1
|
Agrawal V, Amasa S, Karabacak M, Margetis K. Perioperative Glucagon-Like Peptide-1 Agonist Use and Rates of Pseudarthrosis After Single-Level Lumbar Fusion: A Large Retrospective Cohort Study. Neurosurgery 2024:00006123-990000000-01452. [PMID: 39589142 DOI: 10.1227/neu.0000000000003291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/11/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND AND OBJECTIVE Pseudarthrosis is a common surgical complication after arthrodesis and is associated with poor clinical outcomes. The association between glucagon-like peptide-1 (GLP-1) agonist use and pseudarthrosis is yet to be explored. This study aims to examine the association of GLP-1 agonists with rates of pseudarthrosis in patients undergoing single-level lumbar fusion. METHODS This national multicenter cohort study used data spanning from June 19, 2010, to June 19, 2024, from the global health network TriNetX. One-to-one propensity score matching for age, sex, race, comorbidities, body mass index, and A1c was conducted to balance cohorts. The rates of pseudarthrosis were then assessed within the 6-month, 1-year, and 2-year postsurgical follow-up periods. RESULTS A total of 37 147 patients who underwent single-level lumbar fusion (mean [SD] age, 59.3 [13.5] years; 47.7% men and 52.3% women) were enrolled in the study. Among these, 712 individuals (1.9%) were identified as GLP-1 agonist users. After propensity score matching, there were 709 patients in each cohort. Patients who took a GLP-1 agonist had lower odds of developing pseudarthrosis 6 months [odds ratio (OR): 0.70, 95% CI: 0.51-0.96], 1 year [OR: 0.68, 95% CI: 0.50-0.91], and 2 years (OR: 0.68, 95% CI: 0.50-0.91) after a posterior lumbar interbody fusion/transforaminal lumbar interbody fusion procedure. CONCLUSION In this cohort study, patients who were prescribed GLP-1 agonists in the perioperative period had reduced rates of pseudarthrosis compared with patients without GLP-1 agonist prescriptions. These findings suggest a potential therapeutic benefit of GLP-1 agonists in enhancing spinal fusion outcomes and warrant further prospective studies to confirm these results and explore the underlying mechanisms.
Collapse
Affiliation(s)
- Vedant Agrawal
- University of Texas Medical Branch John Sealy School of Medicine, Galveston, Texas, USA
| | - Saketh Amasa
- University of Texas Medical Branch John Sealy School of Medicine, Galveston, Texas, USA
| | - Mert Karabacak
- Department of Neurosurgery, Mount Sinai Health System, New York, New York, USA
| | | |
Collapse
|
2
|
Le R, Nguyen MT, Allahwala MA, Psaltis JP, Marathe CS, Marathe JA, Psaltis PJ. Cardiovascular Protective Properties of GLP-1 Receptor Agonists: More than Just Diabetic and Weight Loss Drugs. J Clin Med 2024; 13:4674. [PMID: 39200816 PMCID: PMC11355214 DOI: 10.3390/jcm13164674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
Owing to their potent glucose-lowering efficacy and substantial weight loss effects, glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are now considered part of the frontline therapeutic options to treat both type 2 diabetes mellitus and nondiabetic overweight/obesity. Stemming from successful demonstration of their cardiometabolic modulation and reduction of major adverse cardiovascular events in clinical outcome trials, GLP-1 RAs have since been validated as agents with compelling cardiovascular protective properties. Studies spanning from the bench to preclinical and large-scale randomised controlled trials have consistently corroborated the cardiovascular benefits of this pharmacological class. Most notably, there is converging evidence that they exert favourable effects on atherosclerotic ischaemic endpoints, with preclinical data indicating that they may do so by directly modifying the burden and composition of atherosclerotic plaques. This narrative review examines the underlying pharmacology and clinical evidence behind the cardiovascular benefits of GLP-1 RAs, with particular focus on atherosclerotic cardiovascular disease. It also delves into the mechanisms that underpin their putative plaque-modifying actions, addresses existing knowledge gaps and therapeutic challenges and looks to future developments in the field, including the use of combination incretin agents for diabetes and weight loss management.
Collapse
Affiliation(s)
- Richard Le
- College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia;
- Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (M.T.N.); (M.A.A.); (J.A.M.)
| | - Mau T. Nguyen
- Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (M.T.N.); (M.A.A.); (J.A.M.)
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (J.P.P.); (C.S.M.)
| | - Momina A. Allahwala
- Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (M.T.N.); (M.A.A.); (J.A.M.)
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (J.P.P.); (C.S.M.)
| | - James P. Psaltis
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (J.P.P.); (C.S.M.)
| | - Chinmay S. Marathe
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (J.P.P.); (C.S.M.)
- Department of Endocrinology, Central Adelaide Local Health Network, Adelaide 5000, Australia
| | - Jessica A. Marathe
- Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (M.T.N.); (M.A.A.); (J.A.M.)
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (J.P.P.); (C.S.M.)
| | - Peter J. Psaltis
- Heart and Vascular Program, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide 5000, Australia; (M.T.N.); (M.A.A.); (J.A.M.)
- Department of Cardiology, Central Adelaide Local Health Network, Adelaide 5000, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (J.P.P.); (C.S.M.)
| |
Collapse
|
3
|
Castiglione R, Vivacqua A, Santoro M, De Rose D, Peluso G, Panza S, Aquila S, D'Agata R. Glucagon-like Peptide-1 Acts as Signaling Mediator to Modulate Human Sperm Performance via Targeting Akt, JNK and IRS-1 Cell Signaling Cascades: Novel Insights into Sperm Physiopathology. J Clin Med 2023; 12:jcm12113844. [PMID: 37298039 DOI: 10.3390/jcm12113844] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/19/2023] [Accepted: 06/01/2023] [Indexed: 06/12/2023] Open
Abstract
Recent evidence suggests that the male gonad is a potential target of glucagon-like peptide-1 (GLP-1). We investigated the effects of glucagon-like peptide-1 (GLP-1) on sperm function and the molecular mechanisms through which it may act. Semen samples of healthy men were incubated in the presence or absence of a GLP-1 mimetic analog, exendin-4 (Exe). In a different analysis, sperm were exposed to tumor necrosis factor (TNF-α) alone and, in some tubes, TNF-α was added after previous exposure to exendin-4 (Exe). Sperm parameters and protein-kinase B (p-Akt), insulin receptor substrate-1 (p-IRS-1 Ser312), and c Jun N-terminal protein kinase (p-JNK Thr183/Tyr185) were considered and evaluated. Sperm parameters, when incubated for 4 h in a simple defined balanced salt solution lacking protein, declined progressively with incubation time. The maximum decline was associated with a significant decrease in phosphorylated protein kinase B (p-Akt), concomitantly to an increase in insulin receptor substrate-1 (p-IRS-1 Ser312) and c Jun N-terminal protein kinase (p-JNK Thr183/Tyr185). Preincubation with exendin-4 (Exe) prevented this decline and maintained sperm motility (progressive-PM and total-TM). TNF-α exposure resulted in decreased sperm motility (PM and TM) and viability (V) in a concentration-dependent manner. Exe addition attenuated this TNF-α negative effect on sperm parameters. Glucagon-like peptide-1 (GLP-1) also acts by reducing levels of the "negative" kinases p-IRS-1Ser312 and p-JNK. An imbalance involving these three kinases in sperm, as it occurs in somatic cells, is a novel scenario that may participate in sperm physiopathology.
Collapse
Affiliation(s)
- Roberto Castiglione
- Department of Experimental and Clinical Medicine, University of Catania, 95123 Catania, Italy
| | - Adele Vivacqua
- Department of Pharmacy and Science of Health and Nutrition, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Marta Santoro
- Department of Pharmacy and Science of Health and Nutrition, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Daniela De Rose
- Department of Pharmacy and Science of Health and Nutrition, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Giuseppina Peluso
- Unit of Physiophatology of Reproduction, Annunziata Hospital, 87100 Cosenza, Italy
| | - Salvatore Panza
- Department of Pharmacy and Science of Health and Nutrition, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Saveria Aquila
- Department of Pharmacy and Science of Health and Nutrition, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
- Centro Sanitario, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Rosario D'Agata
- Department of Experimental and Clinical Medicine, University of Catania, 95123 Catania, Italy
| |
Collapse
|
4
|
Khalifa O, H. Mroue K, Mall R, Ullah E, S. Al-Akl N, Arredouani A. Investigation of the Effect of Exendin-4 on Oleic Acid-Induced Steatosis in HepG2 Cells Using Fourier Transform Infrared Spectroscopy. Biomedicines 2022; 10:biomedicines10102652. [PMID: 36289914 PMCID: PMC9599706 DOI: 10.3390/biomedicines10102652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/10/2022] [Accepted: 07/13/2022] [Indexed: 12/04/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common liver lesion that is untreatable with medications. Glucagon-like peptide-1 receptor (GLP-1R) agonists have recently emerged as a potential NAFLD pharmacotherapy. However, the molecular mechanisms underlying these drugs’ beneficial effects are not fully understood. Using Fourier transform infrared (FTIR) spectroscopy, we sought to investigate the biochemical changes in a steatosis cell model treated or not with the GLP-1R agonist Exendin-4 (Ex-4). HepG2 cells were made steatotic with 400 µM of oleic acid and then treated with 200 nM Ex-4 in order to reduce lipid accumulation. We quantified steatosis using the Oil Red O staining method. We investigated the biochemical alterations induced by steatosis and Ex-4 treatment using Fourier transform infrared (FTIR) spectroscopy and chemometric analyses. Analysis of the Oil Red O staining showed that Ex-4 significantly reduces steatosis. This reduction was confirmed by FTIR analysis, as the phospholipid band (C=O) at 1740 cm−1 in Ex-4 treated cells is significantly decreased compared to steatotic cells. The principal component analysis score plots for both the lipid and protein regions showed that the untreated and Ex-4-treated samples, while still separated, are clustered close to each other, far from the steatotic cells. The biochemical and structural changes induced by OA-induced lipotoxicity are at least partially reversed upon Ex-4 treatment. FTIR and chemometric analyses revealed that Ex-4 significantly reduces OA-induced lipid accumulation, and Ex-4 also restored the lipid and protein biochemical alterations caused by lipotoxicity-induced oxidative stress. In combination with chemometric analyses, FTIR spectroscopy may offer new approaches for investigating the mechanisms underpinning NAFLD.
