignant pleural mesothelioma. Semin Oncol 2019; 46:145-154. [PMID: 31280996 DOI: 10.1053/j.seminoncol.2019.06.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/12/2019] [Indexed: 12/20/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a global health issue, the principal cause of which is exposure to asbestos. The prevalence is anticipated to rise over the next 2 decades, particularly in developing countries, due to the 30-50-year latency period between exposure to asbestos and carcinogenic development. Unresectable MPM has a poor prognosis and limited treatment options and, as such, there is a broad range of therapeutic targets of interest, including angiogenesis, immune checkpoints, mesothelin, as well as chemotherapeutic agents. Recently, the results of several randomized trials in the first-line setting combining antiangiogenic agents with chemotherapy have been reported. This review examines the scientific rationale for targeting angiogenesis in the treatment of unresectable MPM and analyzes recent clinical results with antiangiogenic agents in development (bevacizumab, nintedanib, and cediranib) for the management of MPM.
Collapse
Affiliation(s)
- Anne Tsao
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA.
| | - Takashi Nakano
- Division of Respiratory Medicine, Department of Internal Medicine, Otemae Hospital, Osaka, Japan
| | - Anna K Nowak
- School of Medicine, Faculty of Health and Medical Science, University of Western Australia, Crawley, Western Australia, Australia; Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Sanjay Popat
- Royal Marsden Hospital NHS Foundation Trust, London and Surrey, United Kingdom
| | | | - John Heymach
- Department of Thoracic and Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
25
|
Ma S, Bai Z, Wu H, Wang W. The DPP-4 inhibitor saxagliptin ameliorates ox-LDL-induced endothelial dysfunction by regulating AP-1 and NF-κB. Eur J Pharmacol 2019; 851:186-193. [PMID: 30639312 DOI: 10.1016/j.ejphar.2019.01.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/16/2018] [Accepted: 01/08/2019] [Indexed: 12/12/2022]
Abstract
Diabetes-associated cardiovascular complications are the leading cause of death for diabetic patients. Dipeptidyl peptidase 4 (DPP-4) inhibitor agents, known as gliptins, are a class of potent anti-glycemic agents developed to treat diabetes. Recently, gliptins have been shown to have independent cardiovascular benefits. In this study, we revealed the protective role of saxagliptin in vascular endothelial cells. Our data show that saxagliptin suppresses oxidized low-density lipoprotein cholesterol (ox-LDL)-induced expression of its receptor lectin-like ox-LDL receptor-1 (LOX-1). Saxagliptin treatment reduces ox-LDL-induced production of cytokines and vascular adhesion molecules including tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), vascular cell adhesion molecule 1 (VCAM-1), and intercellular cell adhesion molecule-1 (ICAM-1). The presence of saxagliptin suppressed ox-LDL-induced adhesion of monocytes to endothelial cells in co-culture adhesion experiments. Moreover, saxagliptin mitigated ox-LDL-induced production of reactive oxygen species and suppressed elevated expression of endothelial nicotinamide adenine dinucleotide phosphate oxidase subunit (NOX-4) induced by ox-LDL. Mechanistically, saxagliptin exerted inhibitory effects against ox-LDL-induced phosphorylation of JNK kinase, expression of the activator protein 1 (AP-1) subunits c-Jun/c-fos, and AP-1 promoter activity. Saxagliptin also suppressed nuclear factor κB (NF-κB) p65 accumulation and inhibited its promoter activity. Our data elaborate the molecular mechanism of saxagliptin-mediated endothelial protection and indicate that saxagliptin could have vascular benefits independent on its anti-glycemic function.
Collapse
Affiliation(s)
- Suxia Ma
- Cardiology Department 2, Shangqiu First People's Hospital, Shangqiu, Henan 476100, China
| | - Zhifeng Bai
- Cardiology Department 2, Shangqiu First People's Hospital, Shangqiu, Henan 476100, China.
| | - Huiying Wu
- Cardiology Department, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan 450007, China
| | - Wei Wang
- Cardiology Department, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan 450007, China
| |
Collapse
|
26
|
Zhang M, Jin X, Zhang Z, Li B, Yang G. Vildagliptin protects endothelial cells against high glucose-induced damage. Biomed Pharmacother 2018; 108:1790-1796. [DOI: 10.1016/j.biopha.2018.09.148] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 09/14/2018] [Accepted: 09/26/2018] [Indexed: 12/22/2022] Open
|
27
|
Kawasaki T, Chen W, Htwe YM, Tatsumi K, Dudek SM. DPP4 inhibition by sitagliptin attenuates LPS-induced lung injury in mice. Am J Physiol Lung Cell Mol Physiol 2018; 315:L834-L845. [PMID: 30188745 DOI: 10.1152/ajplung.00031.2018] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a severe clinical condition marked by acute respiratory failure and dysregulated inflammation. Pulmonary vascular endothelial cells (PVECs) function as an important pro-inflammatory source in ARDS, suggesting that modulation of inflammatory events at the endothelial level may have a therapeutic benefit. Dipeptidyl peptidase-4 (DPP4) inhibitors, widely used for the treatment of diabetes mellitus, have been reported to have possible anti-inflammatory effects. However, the potential anti-inflammatory effects of DPP4 inhibition on PVEC function and ARDS pathophysiology are unknown. Therefore, we evaluated the effects of sitagliptin, a DPP4 inhibitor in wide clinical use, on LPS-induced lung injury in mice and in human lung ECs in vitro. In vivo, sitagliptin reduced serum DPP4 activity, bronchoalveolar lavage protein concentration, cell number, and proinflammatory cytokine levels after LPS and alleviated histological findings of lung injury. LPS decreased the expression levels of CD26/DPP4 on pulmonary epithelial cells and PVECs isolated from mouse lungs, and the effect was partially reversed by sitagliptin. In vitro, human lung microvascular ECs (HLMVECs) expressed higher levels of CD26/DPP4 than human pulmonary arterial ECs. LPS induced the release of TNFα, IL-6, and IL-8 by HLMVECs that were inhibited by sitagliptin. LPS promoted the proliferation of HLMVECs, and sitagliptin suppressed this response. However, sitagliptin failed to reverse LPS-induced permeability in cultured ECs or lung epithelial cells in vitro. In summary, sitagliptin attenuates LPS-induced lung injury in mice and exerts anti-inflammatory effects on HLMVECs. These novel observations indicate DPP4 inhibitors may have potential as therapeutic drugs for ARDS.
Collapse
Affiliation(s)
- Takeshi Kawasaki
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois.,Department of Respirology, Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Weiguo Chen
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Yu Maw Htwe
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Steven M Dudek
- Division of Pulmonary, Critical Care, Sleep, and Allergy, Department of Medicine, University of Illinois at Chicago , Chicago, Illinois
| |
Collapse
|
28
|
Cheng XW, Narisawa M, Jin E, Yu C, Xu W, Piao L. Dose rectification of an imbalance between DPP4 and GLP-1 ameliorates chronic stress-related vascular aging and atherosclerosis? Clin Exp Pharmacol Physiol 2018; 45:467-470. [PMID: 29220092 DOI: 10.1111/1440-1681.12903] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/20/2017] [Accepted: 11/20/2017] [Indexed: 12/12/2022]
Abstract
Exposure to psychosocial stress is a risk factor for cardiovascular disease, including vascular aging and regeneration. Dipeptidyl peptidase-4 (DPP-4) exerts many physiological and pharmacological functions by regulating its extremely abundant substrates [eg., glucagon-like peptide-1 (GLP-1), stromal cell-derived factor-1α/C-X-C chemokine receptor type-4, etc.]. Over the past decade, emerging data has revealed unexpected roles for DPP-4 and GLP-1 in intracellular signaling, oxidative stress production, lipid metabolism, cell apoptosis, immune activation, insulin resistance, and inflammation. This mini review focuses on recent findings in this field, highlighting an imbalance between DPP4 and GLP-1 as a potential therapeutic target in the management of vascular aging and atherosclerosis in animals under experimental stress conditions.