Collapse
Affiliation(s)
- Olfa Khalifa
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha 34110, Qatar
| | - Kamal H. Mroue
- Qatar Environment and Energy Research Institute, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha 34110, Qatar
| | - Raghvendra Mall
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha 34110, Qatar
- Department of Immunology, St. Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105-3678, USA
| | - Ehsan Ullah
- Qatar Computing Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha 34110, Qatar
| | - Nayla S. Al-Akl
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha 34110, Qatar
| | - Abdelilah Arredouani
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha 34110, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha 34110, Qatar
- Correspondence:
| |
Collapse
|
5
|
Pahud de Mortanges A, Sinaci E, Salvador D, Bally L, Muka T, Wilhelm M, Bano A. GLP-1 Receptor Agonists and Coronary Arteries: From Mechanisms to Events. Front Pharmacol 2022; 13:856111. [PMID: 35370744 PMCID: PMC8964343 DOI: 10.3389/fphar.2022.856111] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 02/15/2022] [Indexed: 12/13/2022] Open
Abstract
Objective: Glucagon-like peptide 1 receptor agonists (GLP-1 RAs) lower plasma glucose through effects on insulin and glucagon secretion and by decelerating gastric emptying. GLP-1 RAs have many beneficial effects beyond glycemic control, including a protective role on the cardiovascular system. However, underlying mechanisms linking GLP-1 RAs with coronary artery disease are complex and not fully elucidated. In this mini-review, we discuss these mechanisms and subsequent clinical events. Data Sources: We searched PubMed and Google Scholar for evidence on GLP-1 RAs and coronary events. We did not apply restrictions on article type. We reviewed publications for clinical relevance. Synopsis of Content: In the first part, we review the current evidence concerning the role of GLP-1 RAs on potential mechanisms underlying the development of coronary events. Specifically, we discuss the role of GLP-1 RAs on atherosclerosis and vasospasms of epicardial coronary arteries, as well as structural/functional changes of coronary microvasculature. In the second part, we summarize the clinical evidence on the impact of GLP-1 RAs in the prevention of acute and chronic coronary syndromes and coronary revascularization. We conclude by discussing existing gaps in the literature and proposing directions for future research.
Collapse
Affiliation(s)
| | - Eldem Sinaci
- Faculty of Medicine, University of Bern, Bern, Switzerland
| | - Dante Salvador
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.,Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Lia Bally
- Department of Diabetes, Endocrinology, Nutritional Medicine, and Metabolism, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Taulant Muka
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Matthias Wilhelm
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Arjola Bano
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland.,Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
6
|
Shou X, Wang Y, Duan C, Yuan G, Wei N, Yang Y, Hu Y. Knowledge Domain and Emerging Trends of Glucagon-Like Peptide 1 Receptor Agonists in Cardiovascular Research: A Bibliometric Analysis. Curr Probl Cardiol 2022:101194. [PMID: 35395332 DOI: 10.1016/j.cpcardiol.2022.101194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/02/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND Patients with type 2 diabetes (T2DM) are more likely to have cardiovascular disease (CVD). Glucose-lowering drugs with cardiovascular benefits represented by Glucagon-like peptide 1 receptor agonists (GLP1RAs) were discovered and gained more and more attention. METHODS Data from 1985 to the 2021 were downloaded in the Web of Science Core Collection (WoSCC) database. CiteSpaceV was used for bibliometric analysis to find research hotspots and frontiers. RESULTS The 2088 papers were published by 74 countries (regions), 876 institutions, and 2203 authors. The annual publications increased over time from 2005 to 2020. DIABETES OBESITY METABOLISM published the most papers. The USA and China were the top 2 productive nations. The leading institution was the University of Copenhagen, and the most productive researcher was John B Buse. The most cited paper is "Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes" (by Marso SP, 2016). The research hotspots include the effects of GLP1RA on cardiovascular outcomes, efficacy, complicated metabolic abnormalities, protective mechanisms, and other novel anti-diabetic drugs for cardiovascular protection. Research frontiers include cardiovascular studies on semaglutide, as well as the most prominent research approach in the field-placebo-controlled trial. CONCLUSION Numerous countries, institutions, and authors have focused on GLP1RA in cardiovascular research and a great deal of literature has been published. Five research hotspots and two frontiers illustrate the current status and emerging trends of GLP1RA in cardiovascular research. The cardiovascular effects and clinical efficacy of GLP1RA are a current hot topic that is rapidly evolving and of high research value.
Collapse
Affiliation(s)
- Xintian Shou
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yumeng Wang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Chenglin Duan
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Guozhen Yuan
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Namin Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yihan Yang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanhui Hu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
7
|
Peng Y, Lin H, Tian S, Liu S, Li J, Lv X, Chen S, Zhao L, Pu F, Chen X, Shu H, Qing X, Shao Z. Glucagon-like peptide-1 receptor activation maintains extracellular matrix integrity by inhibiting the activity of mitogen-activated protein kinases and activator protein-1. Free Radic Biol Med 2021; 177:247-259. [PMID: 34737144 DOI: 10.1016/j.freeradbiomed.2021.10.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/04/2021] [Accepted: 10/27/2021] [Indexed: 12/25/2022]
Abstract
Disruption of the intervertebral disc extracellular matrix (ECM) is a hallmark of intervertebral disc degeneration (IDD), which is largely attributed to excessive oxidative stress. However, there is a lack of clinically feasible approaches to promote the reconstruction of the disc ECM. Glucagon-like peptide-1 (GLP-1), a safe polypeptide hormone adopted to treat type 2 diabetes mellitus, has shown great potential for relieving oxidative stress-related damage. To our knowledge, this is the first study to reveal that exenatide, a GLP-1 receptor (GLP-1R) agonist, can upregulate disc ECM synthesis and attenuate oxidative stress-induced ECM degradation and IDD. Mechanistically, we found that exenatide inhibited the activation of mitogen-activated protein kinases (MAPK) signaling pathway and the formation of BATF/JUNs heterodimers (an index of activator protein-1 (AP-1) activity). The restoration of MAPK signaling activation reversed the protective effects of exenatide and enhanced downstream BATF/JUNs binding. BATF overexpression was also found to aggravate disc ECM damage, even in the presence of exenatide. In summary, exenatide is an effective agent that regulates ECM anabolic balance and restores disc degeneration by inhibiting MAPK activation and its downstream AP-1 activity. The present study provides a therapeutic rationale for activating the GLP-1 receptor against IDD and establishes the important role of AP-1 activity in the pathogenesis of IDD.
Collapse
Affiliation(s)
- Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shuo Tian
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sheng Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinye Li
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiao Lv
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Songfeng Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450052, China
| | - Lei Zhao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Feifei Pu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xi Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hongyang Shu
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, China; Hubei Key Laboratory of Genetics and Molecular Mechanism of Cardiologic Disorders, Huazhong University of Science and Technology, Wuhan, 430000, China
| | - Xiangcheng Qing
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
8
|
Natarajan SK, Bruett T, Muthuraj PG, Sahoo PK, Power J, Mott JL, Hanson C, Anderson-Berry A. Saturated free fatty acids induce placental trophoblast lipoapoptosis. PLoS One 2021; 16:e0249907. [PMID: 33886600 PMCID: PMC8062006 DOI: 10.1371/journal.pone.0249907] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 03/26/2021] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Obesity during pregnancy increases the risk for maternal complications like gestational diabetes, preeclampsia, and maternal inflammation. Maternal obesity also increases the risk of childhood obesity, intrauterine growth restriction (IUGR) and diabetes to the offspring. Increased circulating free fatty acids (FFAs) in obesity due to adipose tissue lipolysis induces lipoapoptosis to hepatocytes, cholangiocytes, and pancreatic-β-cells. During the third trimester of human pregnancy, there is an increase in maternal lipolysis and release of FFAs into the circulation. It is currently unknown if increased FFAs during gestation as a result of maternal obesity cause placental cell lipoapoptosis. Increased exposure of FFAs during maternal obesity has been shown to result in placental lipotoxicity. The objective of the present study is to determine saturated FFA-induced trophoblast lipoapoptosis and also to test the protective role of monounsaturated fatty acids against FFA-induced trophoblast lipoapoptosis using in vitro cell culture model. Here, we hypothesize that saturated FFAs induce placental trophoblast lipoapoptosis, which was prevented by monounsaturated fatty acids. METHODS Biochemical and structural markers of apoptosis by characteristic nuclear morphological changes with DAPI staining, and caspase 3/7 activity was assessed. Cleaved PARP and cleaved caspase 3 were examined by western blot analysis. RESULTS Treatment of trophoblast cell lines, JEG-3 and JAR cells with palmitate (PA) or stearate (SA) induces trophoblast lipoapoptosis as evidenced by a significant increase in apoptotic nuclear morphological changes and caspase 3/7 activity. We observed that saturated FFAs caused a concentration-dependent increase in placental trophoblast lipoapoptosis. We also observed that monounsaturated fatty acids like palmitoleate and oleate mitigates placental trophoblast lipoapoptosis caused due to PA exposure. CONCLUSION We show that saturated FFAs induce trophoblast lipoapoptosis. Co-treatment of monounsaturated fatty acids like palmitoleate and oleate protects against FFA-induced trophoblast lipoapoptosis.