Collapse
Affiliation(s)
- Xian Wu Cheng
- Department of Cardiology and Heart Center, Yanbian University Hospital, Yanji, Jilin, China.,Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, Republic of Korea.,Institute of Innovation for the Future Society, Nagoya University Graduate School of Medicine, Nagoya, Aichi Prefecture, Japan
| | - Megumi Narisawa
- Department of Cardiology, Tajimikenlitsu General Hospital, Tajimi, Gifu Prefecture, Japan
| | - Enze Jin
- Department of Cardiology, The Forth Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Chenglin Yu
- Department of Cardiology and Heart Center, Yanbian University Hospital, Yanji, Jilin, China
| | - Wenhu Xu
- Department of Cardiology and Heart Center, Yanbian University Hospital, Yanji, Jilin, China
| | - Limei Piao
- Department of Cardiology and Heart Center, Yanbian University Hospital, Yanji, Jilin, China
| |
Collapse
|
29
|
|
30
|
Piao L, Zhao G, Zhu E, Inoue A, Shibata R, Lei Y, Hu L, Yu C, Yang G, Wu H, Xu W, Okumura K, Ouchi N, Murohara T, Kuzuya M, Cheng XW. Chronic Psychological Stress Accelerates Vascular Senescence and Impairs Ischemia-Induced Neovascularization: The Role of Dipeptidyl Peptidase-4/Glucagon-Like Peptide-1-Adiponectin Axis. J Am Heart Assoc 2017; 6:e006421. [PMID: 28963101 PMCID: PMC5721852 DOI: 10.1161/jaha.117.006421] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 08/03/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND Exposure to psychosocial stress is a risk factor for cardiovascular disease, including vascular aging and regeneration. Given that dipeptidyl peptidase-4 (DPP4) regulates several intracellular signaling pathways associated with the glucagon-like peptide-1 (GLP-1) metabolism, we investigated the role of DPP4/GLP-1 axis in vascular senescence and ischemia-induced neovascularization in mice under chronic stress, with a special focus on adiponectin -mediated peroxisome proliferator activated receptor-γ/its co-activator 1α (PGC-1α) activation. METHODS AND RESULTS Seven-week-old mice subjected to restraint stress for 4 weeks underwent ischemic surgery and were kept under immobilization stress conditions. Mice that underwent ischemic surgery alone served as controls. We demonstrated that stress impaired the recovery of the ischemic/normal blood-flow ratio throughout the follow-up period and capillary formation. On postoperative day 4, stressed mice showed the following: increased levels of plasma and ischemic muscle DPP4 and decreased levels of GLP-1 and adiponectin in plasma and phospho-AMP-activated protein kinase α (p-AMPKα), vascular endothelial growth factor, peroxisome proliferator activated receptor-γ, PGC-1α, and Sirt1 proteins and insulin receptor 1 and glucose transporter 4 genes in the ischemic tissues, vessels, and/or adipose tissues and numbers of circulating endothelial CD31+/c-Kit+ progenitor cells. Chronic stress accelerated aortic senescence and impaired aortic endothelial sprouting. DPP4 inhibition and GLP-1 receptor activation improved these changes; these benefits were abrogated by adiponectin blocking and genetic depletion. CONCLUSIONS These results indicate that the DPP4/GLP-1-adiponectin axis is a novel therapeutic target for the treatment of vascular aging and cardiovascular disease under chronic stress conditions.
Collapse
MESH Headings
- Adiponectin/metabolism
- Animals
- Cells, Cultured
- Cellular Senescence
- Chronic Disease
- Dipeptidyl Peptidase 4/deficiency
- Dipeptidyl Peptidase 4/genetics
- Dipeptidyl Peptidase 4/metabolism
- Disease Models, Animal
- Endothelial Progenitor Cells/enzymology
- Endothelial Progenitor Cells/pathology
- Glucagon-Like Peptide 1/metabolism
- Ischemia/enzymology
- Ischemia/genetics
- Ischemia/pathology
- Ischemia/physiopathology
- Male
- Mice, Inbred C57BL
- Neovascularization, Physiologic
- PPAR gamma/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Proteolysis
- Rats, Inbred F344
- Rats, Transgenic
- Receptors, Adiponectin/metabolism
- Signal Transduction
- Stress, Psychological/enzymology
- Stress, Psychological/genetics
- Stress, Psychological/pathology
- Stress, Psychological/physiopathology
- Time Factors
- Tissue Culture Techniques
Collapse
Affiliation(s)
- Limei Piao
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Cardiology and ICU, Yanbian University Hospital, Yanji, Jilin Province, China
| | - Guangxian Zhao
- Cardiology and ICU, Yanbian University Hospital, Yanji, Jilin Province, China
| | - Enbo Zhu
- Cardiology and ICU, Yanbian University Hospital, Yanji, Jilin Province, China
| | - Aiko Inoue
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rei Shibata
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yanna Lei
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Cardiology and ICU, Yanbian University Hospital, Yanji, Jilin Province, China
| | - Lina Hu
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Public Health, Guilin Medical College, Guilin, Guangxi Province, China
| | - Chenglin Yu
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Cardiology and ICU, Yanbian University Hospital, Yanji, Jilin Province, China
| | - Guang Yang
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Cardiology and ICU, Yanbian University Hospital, Yanji, Jilin Province, China
| | - Hongxian Wu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Cardiology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenhu Xu
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Cardiology and ICU, Yanbian University Hospital, Yanji, Jilin Province, China
| | - Kenji Okumura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Noriyuki Ouchi
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masafumi Kuzuya
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xian Wu Cheng
- Department of Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Cardiology and ICU, Yanbian University Hospital, Yanji, Jilin Province, China
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
31
|
Zhu E, Hu L, Wu H, Piao L, Zhao G, Inoue A, Kim W, Yu C, Xu W, Bando YK, Li X, Lei Y, Hao CN, Takeshita K, Kim WS, Okumura K, Murohara T, Kuzuya M, Cheng XW. Dipeptidyl Peptidase-4 Regulates Hematopoietic Stem Cell Activation in Response to Chronic Stress. J Am Heart Assoc 2017; 6:JAHA.117.006394. [PMID: 28710180 PMCID: PMC5586325 DOI: 10.1161/jaha.117.006394] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Background DPP4 (Dipeptidyl peptidase‐4)‐GLP‐1 (glucagon‐like peptide‐1) and its receptor (GLP‐1R) axis has been involved in several intracellular signaling pathways. The Adrβ3 (β3‐adrenergic receptor)/CXCL12 (C‐X‐C motif chemokine 12) signal was required for the hematopoiesis. We investigated the novel molecular requirements between DPP4‐GLP‐1/GLP‐1 and Adrβ3/CXCL12 signals in bone marrow (BM) hematopoietic stem cell (HSC) activation in response to chronic stress. Methods and Results Male 8‐week‐old mice were subjected to 4‐week intermittent restrain stress and orally treated with vehicle or the DPP4 inhibitor anagliptin (30 mg/kg per day). Control mice were left undisturbed. The stress increased the blood and brain DPP4 levels, the plasma epinephrine and norepinephrine levels, and the BM niche cell Adrβ3 expression, and it decreased the plasma GLP‐1 levels and the brain GLP‐1R and BM CXCL12 expressions. These changes were reversed by DPP4 inhibition. The stress activated BM sca‐1highc‐KithighCD48lowCD150highHSC proliferation, giving rise to high levels of blood leukocytes and monocytes. The stress‐activated HSC proliferation was reversed by DPP4 depletion and by GLP‐1R activation. Finally, the selective pharmacological blocking of Adrβ3 mitigated HSC activation, accompanied by an improvement of CXCL12 gene expression in BM niche cells in response to chronic stress. Conclusions These findings suggest that DPP4 can regulate chronic stress‐induced BM HSC activation and inflammatory cell production via an Adrβ3/CXCL12‐dependent mechanism that is mediated by the GLP‐1/GLP‐1R axis, suggesting that the DPP4 inhibition or the GLP‐1R stimulation may have applications for treating inflammatory diseases.