Collapse
Affiliation(s)
- Sathish Kumar Natarajan
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
- * E-mail:
| | - Taylor Bruett
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Philma Glora Muthuraj
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Prakash K. Sahoo
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Jillian Power
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE, United States of America
| | - Justin L. Mott
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Corrine Hanson
- College of Allied Health Professions Medical Nutrition Education, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Ann Anderson-Berry
- Department of Pediatrics, University of Nebraska Medical Center, Omaha, NE, United States of America
| |
Collapse
|
9
|
Role of calcineurin biosignaling in cell secretion and the possible regulatory mechanisms. Saudi J Biol Sci 2021; 28:116-124. [PMID: 33424288 PMCID: PMC7783665 DOI: 10.1016/j.sjbs.2020.08.042] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/02/2020] [Accepted: 08/30/2020] [Indexed: 11/22/2022] Open
Abstract
Cyclic adenosine monophosphate (cAMP) and calcium ions (Ca2+) are two chemical molecules that play a central role in the stimulus-dependent secretion processes within cells. Ca2+ acts as the basal signaling molecule responsible to initiate cell secretion. cAMP primarily acts as an intracellular second messenger in a myriad of cellular processes by activating cAMP-dependent protein kinases through association with such kinases in order to mediate post-translational phosphorylation of those protein targets. Put succinctly, both Ca2+ and cAMP act by associating or activating other proteins to ensure successful secretion. Calcineurin is one such protein regulated by Ca2+; its action depends on the intracellular levels of Ca2+. Being a phosphatase, calcineurin dephosphorylate and other proteins, as is the case with most other phosphatases, such as protein phosphatase 2A (PP2A), PP2C, and protein phosphatase-1 (PP1), will likely be activated by phosphorylation. Via this process, calcineurin is able to affect different intracellular signaling with clinical importance, some of which has been the basis for development of different calcineurin inhibitors. In this review, the cAMP-dependent calcineurin bio-signaling, protein-protein interactions and their physiological implications as well as regulatory signaling within the context of cellular secretion are explored.
Collapse
|
10
|
Beneficial Effects of Glucagon-Like Peptide-1 (GLP-1) in Diabetes-Induced Retinal Abnormalities: Involvement of Oxidative Stress. Antioxidants (Basel) 2020; 9:antiox9090846. [PMID: 32927585 PMCID: PMC7554849 DOI: 10.3390/antiox9090846] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/01/2020] [Accepted: 09/07/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Hyperglycemia-induced oxidative stress plays a key role in diabetic complications, including diabetic retinopathy. The main goal of this study was to assess whether the topical administration (eye drops) of glucagon-like peptide-1 (GLP-1) has any effect on oxidative stress in the retina. Methods: db/db mice were treated with eye drops of GLP-1 or vehicle for three weeks, with db/+ mice being used as control. Studies included the assessment by western blot of the antioxidant defense markers CuZnSOD, MnSOD, glutathione peroxidase and reductase; immunofluorescence measurements of DNA/RNA damage, nitro tyrosine and Ki67 and Babam2 proteins. Results: GLP-1 eye drops protected from oxidative stress by increasing the protein levels of glutathione reductase, glutathione peroxidase and CuZnSOD and MnSOD in diabetic retinas. This was associated with a significant reduction of DNA/RNA damage and the activation of proteins involved in DNA repair in the retina (Babam2) and Ki67 (a biomarker of cell proliferation). Conclusions: GLP-1 modulates the antioxidant defense system in the diabetic retina and has a neuroprotective action favoring DNA repair and neuron cells proliferation.
Collapse
|
11
|
Neuroprotection of GLP-1/GIP receptor agonist via inhibition of mitochondrial stress by AKT/JNK pathway in a Parkinson's disease model. Life Sci 2020; 256:117824. [PMID: 32445758 DOI: 10.1016/j.lfs.2020.117824] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 05/10/2020] [Accepted: 05/18/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To investigate the effect of glucagon-like peptide-1 (GLP-1) receptor and glucose dependent insulinotrophic polypeptide (GIP) receptor dual agonist DA-JC4 on alleviating Parkinson's disease (PD) and unveil related cellular mechanisms. METHODS Rotenone was injected to generate a rat PD model, on which the effect of DA-JC4 on motor functions was evaluated by rotational behavioral assay and open field test. The survival of dopaminergic neurons was analyzed, in addition to assays for mitochondrial stress and quantification of neurotransmitter levels using high performance liquid chromatography (HPLC). In cultured hippocampal neurons, the effect of DA-JC4 on mitochondrial stress and related cellular mechanism was analyzed by Flow cytometry, western blotting and reactive oxygen species (ROS). RESULTS DA-JC4 significantly improved motor functions in PD rats, and elevated levels of major neurotransmitters. By histological analysis, DA-JC4 protected dopaminergic neurons from rotenone-induced cell death, which was associated with reduced mitochondrial stress. Experiments in cultured rat hippocampal neurons validated the neuroprotective role of DA-JC4 against cell apoptosis and mitochondrial stress induced by rotenone. The protective effect of DA-JC4 was later found to be dependent on AKT/JNK signal pathway, as treatment using AKT inhibitor or JNK activator abolished such effects. CONCLUSION Our results showed that the dual agonist of GLP-1/GIP receptor could ameliorate motor dysfunctions of PD by protecting dopaminergic neurons which was mediated by relieved mitochondrial stress and apoptosis via AKT/JNK signal pathway.
Collapse
|
12
|
Aini K, Fukuda D, Tanaka K, Higashikuni Y, Hirata Y, Yagi S, Kusunose K, Yamada H, Soeki T, Sata M. Vildagliptin, a DPP-4 Inhibitor, Attenuates Endothelial Dysfunction and Atherogenesis in Nondiabetic Apolipoprotein E-Deficient Mice. Int Heart J 2019; 60:1421-1429. [PMID: 31735774 DOI: 10.1536/ihj.19-117] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors are novel antidiabetic agents with possible vascular protection effects. Endothelial dysfunction is an initiation step in atherogenesis. The purpose of this study was to investigate whether vildagliptin (Vilda) attenuates the development of endothelial dysfunction and atherosclerotic lesions in nondiabetic apolipoprotein E-deficient (ApoE-/-) mice. Eight-week-old nondiabetic ApoE-/- mice fed a Western-type diet received Vilda (50 mg/kg/day) for 20 weeks or 8 weeks. After 20 weeks of treatment, Vilda administration reduced atherogenesis in the aortic arch as determined by en face Sudan IV staining compared with the vehicle group (P < 0.05). Vilda also reduced lipid accumulation (P < 0.05) and vascular cell adhesion molecule-1 (VCAM-1) expression (P < 0.05) and tended to decrease macrophage infiltration (P = 0.05) into atherosclerotic plaques compared with vehicle. After 8 weeks of treatment, endothelium-dependent vascular reactivity was examined. Vilda administration significantly attenuated the impairment of endothelial function in nondiabetic ApoE-/- mice compared with the vehicle group (P < 0.05). Vilda treatment did not alter metabolic parameters, including blood glucose level, in both study protocols. To investigate the mechanism, aortic segments obtained from wild-type mice were incubated with exendin-4 (Ex-4), a glucagon-like peptide-1 (GLP-1) analog, in the presence or absence of lipopolysaccharide (LPS). Ex-4 attenuated the impairment of endothelium-dependent vasodilation induced by LPS (P < 0.01). Furthermore, Ex-4 promoted phosphorylation of eNOS at Ser1177 which was decreased by LPS in human umbilical endothelial cells (P < 0.05). Vilda inhibited the development of endothelial dysfunction and prevented atherogenesis in nondiabetic ApoE-/- mice. Our results suggested that GLP-1-dependent amelioration of endothelial dysfunction is associated with the atheroprotective effects of Vilda.