Collapse
Affiliation(s)
- Enbo Zhu
- Department of Cardiology and ICU, Yanbian University Hospital, Yanji, China.,Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Lina Hu
- Department of Public Health, Guilin Medical College, Guilin Guangxi, China.,Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hongxian Wu
- Department of Cardiology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, China.,Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Limei Piao
- Department of Cardiology and ICU, Yanbian University Hospital, Yanji, China.,Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Guangxian Zhao
- Department of Cardiology and ICU, Yanbian University Hospital, Yanji, China
| | - Aiko Inoue
- Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Weon Kim
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea
| | - Chenglin Yu
- Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Wenhu Xu
- Department of Cardiology and ICU, Yanbian University Hospital, Yanji, China.,Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuko K Bando
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xiang Li
- Department of Cardiology and ICU, Yanbian University Hospital, Yanji, China.,Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yanna Lei
- Department of Cardiology and ICU, Yanbian University Hospital, Yanji, China
| | - Chang-Ning Hao
- Department of Vascular Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kyosuke Takeshita
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Woo-Shik Kim
- Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea
| | - Kenji Okumura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masafumi Kuzuya
- Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Xian Wu Cheng
- Department of Cardiology and ICU, Yanbian University Hospital, Yanji, China .,Department of and Community Healthcare & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya, Japan.,Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
32
|
Yang G, Lei Y, Inoue A, Piao L, Hu L, Jiang H, Sasaki T, Wu H, Xu W, Yu C, Zhao G, Ogasawara S, Okumura K, Kuzuya M, Cheng XW. Exenatide mitigated diet-induced vascular aging and atherosclerotic plaque growth in ApoE-deficient mice under chronic stress. Atherosclerosis 2017; 264:1-10. [PMID: 28734203 DOI: 10.1016/j.atherosclerosis.2017.07.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 07/03/2017] [Accepted: 07/12/2017] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND AIMS Exposure to psychosocial stress is a risk factor for cardiovascular disorders. Because the glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) agonist prevents cardiovascular injury, we investigated the beneficial effects and mechanism of the GLP-1 analogue exenatide on stress-related vascular senescence and atherosclerosis in apolipoprotein E-deficient (ApoE-/-) mice fed a high-fat (HF) diet. METHODS ApoE-/- mice fed the HF diet were assigned to non-stressed and immobilized-stress groups for 12 weeks. Mice fed the HF diet were divided into 2 groups and administered vehicle or exenatide for 12 weeks under stress conditions. RESULTS Chronic stress enhanced vascular endothelial senescence and atherosclerotic plaque growth. The stress increased the levels of plasma depeptidyl peptidase-4 activity and decreased the levels of plasma GLP-1 and both plasma and adipose adiponectin (APN). As compared with the mice subjected to stress alone, the exenatide-treated mice had decreased plaque microvessel density, macrophage accumulation, broken elastin, and enhanced plaque collagen volume, and lowered levels of peroxisome proliferator-activated receptor-α, gp91phox osteopontin, C-X-C chemokine receptor-4, toll-like receptor-2 (TLR2), TLR4, and cathepsins K, L, and S mRNAs and/or proteins. Exenatide reduced aortic matrix metalloproteinase-9 (MMP-9) and MMP-2 gene expression and activities. Exenatide also stimulated APN expression of preadipocytes and inhibited ox-low density lipoprotein-induced foam cell formation of monocytes in stressed mice. CONCLUSIONS These results indicate that the exenatide-mediated beneficial vascular actions are likely attributable, at least in part, to the enhancement of APN production and the attenuation of plaque oxidative stress, inflammation, and proteolysis in ApoE-/- mice under chronic stress.