Collapse
Affiliation(s)
- Kunduziayi Aini
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Daiju Fukuda
- Department of Cardio-Diabetes Medicine, Tokushima University Graduate School of Biomedical Science
| | - Kimie Tanaka
- Division for Health Service Promotion, The University of Tokyo
| | | | | | - Shusuke Yagi
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Kenya Kusunose
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Hirotsugu Yamada
- Department of Community Medicine for Cardiology, Tokushima University Graduate School of Biomedical Sciences
| | - Takeshi Soeki
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| | - Masataka Sata
- Department of Cardiovascular Medicine, Tokushima University Graduate School of Biomedical Sciences
| |
Collapse
|
13
|
Luo X, Hu Y, He S, Ye Q, Lv Z, Liu J, Chen X. Dulaglutide inhibits high glucose- induced endothelial dysfunction and NLRP3 inflammasome activation. Arch Biochem Biophys 2019; 671:203-209. [DOI: 10.1016/j.abb.2019.07.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/08/2019] [Accepted: 07/10/2019] [Indexed: 02/07/2023]
|
14
|
Koshibu M, Mori Y, Saito T, Kushima H, Hiromura M, Terasaki M, Takada M, Fukui T, Hirano T. Antiatherogenic effects of liraglutide in hyperglycemic apolipoprotein E-null mice via AMP-activated protein kinase-independent mechanisms. Am J Physiol Endocrinol Metab 2019; 316:E895-E907. [PMID: 30860874 DOI: 10.1152/ajpendo.00511.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1RAs) exert potent glucose-lowering effects without increasing risks for hypoglycemia and weight gain. Preclinical studies have demonstrated direct antiatherogenic effects of GLP-1RAs in normoglycemic animal models; however, the underlying mechanisms in hyperglycemic conditions have not been fully clarified. Here we aimed to elucidate the role of AMP-activated protein kinase (AMPK) in antiatherogenic effects of GLP-1RAs in hyperglycemic mice. Streptozotocin-induced hyperglycemic apolipoprotein E-null mice were treated with vehicle, low-dose liraglutide (17 nmol·kg-1·day-1), or high-dose liraglutide (107 nmol·kg-1·day-1) in experiment 1 and the AMPK inhibitor dorsomorphin, dorsomorphin + low-dose liraglutide, or dorsomorphin + high-dose liraglutide in experiment 2. Four weeks after treatment, aortas were collected to assess atherosclerosis. In experiment 1, metabolic parameters were similar among the groups. Assessment of atherosclerosis revealed that high-dose liraglutide treatments reduced lipid deposition on the aortic surface and plaque volume and intraplaque macrophage accumulation at the aortic sinus. In experiment 2, liraglutide-induced AMPK phosphorylation in the aorta was abolished by dorsomorphin; however, the antiatherogenic effects of high-dose liraglutide were preserved. In cultured human umbilical vein endothelial cells, liraglutide suppressed tumor necrosis factor-induced expression of proatherogenic molecules; these effects were maintained under small interfering RNA-mediated knockdown of AMPKα1 and in the presence of dorsomorphin. Conversely, in human monocytic U937 cells, the anti-inflammatory effects of liraglutide were abolished by dorsomorphin. In conclusion, liraglutide exerted AMPK-independent antiatherogenic effects in hyperlipidemic mice with streptozotocin-induced hyperglycemia, with the possible involvement of AMPK-independent suppression of proatherogenic molecules in vascular endothelial cells.
Collapse
Affiliation(s)
- Masakazu Koshibu
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Yusaku Mori
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Tomomi Saito
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Hideki Kushima
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Munenori Hiromura
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Michishige Terasaki
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Michiya Takada
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Tomoyasu Fukui
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| | - Tsutomu Hirano
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, Showa University School of Medicine, Shinagawa, Tokyo , Japan
| |
Collapse
|
15
|
Reactive oxygen species (ROS) in macrophage activation and function in diabetes. Immunobiology 2018; 224:242-253. [PMID: 30739804 DOI: 10.1016/j.imbio.2018.11.010] [Citation(s) in RCA: 333] [Impact Index Per Article: 47.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/27/2018] [Accepted: 11/27/2018] [Indexed: 12/14/2022]
Abstract
In a diabetic milieu high levels of reactive oxygen species (ROS) are induced. This contributes to the vascular complications of diabetes. Recent studies have shown that ROS formation is exacerbated in diabetic monocytes and macrophages due to a glycolytic metabolic shift. Macrophages are important players in the progression of diabetes and promote inflammation through the release of pro-inflammatory cytokines and proteases. Because ROS is an important mediator for the activation of pro-inflammatory signaling pathways, obesity and hyperglycemia-induced ROS production may favor induction of M1-like pro-inflammatory macrophages during diabetes onset and progression. ROS induces MAPK, STAT1, STAT6 and NFκB signaling, and interferes with macrophage differentiation via epigenetic (re)programming. Therefore, a comprehensive understanding of the impact of ROS on macrophage phenotype and function is needed in order to improve treatment of diabetes and its vascular complications. In the current comprehensive review, we dissect the role of ROS in macrophage polarization, and analyze how ROS production links metabolism and inflammation in diabetes and its complications. Finally, we discuss the contribution of ROS to the crosstalk between macrophages and endothelial cells in diabetic complications.
Collapse
|
16
|
Tang Z, Liu L, Guo Y, Deng G, Chen M, Wei J. Exendin‑4 reverses endothelial dysfunction in mice fed a high‑cholesterol diet by a GTP cyclohydrolase‑1/tetrahydrobiopterin pathway. Mol Med Rep 2018; 18:3350-3358. [PMID: 30085331 PMCID: PMC6102738 DOI: 10.3892/mmr.2018.9345] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 06/26/2018] [Indexed: 12/25/2022] Open
Abstract
The present study examined whether exendin‑4 (Ex4) can improve the endothelial dysfunction of apolipoprotein E knockout (APOE‑KO) mice fed a high‑cholesterol diet and the potential mechanism by which it acts. Genetically wild‑type (WT) C57BL/6 mice and APOE‑KO mice of C57BL/6 background, were each randomly assigned to receive either Ex4 treatment (Ex4‑treated, for 8 weeks) or not (control). The 4 groups were fed the same high‑cholesterol diet for 8 weeks. The following were measured at the end of the eighth week: Endothelium‑dependent vasodilation of the arteries; plasma nitric oxide (NO) and metabolic index; levels of endothelial NO synthase (eNOS); phosphorylated eNOS (p‑eNOS; Ser‑1,177); guanosine triphosphate cyclohydrolase‑1 (GCH1); and tetrahydrobiopterin (THB). Ex4 treatment was associated with higher p‑eNOS levels in the WT group and in the APOE‑KO group, and higher vascular expression of GCH1 and higher arterial THB content, compared with baseline values. The results of the present study suggested that Ex4 may exert cardioprotective effects by reversing high‑cholesterol diet‑induced endothelial dysfunction in APOE‑KO mice. The protective mechanism is probably associated with the promotion of the expression levels of GCH1 protein and THB that maintain the normal function of eNOS.
Collapse
Affiliation(s)
- Zhiqi Tang
- Department of Cardiology, The First People's Hospital of Nanning City, Nanning, Guangxi 530021, P.R. China
| | - Lijuan Liu
- School of Continuing Education, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yujie Guo
- Department of Cardiology, The People's Hospital of Liuzhou City, Liuzhou, Guangxi 545006, P.R. China
| | - Guoxiong Deng
- Department of Cardiology, The First People's Hospital of Nanning City, Nanning, Guangxi 530021, P.R. China
| | - Meixiang Chen
- Department of Cardiology, The First People's Hospital of Nanning City, Nanning, Guangxi 530021, P.R. China
| | - Jinru Wei
- Department of Cardiology, The First People's Hospital of Nanning City, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW In addition to their effects on glycemic control, two specific classes of relatively new anti-diabetic drugs, namely the sodium glucose co-transporter-2 inhibitors (SGLT2i) and glucagon-like peptide-1 receptor agonists (GLP-1RA) have demonstrated reduced rates of major adverse cardiovascular events (MACE) in subjects with type 2 diabetes (T2D) at high risk for cardiovascular disease (CVD). This review summarizes recent experimental results that inform putative molecular mechanisms underlying these benefits. RECENT FINDINGS SGLT2i and GLP-1RA exert cardiovascular effects by targeting in both common and distinctive ways (A) several mediators of macro- and microvascular pathophysiology: namely (A1) inflammation and atherogenesis, (A2) oxidative stress-induced endothelial dysfunction, (A3) vascular smooth muscle cell reactive oxygen species (ROS) production and proliferation, and (A4) thrombosis. These agents also exhibit (B) hemodynamic effects through modulation of (B1) natriuresis/diuresis and (B2) the renin-angiotensin-aldosterone system. This review highlights that while GLP-1RA exert direct effects on vascular (endothelial and smooth muscle) cells, the effects of SGLT2i appear to include the activation of signaling pathways that prevent adverse vascular remodeling. Both SGLT2i and GLP-1RA confer hemodynamic effects that counter adverse cardiac remodeling.
Collapse
Affiliation(s)
- Dorrin Zarrin Khat
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada
| | - Mansoor Husain
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
- Department of Medicine, University of Toronto, Toronto, Canada.
- Toronto General Hospital Research Institute, University Health Network, Toronto, Canada.
- Heart and Stroke Richard Lewar Centre of Excellence, University of Toronto, Toronto, Canada.
- Ted Rogers Centre for Heart Research, University Health Network, Toronto, Canada.
- Peter Munk Cardiac Centre, University Health Network, Toronto, Canada.
| |
Collapse
|
18
|
Lim S, Kim KM, Nauck MA. Glucagon-like Peptide-1 Receptor Agonists and Cardiovascular Events: Class Effects versus Individual Patterns. Trends Endocrinol Metab 2018; 29:238-248. [PMID: 29463450 DOI: 10.1016/j.tem.2018.01.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 01/28/2018] [Accepted: 01/29/2018] [Indexed: 02/06/2023]
Abstract
Several new glucose-lowering medications have been approved, such as dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 receptor agonists (GLP-1RAs), and sodium glucose cotransporter-2 inhibitors. Among GLP-1RAs, lixisenatide, a short-acting drug, did not show cardiovascular (CV) benefits in patients with Type 2 diabetes mellitus (T2D) and acute coronary syndrome. Extended-release exenatide was also not significantly better for CV outcomes. By contrast, once daily liraglutide and once weekly semaglutide, both long-acting GLP-1RAs, decreased the incidence of major adverse CV events and mortality. This Review attempts to explain favorable CV results with some, but not all, GLP-1RAs, to aid in their differential prescription with the aim of further reducing the adverse CV burden of T2D.
Collapse
Affiliation(s)
- Soo Lim
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, South Korea.
| | - Kyoung Min Kim
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Michael A Nauck
- Diabetes Center Bochum-Hattingen, Department of Medicine I, St Josef-Hospital (Ruhr-Universität Bochum), Bochum, Germany.
| |
Collapse
|
19
|
Sun ZL, Jiang XF, Cheng YC, Liu YF, Yang K, Zhu SL, Kong XB, Tu Y, Bian KF, Liu ZL, Chen XY. Exendin-4 inhibits high-altitude cerebral edema by protecting against neurobiological dysfunction. Neural Regen Res 2018; 13:653-663. [PMID: 29722317 PMCID: PMC5950675 DOI: 10.4103/1673-5374.230291] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The anti-inflammatory and antioxidant effects of exendin-4 (Ex-4) have been reported previously. However, whether (Ex-4) has anti-inflammatory and antioxidant effects on high-altitude cerebral edema (HACE) remains poorly understood. In this study, two rat models of HACE were established by placing rats in a hypoxic environment with a simulated altitude of either 6000- or 7000-m above sea level (MASL) for 72 hours. An altitude of 7000 MASL with 72-hours of hypoxia was found to be the optimized experimental paradigm for establishing HACE models. Then, in rats where a model of HACE was established by introducing them to a 7000 MASL environment with 72-hours of hypoxia treatment, 2, 10 and, 100 μg of Ex-4 was intraperitoneally administrated. The open field test and tail suspension test were used to test animal behavior. Routine methods were used to detect change in inflammatory cells. Hematoxylin-eosin staining was performed to determine pathological changes to brain tissue. Wet/dry weight ratios were used to measure brain water content. Evans blue leakage was used to determine blood-brain barrier integrity. Enzyme-linked immunosorbent assay (ELISA) was performed to measure markers of inflammation and oxidative stress including superoxide dismutase, glutathione, and malonaldehyde values, as well as interleukin-6, tumor necrosis factor-alpha, cyclic adenosine monophosphate levels in the brain tissue. Western blot analysis was performed to determine the levels of occludin, ZO-1, SOCS-3, vascular endothelial growth factor, EPAC1, nuclear factor-kappa B, and aquaporin-4. Our results demonstrate that Ex-4 preconditioning decreased brain water content, inhibited inflammation and oxidative stress, alleviated brain tissue injury, maintain blood-brain barrier integrity, and effectively improved motor function in rat models of HACE. These findings suggest that Ex-4 exhibits therapeutic potential in the treatment of HACE.
Collapse
Affiliation(s)
- Zhong-Lei Sun
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin, Tianjin; Jinzhou Medical University, Jinzhou, Liaoning Province, China
| | - Xian-Feng Jiang
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin; Tianjin Medical University, Tianjin, China
| | - Yuan-Chi Cheng
- Central Hospital of Fengxian District of Shanghai, Shanghai, China
| | - Ying-Fu Liu
- Logistics University of People's Armed Police Force, Tianjin, China
| | - Kai Yang
- The No. 2 Hospital of Nanjing, Nanjing, Jiangsu Province, China
| | | | - Xian-Bin Kong
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yue Tu
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin, Tianjin, China
| | - Ke-Feng Bian
- Logistics University of People's Armed Police Force, Tianjin, China
| | - Zhen-Lin Liu
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin, Tianjin, China
| | - Xu-Yi Chen
- Affiliated Hospital of Logistics University of Chinese People's Armed Police Forces, Institute of Traumatic Brain Trauma and Neurological of CAPF, Neurotrauma Repair Key Laboratory of Tianjin, Tianjin, China
| |
Collapse
|
20
|
Effects of Linagliptin on Vessel Wall Healing in the Rat Model of Arterial Injury Under Normal and Diabetic Conditions. J Cardiovasc Pharmacol 2017; 69:101-109. [PMID: 27875385 DOI: 10.1097/fjc.0000000000000447] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Diabetic patients suffer an increased risk of restenosis and late stent thrombosis after angioplasty, complications which are related to a defective reendothelialization. Dipeptidyl peptidase-4 inhibitors have been suggested to exert a direct effect on endothelial and smooth muscle cells (SMCs). Therefore, the objective was to study if the dipeptidyl peptidase-4 inhibitor linagliptin could influence vascular repair and accelerate reendothelialization after arterial injury in healthy and diabetic animals. Diabetic Goto-Kakizaki and healthy Wistar rats were subjected to arterial injury and treated with linagliptin or vehicle. Vessel wall healing was monitored noninvasively using ultrasound, and on sacrifice, with Evans blue staining and immunohistochemistry. The effect of linagliptin on SMCs was also studied in vitro. We found that linagliptin reduced the proliferation and dedifferentiation of SMCs in vitro, and modulated the inflammatory response in the SMCs after arterial injury in vivo. However, these effects of linagliptin did not affect the neointima formation or the reendothelialization under normal and diabetic conditions. Although linagliptin did not influence vessel wall healing, it seems to possess a desirable antiproliferative influence on SMCs in vitro and an antiinflammatory effect in vivo. These pharmacological properties might carry a potential significance for favorable outcome after vascular interventions in diabetic patients.
Collapse
|
21
|
Doytcheva P, Bächler T, Tarasco E, Marzolla V, Engeli M, Pellegrini G, Stivala S, Rohrer L, Tona F, Camici GG, Vanhoutte PM, Matter CM, Lutz TA, Lüscher TF, Osto E. Inhibition of Vascular c-Jun N-Terminal Kinase 2 Improves Obesity-Induced Endothelial Dysfunction After Roux-en-Y Gastric Bypass. J Am Heart Assoc 2017; 6:JAHA.117.006441. [PMID: 29138180 PMCID: PMC5721746 DOI: 10.1161/jaha.117.006441] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background Roux‐en‐Y gastric bypass (RYGB) reduces obesity‐associated comorbidities and cardiovascular mortality. RYGB improves endothelial dysfunction, reducing c‐Jun N‐terminal kinase (JNK) vascular phosphorylation. JNK activation links obesity with insulin resistance and endothelial dysfunction. Herein, we examined whether JNK1 or JNK2 mediates obesity‐induced endothelial dysfunction and if pharmacological JNK inhibition can mimic RYGB vascular benefits. Methods and Results After 7 weeks of a high‐fat high‐cholesterol diet, obese rats underwent RYGB or sham surgery; sham–operated ad libitum–fed rats received, for 8 days, either the control peptide D‐TAT or the JNK peptide inhibitor D‐JNKi‐1 (20 mg/kg per day subcutaneous). JNK peptide inhibitor D‐JNKi‐1 treatment improved endothelial vasorelaxation in response to insulin and glucagon‐like peptide‐1, as observed after RYGB. Obesity increased aortic phosphorylation of JNK2, but not of JNK1. RYGB and JNK peptide inhibitor D‐JNKi‐1 treatment blunted aortic JNK2 phosphorylation via activation of glucagon‐like peptide‐1–mediated signaling. The inhibitory phosphorylation of insulin receptor substrate‐1 was reduced, whereas the protein kinase B/endothelial NO synthase pathway was increased and oxidative stress was decreased, resulting in improved vascular NO bioavailability. Conclusions Decreased aortic JNK2 phosphorylation after RYGB rapidly improves obesity‐induced endothelial dysfunction. Pharmacological JNK inhibition mimics the endothelial protective effects of RYGB. These findings highlight the therapeutic potential of novel strategies targeting vascular JNK2 against the severe cardiovascular disease associated with obesity.
Collapse
Affiliation(s)
- Petia Doytcheva
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Thomas Bächler
- Department of Surgery, Cantonal Hospital Fribourg, Fribourg, Switzerland
| | - Erika Tarasco
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Vincenzo Marzolla
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Laboratory of Cardiovascular Endocrinology, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Pisana, Rome, Italy
| | - Michael Engeli
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland
| | - Giovanni Pellegrini
- Laboratory for Animal Model Pathology, Institute for Veterinary Pathology, Vetsuisse Faculty University of Zurich, Switzerland
| | - Simona Stivala
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Lucia Rohrer
- Institute of Clinical Chemistry, University Hospital Zurich, Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Francesco Tona
- Department of Cardiac, Thoracic and Vascular Sciences, University of Padova, Italy
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Paul M Vanhoutte
- State Key Laboratory for Pharmaceutical Biotechnologies & Department of Pharmacology & Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Schwerzenbach, Switzerland
| | - Christian M Matter
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Thomas A Lutz
- Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Switzerland.,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland
| | - Elena Osto
- Center for Molecular Cardiology, University of Zurich, Switzerland .,University Heart Center, Cardiology, University Hospital Zurich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Switzerland.,Laboratory of Translational Nutrition Biology Federal Institute of Technology Zurich (ETHZ), Schwerzenbach, Switzerland
| |
Collapse
|
22
|
Leonardini A, D'Oria R, Incalza MA, Caccioppoli C, Andrulli Buccheri V, Cignarelli A, Paparella D, Margari V, Natalicchio A, Perrini S, Giorgino F, Laviola L. GLP-1 Receptor Activation Inhibits Palmitate-Induced Apoptosis via Ceramide in Human Cardiac Progenitor Cells. J Clin Endocrinol Metab 2017; 102:4136-4147. [PMID: 28938428 DOI: 10.1210/jc.2017-00970] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/11/2017] [Indexed: 11/19/2022]
Abstract
CONTEXT Increased apoptosis of cardiomyocytes and cardiac progenitor cells (CPCs) in response to saturated fatty acids (SFAs) can lead to myocardial damage and dysfunction. Ceramides mediate lipotoxicity-induced apoptosis. Glucagonlike peptide-1 receptor (GLP1R) agonists exert beneficial effects on cardiac cells in experimental models. OBJECTIVE To investigate the protective effects of GLP1R activation on SFA-mediated apoptotic death of human CPCs. DESIGN Human CPCs were isolated from cardiac appendages of nondiabetic donors and then exposed to palmitate with or without pretreatment with the GLP1R agonist exendin-4. Ceramide accumulation was evaluated by immunofluorescence. Expression of key enzymes in de novo ceramide biosynthesis was studied by quantitative reverse-transcription polymerase chain reaction and immunoblotting. Apoptosis was evaluated by measuring release of oligonucleosomes, caspase-3 cleavage, caspase activity, and terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling. RESULTS Exposure of the CPCs to palmitate resulted in 2.3- and 1.9-fold higher expression of ceramide synthase 5 (CERS5) and ceramide desaturase-1, respectively (P < 0.05). This was associated with intracellular accumulation of ceramide and activation of c-Jun NH2-terminal protein kinase (JNK) signaling and apoptosis (P < 0.05). Both coincubation with fumonisin B1, a specific ceramide synthase inhibitor, and CERS5 knockdown prevented ceramide accumulation, JNK activation, and apoptosis in response to palmitate (P < 0.05). Exendin-4 also prevented the activation of the ceramide biosynthesis and JNK in response to palmitate, inhibiting apoptosis (P < 0.05). CONCLUSIONS Excess palmitate results in activation of ceramide biosynthesis, JNK signaling, and apoptosis in human CPCs. GLP1R activation counteracts this lipotoxic damage via inhibition of ceramide generation, and this may represent a cardioprotective mechanism.