Collapse
Affiliation(s)
- Guang Yang
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China
| | - Yanna Lei
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China
| | - Aiko Inoue
- Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan; Department of Community Health & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 4668550, Japan
| | - Limei Piao
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China; Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan
| | - Lina Hu
- Department of Public Health, Guilin Medical College, Guilin, Guangxi P. R., 541004, China
| | - Haiying Jiang
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China
| | - Takeshi Sasaki
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, 4313192, Japan
| | - Hongxian Wu
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Wenhu Xu
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China; Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan
| | - Chenglin Yu
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China; Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan
| | - Guangxian Zhao
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China
| | - Shinyu Ogasawara
- Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan
| | - Kenji Okumura
- Department of Cardiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Masafumi Kuzuya
- Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan; Department of Community Health & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 4668550, Japan
| | - Xian-Wu Cheng
- Department of Cardiology and ICU, Yanbian University Hospital, Yanjin, Jilin PR., 13000, China; Institute of Innovation for Future Society, Nagoya University, Nagoya, 4668550, Japan; Department of Community Health & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya, 4668550, Japan; Department of Internal Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
33
|
Li JW, Chen YD, Chen WR, You Q, Li B, Zhou H, Zhang Y, Han TW. Prognostic value of plasma DPP4 activity in ST-elevation myocardial infarction. Cardiovasc Diabetol 2017; 16:72. [PMID: 28587613 PMCID: PMC5461628 DOI: 10.1186/s12933-017-0553-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/20/2017] [Indexed: 01/03/2023] Open
Abstract
Background Dipeptidyl peptidase-4 (DPP4) regulates blood glucose levels and inflammation, and it is also implicated in the pathophysiological process of myocardial infarction (MI). Plasma DPP4 activity (DPP4a) may provide prognostic information regarding outcomes for ST-segment elevation MI (STEMI) patients. Methods Blood samples were obtained from 625 consecutively admitted, percutaneous coronary intervention-treated STEMI patients with a mean age of 57 years old. DPP4a was quantified using enzymatic assays. Results The median follow-up period was 30 months. Multivariate Cox-regression analyses (adjusted for confounding variables) showed that a 1 U/L increase of DPP4a did not associate with risks of major adverse cardiac or cerebrovascular events (MACCE), cardiovascular mortality, MI, heart failure readmission, stroke, non-cardiovascular mortality and repeated revascularization. However, in a subset of 149 diabetic STEMI patients, DPP4a associated with an increased risk of MACCE (HR 1.16; 95% CI 1.04–1.30; p = 0.01). Conclusions DPP4a did not associate with cardiovascular events and non-cardiovascular mortality in non-diabetic STEMI patients. However, DPP4a may be associated with future MACCE in diabetic STEMI patients. Trial registration NCT03046576, registered on 5 February, 2017, retrospectively registered Electronic supplementary material The online version of this article (doi:10.1186/s12933-017-0553-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jing-Wei Li
- Department of Cardiology, People's Liberation Army General Hospital, No. 28 Fuxing Road, Wukesong, Haidian District, Beijing, 100853, China.,Department of Cardiology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yun-Dai Chen
- Department of Cardiology, People's Liberation Army General Hospital, No. 28 Fuxing Road, Wukesong, Haidian District, Beijing, 100853, China.
| | - Wei-Ren Chen
- Department of Cardiology, People's Liberation Army General Hospital, No. 28 Fuxing Road, Wukesong, Haidian District, Beijing, 100853, China
| | - Qi You
- Department of Cardiology, People's Liberation Army General Hospital, No. 28 Fuxing Road, Wukesong, Haidian District, Beijing, 100853, China
| | - Bo Li
- Department of Cardiology, People's Liberation Army General Hospital, No. 28 Fuxing Road, Wukesong, Haidian District, Beijing, 100853, China
| | - Hao Zhou
- Department of Cardiology, People's Liberation Army General Hospital, No. 28 Fuxing Road, Wukesong, Haidian District, Beijing, 100853, China
| | - Ying Zhang
- Department of Cardiology, People's Liberation Army General Hospital, No. 28 Fuxing Road, Wukesong, Haidian District, Beijing, 100853, China
| | - Tian-Wen Han
- Department of Cardiology, People's Liberation Army General Hospital, No. 28 Fuxing Road, Wukesong, Haidian District, Beijing, 100853, China
| |
Collapse
|
34
|
Lei Y, Yang G, Hu L, Piao L, Inoue A, Jiang H, Sasaki T, Zhao G, Yisireyili M, Yu C, Xu W, Takeshita K, Okumura K, Kuzuya M, Cheng XW. Increased dipeptidyl peptidase-4 accelerates diet-related vascular aging and atherosclerosis in ApoE-deficient mice under chronic stress. Int J Cardiol 2017; 243:413-420. [PMID: 28549747 DOI: 10.1016/j.ijcard.2017.05.062] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 04/21/2017] [Accepted: 05/15/2017] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Exposure to psychosocial stress is a risk factor for cardiovascular disease. Given that dipeptidyl peptidase-4 (DPP4) regulates several intracellular signaling pathways associated with glucagon-like peptide-1 (GLP-1) metabolism, we investigated the role of DPP4 in stress-related vascular senescence and atherosclerosis in apolipoprotein E-deficient (ApoE-/-) mice. METHODS AND RESULTS ApoE-/- mice fed a high-fat (HF) diet were randomly assigned to one of non-stress and immobilized stress groups for 12weeks. Chronic stress accelerated vascular senescence and atherosclerotic plaque growth at the aortic roots. Stressed mice had increased levels of plasma DPP4 and decreased levels of plasma GLP-1 and adiponectin (APN) and adipose APN expression. Stress increased plaque macrophage infiltration, neovessel density, and elastin fragmentation, lessened the plaque collagen content, and increased the levels of toll-like receptor-2 (TLR2), TLR4, C-X-C chemokine receptor-4, cathepsins S and K, osteopontin, peroxisome proliferator-activated receptor-α, p16INK4A, p21, and gp91phox mRNAs and/or proteins. Stressed aortas had also increased matrix metalloproteinase-2 (MMP-2) and MMP-9 activities. DPP4 inhibition with anagliptin reversed stress-related atherosclerotic lesion formation, and this benefit was abrogated by APN blocking. In vitro, the GLP-1 receptor agonist exenatide stimulated APN expression in 3T3-L1 cells. CONCLUSIONS These results indicate that the DPP4 inhibition-mediated benefits are likely attributable, at least in part, to attenuation of plaque inflammation, oxidative stress and proteolysis associated with GLP-1-mediated APN production in ApoE-/- mice under stress. Thus, DPP4 will be a novel therapeutic target for the treatment of stress-related cardiovascular disease.
Collapse
Affiliation(s)
- Yanna Lei
- Department of ICU and Cardiology, Yanbian University Hospital, Yanji 133000, Jilin, PR China
| | - Guang Yang
- Department of ICU and Cardiology, Yanbian University Hospital, Yanji 133000, Jilin, PR China; Department of Community Health & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Lina Hu
- Department of Public Health, Guilin Medical College, Guilin 541004, Guangxi, PR China
| | - Limei Piao
- Department of ICU and Cardiology, Yanbian University Hospital, Yanji 133000, Jilin, PR China; Department of Community Health & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Aiko Inoue
- Department of ICU and Cardiology, Yanbian University Hospital, Yanji 133000, Jilin, PR China
| | - Haiying Jiang
- Department of Physiology and Pathophysiology, Yanbian University College of Medicine, Yanji 133000, Jilin, PR China
| | - Takeshi Sasaki
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine, Hamamatsu 4313192, Shizuokaken, Japan
| | - Guangxian Zhao
- Department of ICU and Cardiology, Yanbian University Hospital, Yanji 133000, Jilin, PR China
| | - Maimaiti Yisireyili
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Aichiken, Japan; Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul 02447, South Korea
| | - Chenglin Yu
- Department of ICU and Cardiology, Yanbian University Hospital, Yanji 133000, Jilin, PR China
| | - Wenhu Xu
- Department of ICU and Cardiology, Yanbian University Hospital, Yanji 133000, Jilin, PR China
| | - Kyosuke Takeshita
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Aichiken, Japan; Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul 02447, South Korea
| | - Kenji Okumura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Aichiken, Japan; Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul 02447, South Korea
| | - Masafumi Kuzuya
- Department of Community Health & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Aichiken, Japan
| | - Xian Wu Cheng
- Department of ICU and Cardiology, Yanbian University Hospital, Yanji 133000, Jilin, PR China; Department of Community Health & Geriatrics, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Division of Cardiology, Department of Internal Medicine, Kyung Hee University, Seoul 02447, South Korea; Institute of Innovation for Future Society, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Aichiken, Japan.
| | | |
Collapse
|