Collapse
Affiliation(s)
- Anna Leonardini
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari Aldo Moro, I-70124 Bari, Italy
| | - Rossella D'Oria
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari Aldo Moro, I-70124 Bari, Italy
| | - Maria Angela Incalza
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari Aldo Moro, I-70124 Bari, Italy
| | - Cristina Caccioppoli
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari Aldo Moro, I-70124 Bari, Italy
| | - Valentina Andrulli Buccheri
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari Aldo Moro, I-70124 Bari, Italy
| | - Angelo Cignarelli
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari Aldo Moro, I-70124 Bari, Italy
| | - Domenico Paparella
- Department of Emergency and Organ Transplantation, Section of Cardiac Surgery, University of Bari Aldo Moro, I-70124 Bari, Italy
- Cardiac Surgery, Santa Maria Hospital, I-70124 Bari, Italy
| | - Vito Margari
- Cardiac Surgery, Santa Maria Hospital, I-70124 Bari, Italy
| | - Annalisa Natalicchio
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari Aldo Moro, I-70124 Bari, Italy
| | - Sebastio Perrini
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari Aldo Moro, I-70124 Bari, Italy
| | - Francesco Giorgino
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari Aldo Moro, I-70124 Bari, Italy
| | - Luigi Laviola
- Department of Emergency and Organ Transplantation, Section of Internal Medicine, Endocrinology, Andrology, and Metabolic Diseases, University of Bari Aldo Moro, I-70124 Bari, Italy
| |
Collapse
|
23
|
Niu B, Li C, Su H, Li Q, He Q, Liu L, Xue Y, Shen T, Xia X. Glucagon-like peptide-1 receptor agonist exendin-4 protects against interleukin-1β-mediated inhibition of glucose-stimulated insulin secretion by mouse insulinoma β cells. Exp Ther Med 2017; 14:2671-2676. [PMID: 28947919 DOI: 10.3892/etm.2017.4803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Accepted: 11/04/2016] [Indexed: 01/01/2023] Open
Abstract
The aim of the present study was to investigate the protective effect of the glucagon-like peptide-1 receptor agonist exendin-4 on the interleukin (IL)-1β-induced impairment of glucose-stimulated insulin secretion (GSIS) in β-TC-6 cells. β-TC-6 cells were pretreated with various concentrations of IL-1β (0.15, 1.5 or 15 ng/ml) and exendin-4 (0.1 or 1 mM). Exendin-4 was administered to β-TC-6 cells prior to, during and following pretreatment. Cells were stimulated with various concentrations of glucose (0, 1.38, 5.5 and 11.1 mM), and insulin was measured via radioimmunoassay of the supernatant; furthermore, western blot analysis was used to detect phosphorylated extracellular receptor kinase (ERK)1/2. The insulin levels (151.08±14.34 µIU/ml) and ERK1/2 phosphorylation in β-TC-6 cells peaked in response to 1.38 mM glucose stimulation compared with 0, 5.5 and 11.1 mM glucose stimulation. IL-1β inhibited GSIS in a dose-dependent manner: Insulin levels were 83.76±1.16 µIU/ml when 0.15 ng/ml IL-1β was added under GSIS, 59.46±3.20 µIU/ml when 1.5 ng/ml IL-1β was added under GSIS, and 56.98±1.19 µIU/ml when 15 ng/ml IL-1β was added under GSIS. Exendin-4 exerted a protective effect against IL-1β-induced GSIS inhibition in a dose-dependent manner. The greatest protective effect was observed when exendin-4 was added prior to IL-1β pretreatment, which was statistically significant (P<0.05). These findings suggested that exendin-4 was able to reverse the IL-1β-induced inhibition of ERK1/2 phosphorylation and serves a protective role by impairing GSIS induced by IL-1β in β-TC-6 cells. This mechanism may be associated with the recovery of ERK1/2 activation.
Collapse
Affiliation(s)
- Ben Niu
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China.,Department of Endocrinology, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan, Kunming, Yunnan 650032, P.R. China
| | - Chao Li
- Department of Emergency Medicine, The People's Hospital of Yuxi City, Yuxi, Yunnan 653100, P.R. China
| | - Heng Su
- Department of Endocrinology, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan, Kunming, Yunnan 650032, P.R. China
| | - Qingzhu Li
- Department of Endocrinology, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan, Kunming, Yunnan 650032, P.R. China
| | - Qiu He
- Department of Endocrinology, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan, Kunming, Yunnan 650032, P.R. China
| | - Lijuan Liu
- Department of Cadre Ward, WISCO General Hospital, Wuhan, Hubei 430080, P.R. China
| | - Yuanming Xue
- Department of Endocrinology, The Affiliated Hospital of Kunming University of Science and Technology, The First People's Hospital of Yunnan, Kunming, Yunnan 650032, P.R. China
| | - Tao Shen
- Institute of Basic and Clinical Medicine, Center of Clinical Molecular Biology of Yunnan, Affiliated Hospital of Kunming University of Science and Technology, First People's Hospital of Yunnan, Kunming, Yunnan 650032, P.R. China
| | - Xueshan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
24
|
Incalza MA, D'Oria R, Natalicchio A, Perrini S, Laviola L, Giorgino F. Oxidative stress and reactive oxygen species in endothelial dysfunction associated with cardiovascular and metabolic diseases. Vascul Pharmacol 2017; 100:1-19. [PMID: 28579545 DOI: 10.1016/j.vph.2017.05.005] [Citation(s) in RCA: 786] [Impact Index Per Article: 98.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 05/21/2017] [Accepted: 05/31/2017] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) are reactive intermediates of molecular oxygen that act as important second messengers within the cells; however, an imbalance between generation of reactive ROS and antioxidant defense systems represents the primary cause of endothelial dysfunction, leading to vascular damage in both metabolic and atherosclerotic diseases. Endothelial activation is the first alteration observed, and is characterized by an abnormal pro-inflammatory and pro-thrombotic phenotype of the endothelial cells lining the lumen of blood vessels. This ultimately leads to reduced nitric oxide (NO) bioavailability, impairment of the vascular tone and other endothelial phenotypic changes collectively termed endothelial dysfunction(s). This review will focus on the main mechanisms involved in the onset of endothelial dysfunction, with particular focus on inflammation and aberrant ROS production and on their relationship with classical and non-classical cardiovascular risk factors, such as hypertension, metabolic disorders, and aging. Furthermore, new mediators of vascular damage, such as microRNAs, will be discussed. Understanding mechanisms underlying the development of endothelial dysfunction is an important base of knowledge to prevent vascular damage in metabolic and cardiovascular diseases.
Collapse
Affiliation(s)
- Maria Angela Incalza
- Department of Emergency and Organ Transplantation, Section on Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Rossella D'Oria
- Department of Emergency and Organ Transplantation, Section on Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Annalisa Natalicchio
- Department of Emergency and Organ Transplantation, Section on Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Sebastio Perrini
- Department of Emergency and Organ Transplantation, Section on Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Luigi Laviola
- Department of Emergency and Organ Transplantation, Section on Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy
| | - Francesco Giorgino
- Department of Emergency and Organ Transplantation, Section on Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari, Italy.
| |
Collapse
|
25
|
Li L, Huang B, Song S, Sohun H, Rao Z, Tao L, Jin Q, Zeng J, Wu R, Ji K, Lin J, Wu L, Chu M. A20 functions as mediator in TNFα-induced injury of human umbilical vein endothelial cells through TAK1-dependent MAPK/eNOS pathway. Oncotarget 2017; 8:65230-65239. [PMID: 29029426 PMCID: PMC5630326 DOI: 10.18632/oncotarget.18191] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 04/27/2017] [Indexed: 12/15/2022] Open
Abstract
A20, a negative regulator of nuclear factor κB signaling, has been shown to attenuate atherosclerotic events. Transforming growth factor beta-activated kinase 1 (TAK1) plays a critical role in TNFα-induced atherosclerosis via endothelial nitric oxide (NO) synthase (eNOS) uncoupling and NO reduction. In the study, we investigated the hypothesis that A20 protected endothelial cell injury induced by TNFα through modulating eNOS activity and TAK1 signalling. Human umbilical vein endothelial cells (HUVECs) were stimulated by TNFα. The impact of A20 on cell apoptosis, eNOS expression and NO production and related TAK1 pathway were detected. Both eNOS and NO production were remarkably reduced. TAK1, p38 MAPK phosphorylation and HUVECs apoptosis were enhanced after TNFα stimulation for 2 hrs. Inhibition of A20 significantly activated TAK1, p38 MAPK phosphorylation, and cell apoptosis, but blocked eNOS expression and NO production. Furthermore, p38 MAPK expression was suppressed by A20 over-expression, but re-enhanced by inhibiting A20 or activation of TAK1. Furtherly, TNFα-induced suppression of eNOS and NO production were largely prevented by silencing p38 MAPK. Collectively, our results suggested that A20-mediated TAK1 inactivation suppresses p38 MAPK and regulated MAPK/eNOS pathway, which contributes to endothelial cell survival and function preservation.
Collapse
Affiliation(s)
- Lei Li
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | | | - Shiyang Song
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Hareshwaree Sohun
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Zhiheng Rao
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Luyuan Tao
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Qike Jin
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jingjing Zeng
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Rongzhou Wu
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Kangting Ji
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jiafeng Lin
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Lianpin Wu
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Maoping Chu
- Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
26
|
Li J, Zheng J, Wang S, Lau HK, Fathi A, Wang Q. Cardiovascular Benefits of Native GLP-1 and its Metabolites: An Indicator for GLP-1-Therapy Strategies. Front Physiol 2017; 8:15. [PMID: 28194113 PMCID: PMC5276855 DOI: 10.3389/fphys.2017.00015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 01/09/2017] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular disease is a common co-morbidity and leading cause of death in patients with type 2 diabetes mellitus (T2DM). Glucagon-like peptide 1 (GLP-1) is a peptide hormone produced by intestinal L cells in response to feeding. Native GLP-1 (7-36) amide is rapidly degraded by diaminopeptidyl peptidase-4 (DPP4) to GLP-1 (9-36) amide, making 9-36a the major circulating form. While it is 7-36a, and not its metabolites, which exerts trophic effects on islet β-cells, recent studies suggest that both 7-36a and its metabolites have direct cardiovascular effects, including preserving cardiomyocyte viability, ameliorating cardiac function, and vasodilation. In particular, the difference in cardiovascular effects between 7-36a and 9-36a is attracting attention. Growing evidence has strengthened the presumption that their cardiovascular effects are overlapping, but distinct and complementary to each other; 7-36a exerts cardiovascular effects in a GLP-1 receptor (GLP-1R) dependent pathway, whereas 9-36a does so in a GLP-1R independent pathway. GLP-1 therapies have been developed using two main strategies: DPP4-resistant GLP-1 analogs/GLP-1R agonists and DPP4 inhibitors, which both aim to prolong the life-time of circulating 7-36a. One prominent concern that should be addressed is that the cardiovascular benefits of 9-36a are lacking in these strategies. This review attempts to differentiate the cardiovascular effects between 7-36a and 9-36a in order to provide new insights into GLP-1 physiology, and facilitate our efforts to develop a superior GLP-1-therapy strategy for T2DM and cardiovascular diseases.
Collapse
Affiliation(s)
- Junfeng Li
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical College, Fudan UniversityShanghai, China; Department of Endocrinology, Renmin Hospital of Wuhan UniversityWuhan, China
| | - Juan Zheng
- Division of Endocrinology and Metabolism, the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's HospitalToronto, ON, Canada; Department of Physiology, Faculty of Medicine, University of TorontoToronto, ON, Canada
| | - Susanne Wang
- Division of Endocrinology and Metabolism, the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's HospitalToronto, ON, Canada; Department of Physiology, Faculty of Medicine, University of TorontoToronto, ON, Canada
| | - Harry K Lau
- Division of Endocrinology and Metabolism, the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's HospitalToronto, ON, Canada; Department of Physiology, Faculty of Medicine, University of TorontoToronto, ON, Canada
| | - Ali Fathi
- Division of Endocrinology and Metabolism, the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's Hospital Toronto, ON, Canada
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical College, Fudan UniversityShanghai, China; Division of Endocrinology and Metabolism, the Keenan Research Centre in the Li Ka Shing Knowledge Institute, St. Michael's HospitalToronto, ON, Canada; Department of Physiology, Faculty of Medicine, University of TorontoToronto, ON, Canada
| |
Collapse
|
27
|
Pujadas G, Drucker DJ. Vascular Biology of Glucagon Receptor Superfamily Peptides: Mechanistic and Clinical Relevance. Endocr Rev 2016; 37:554-583. [PMID: 27732058 DOI: 10.1210/er.2016-1078] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Regulatory peptides produced in islet and gut endocrine cells, including glucagon, glucagon-like peptide-1 (GLP-1), GLP-2, and glucose-dependent insulinotropic polypeptide, exert actions with considerable metabolic importance and translational relevance. Although the clinical development of GLP-1 receptor agonists and dipeptidyl peptidase-4 inhibitors has fostered research into how these hormones act on the normal and diseased heart, less is known about the actions of these peptides on blood vessels. Here we review the effects of these peptide hormones on normal blood vessels and highlight their vascular actions in the setting of experimental and clinical vascular injury. The cellular localization and signal transduction properties of the receptors for glucagon, GLP-1, GLP-2, and glucose-dependent insulinotropic polypeptide are discussed, with emphasis on endothelial cells and vascular smooth muscle cells. The actions of these peptides on the control of blood flow, blood pressure, angiogenesis, atherosclerosis, and vascular inflammation are reviewed with a focus on elucidating direct and indirect mechanisms of action. How these peptides traverse the blood-brain barrier is highlighted, with relevance to the use of GLP-1 receptor agonists to treat obesity and neurodegenerative disorders. Wherever possible, we compare actions identified in cell lines and primary cell culture with data from preclinical studies and, when available, results of human investigation, including studies in subjects with diabetes, obesity, and cardiovascular disease. Throughout the review, we discuss pitfalls, limitations, and challenges of the existing literature and highlight areas of controversy and uncertainty. The increasing use of peptide-based therapies for the treatment of diabetes and obesity underscores the importance of understanding the vascular biology of peptide hormone action.
Collapse
Affiliation(s)
- Gemma Pujadas
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Daniel J Drucker
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, ON M5G 1X5, Canada
| |
Collapse
|
28
|
Bułdak Ł, Machnik G, Bułdak RJ, Łabuzek K, Bołdys A, Okopień B. Exenatide and metformin express their anti-inflammatory effects on human monocytes/macrophages by the attenuation of MAPKs and NFκB signaling. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:1103-15. [PMID: 27424158 DOI: 10.1007/s00210-016-1277-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/10/2016] [Indexed: 12/13/2022]
Abstract
Metformin and exenatide are effective antidiabetic drugs, and they seem to have pleiotropic properties improving cardiovascular outcomes. Macrophages' phenotype is essential in the development of atherosclerosis, and it can be modified during antidiabetic therapy, resulting in attenuated atherogenesis. The mechanism orchestrating this phenomenon is not fully clear. We examined the impact of exenatide and metformin on the level of TNF alpha, MCP-1, reactive oxygen species (ROS), and the activation of mitogen-activated protein kinases (MAPK), nuclear factor kappa B (NFκB), and CCAAT/enhancer-binding protein beta (C/EBP beta) in human monocytes/macrophages. We found that both drugs reduced levels of TNF alpha, ROS, and NFκB binding activity to a similar extent. Compared to metformin, exenatide was more effective in reducing MCP-1 levels. We noted that Compound C (AMPK inhibitor) reduced the impact of exenatide on cytokines, ROS, and NFκB in cultures. Both drugs elevated the C/EBP beta phosphorylation level. Experiments on MAPKs showed effective inhibitory potential of exenatide toward p38, JNK, and ERK, whereas metformin inhibited JNK and ERK only. Exenatide was more effective in the inhibition of JNK than metformin. Interestingly, an in vitro setting additive effect of drugs was absent. In conclusion, here, we report that metformin and exenatide inhibit the proinflammatory phenotype of human monocytes/macrophages via influence on MAPK, C/EBP beta, and NFκB. Exenatide was more effective than metformin in reducing MCP-1 expression and JNK activity. We also showed that some effects of exenatide relied on AMPK activation. This shed light on the possible mechanisms responsible for pleiotropic effects of metformin and exenatide.
Collapse
Affiliation(s)
- Łukasz Bułdak
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Medykow 18, 40-752, Katowice, Poland.
| | - Grzegorz Machnik
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Medykow 18, 40-752, Katowice, Poland
| | - Rafał Jakub Bułdak
- Department of Physiology, School of Medicine in Zabrze, Medical University of Silesia, Jordana 19, 41-808, Zabrze, Poland
| | - Krzysztof Łabuzek
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Medykow 18, 40-752, Katowice, Poland
| | - Aleksandra Bołdys
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Medykow 18, 40-752, Katowice, Poland
| | - Bogusław Okopień
- Department of Internal Medicine and Clinical Pharmacology, School of Medicine in Katowice, Medical University of Silesia, Medykow 18, 40-752, Katowice, Poland
| |
Collapse
|
29
|
Wei R, Ma S, Wang C, Ke J, Yang J, Li W, Liu Y, Hou W, Feng X, Wang G, Hong T. Exenatide exerts direct protective effects on endothelial cells through the AMPK/Akt/eNOS pathway in a GLP-1 receptor-dependent manner. Am J Physiol Endocrinol Metab 2016; 310:E947-57. [PMID: 27072494 DOI: 10.1152/ajpendo.00400.2015] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 04/12/2016] [Indexed: 01/30/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) may have direct favorable effects on cardiovascular system. The aim of this study was to investigate the effects of the GLP-1 analog exenatide on improving coronary endothelial function in patients with type 2 diabetes and to investigate the underlying mechanisms. The newly diagnosed type 2 diabetic subjects were enrolled and given either lifestyle intervention or lifestyle intervention plus exenatide treatment. After 12-wk treatment, coronary flow velocity reserve (CFVR), an important indicator of coronary endothelial function, was improved significantly, and serum levels of soluble intercellular adhesion molecule-1 (sICAM-1) and soluble vascular cell adhesion molecule-1 (sVCAM-1) were remarkably decreased in the exenatide treatment group compared with the baseline and the control group. Notably, CFVR was correlated inversely with hemoglobin A1c (Hb A1c) and positively with high-density lipoprotein cholesterol (HDL-C). In human umbilical vein endothelial cells, exendin-4 (a form of exenatide) significantly increased NO production, endothelial NO synthase (eNOS) phosphorylation, and GTP cyclohydrolase 1 (GTPCH1) level in a dose-dependent manner. The GLP-1 receptor (GLP-1R) antagonist exendin (9-39) or GLP-1R siRNA, adenylyl cyclase inhibitor SQ-22536, AMPK inhibitor compound C, and PI3K inhibitor LY-294002 abolished the effects of exendin-4. Furthermore, exendin-4 reversed homocysteine-induced endothelial dysfunction by decreasing sICAM-1 and reactive oxygen species (ROS) levels and upregulating NO production and eNOS phosphorylation. Likewise, exendin (9-39) diminished the protective effects of exendin-4 on the homocysteine-induced endothelial dysfunction. In conclusion, exenatide significantly improves coronary endothelial function in patients with newly diagnosed type 2 diabetes. The effect may be mediated through activation of AMPK/PI3K-Akt/eNOS pathway via a GLP-1R/cAMP-dependent mechanism.
Collapse
Affiliation(s)
- Rui Wei
- Department of Endocrinology and Metabolism and
| | - Shifeng Ma
- Department of Endocrinology and Metabolism and
| | - Chen Wang
- Department of Endocrinology and Metabolism and
| | - Jing Ke
- Department of Endocrinology and Metabolism and
| | - Jin Yang
- Department of Endocrinology and Metabolism and
| | - Weihong Li
- Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Ye Liu
- Department of Endocrinology and Metabolism and
| | - Wenfang Hou
- Department of Endocrinology and Metabolism and
| | - Xinheng Feng
- Department of Cardiology, Peking University Third Hospital, Beijing, China
| | - Guang Wang
- Department of Endocrinology and Metabolism and
| | | |
Collapse
|
30
|
Zhang Y, Zhou H, Wu W, Shi C, Hu S, Yin T, Ma Q, Han T, Zhang Y, Tian F, Chen Y. Liraglutide protects cardiac microvascular endothelial cells against hypoxia/reoxygenation injury through the suppression of the SR-Ca(2+)-XO-ROS axis via activation of the GLP-1R/PI3K/Akt/survivin pathways. Free Radic Biol Med 2016; 95:278-92. [PMID: 27038735 DOI: 10.1016/j.freeradbiomed.2016.03.035] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 03/04/2016] [Accepted: 03/29/2016] [Indexed: 01/15/2023]
Abstract
Microvascular endothelial cells (CMECs) oxidative damage resulting from hypoxia/reoxygenation (H/R) injury is responsible for microcirculation perfusion disturbances and the progression of cardiac dysfunction. However, few strategies are available to reverse such pathologies. Here, we studied the effects and mechanisms of liraglutide on CEMCs oxidative damage, focusing in particular on calcium overload-triggered free radical injury signals and the GLP-1R/PI3K/Akt/survivin survival pathways. The results indicate that H/R increased IP3R expression but reduced SERCA2a expression, which rapidly raised intracellular Ca(2+) levels, subsequently leading to Ca(2+)-dependent xanthine oxidase (XO) activation, reactive oxygen species (ROS) production and the cellular apoptosis of CMECs. However, liraglutide pretreatment abrogated Ca(2+)-mediated oxidative apoptosis. Furthermore, liraglutide regulated the rate of IP3R/SERCA2a gene transcription and conserved SERCA2a-ATPase activity via the maintenance of ATP production under H/R, which drove excessive Ca(2+) reflux to the sarcoplasmic reticulum (SR) and inhibited Ca(2+) release from the SR, ultimately restoring Ca(2+) homeostasis. Furthermore, the regulatory role of liraglutide on Ca(2+) balance in conjunction with its up-regulation of superoxide dismutase, glutathione and glutathione peroxidase collectively scavenged the excess ROS under H/R. Moreover, we showed that liraglutide strengthened Akt phosphorylation and subsequently survivin expression. In addition, both the blockade of the GLP-1R/PI3K/Akt pathways and the siRNA-mediated knockdown of survivin abolished the protective effects of liraglutide on SR-Ca(2+) function and CMECs oxidative apoptosis. In summary, this study confirmed that H/R induced CMECs oxidative damage through the SR-Ca(2+)-XO-ROS injury signals and that liraglutide pretreatment may suppress such CMECs damage through the PI3K/Akt/survivin pathways.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| | - Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| | - Wenbo Wu
- Department of Burn surgery and Plastic surgery, First Hospital Affiliated to the Chinese PLA General Hospital, Beijing, China
| | - Chen Shi
- Department of Radiotherapy, Beijing Cancer Hospital, Beijing, China
| | - Shunying Hu
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| | - Tong Yin
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| | - Qiang Ma
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| | - Tianwen Han
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| | - Yingqian Zhang
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| | - Feng Tian
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China
| | - Yundai Chen
- Department of Cardiology, Chinese PLA General Hospital, #28 Fuxing Road, Beijing 100853, China.
| |
Collapse
|
31
|
Abstract
Glucagon-like peptide-1 (GLP-1) is a peptide hormone, released from intestinal L-cells in response to hormonal, neural and nutrient stimuli. In addition to potentiation of meal-stimulated insulin secretion, GLP-1 signalling exerts numerous pleiotropic effects on various tissues, regulating energy absorption and disposal, as well as cell proliferation and survival. In Type 2 Diabetes (T2D) reduced plasma levels of GLP-1 have been observed, and plasma levels of GLP-1, as well as reduced numbers of GLP-1 producing cells, have been correlated to obesity and insulin resistance. Increasing endogenous secretion of GLP-1 by selective targeting of the molecular mechanisms regulating secretion from the L-cell has been the focus of much recent research. An additional and promising strategy for enhancing endogenous secretion may be to increase the L-cell mass in the intestinal epithelium, but the mechanisms that regulate the growth, survival and function of these cells are largely unknown. We recently showed that prolonged exposure to high concentrations of the fatty acid palmitate induced lipotoxic effects, similar to those operative in insulin-producing cells, in an in vitro model of GLP-1-producing cells. The mechanisms inducing this lipototoxicity involved increased production of reactive oxygen species (ROS). In this review, regulation of GLP-1-secreting cells is discussed, with a focus on the mechanisms underlying GLP-1 secretion, long-term regulation of growth, differentiation and survival under normal as well as diabetic conditions of hypernutrition.
Collapse
|
32
|
Petersen KE, Rakipovski G, Raun K, Lykkesfeldt J. Does Glucagon-like Peptide-1 Ameliorate Oxidative Stress in Diabetes? Evidence Based on Experimental and Clinical Studies. Curr Diabetes Rev 2016; 12:331-358. [PMID: 26381142 PMCID: PMC5101636 DOI: 10.2174/1573399812666150918150608] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 09/15/2015] [Accepted: 09/18/2015] [Indexed: 02/07/2023]
Abstract
Glucagon-like peptide-1 (GLP-1) has shown to influence the oxidative stress status in a number of in vitro, in vivo and clinical studies. Well-known effects of GLP-1 including better glycemic control, decreased food intake, increased insulin release and increased insulin sensitivity may indirectly contribute to this phenomenon, but glucose-independent effects on ROS level, production and antioxidant capacity have been suggested to also play a role. The potential 'antioxidant' activity of GLP-1 along with other proposed glucose-independent modes of action related to ameliorating redox imbalance remains a controversial topic but could hold a therapeutic potential against micro- and macrovascular diabetic complications. This review discusses the presently available knowledge from experimental and clinical studies on the effects of GLP-1 on oxidative stress in diabetes and diabetes-related complications.
Collapse
Affiliation(s)
| | | | | | - Jens Lykkesfeldt
- Faculty of Health and Medical Sciences, University of Copenhagen, DK-1870, Frederiksberg C, Denmark.
| |
Collapse
|
33
|
Exenatide (a GLP-1 agonist) expresses anti-inflammatory properties in cultured human monocytes/macrophages in a protein kinase A and B/Akt manner. Pharmacol Rep 2015; 68:329-37. [PMID: 26922535 DOI: 10.1016/j.pharep.2015.10.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 10/21/2015] [Accepted: 10/22/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Incretin-based therapies in the treatment of type 2 diabetes mellitus are associated with significant improvements in glycemic control, which are accompanied by a beneficial impact on atherosclerosis. Macrophages are essential in the development of atherosclerotic plaques and may develop features that accelerate atherosclerosis (classically activated macrophages) or protect arterial walls against it (alternatively activated macrophages). Therefore, we explored whether beneficial actions of exenatide are connected with the influence on the macrophages' phenotype and synthesis of inflammatory and anti-inflammatory cytokines. METHODS Monocytes/macrophages were harvested from 10 healthy subjects. Cells were cultured in the presence of exenatide, exendin 9-39 (GLP-1 antagonist), LPS, IL-4, PKI (PKA inhibitor) and triciribine (PKB/Akt inhibitor). We measured the effects of the above-mentioned compounds on markers of macrophages' phenotype (inducible nitrous oxide (iNOS), arginase 1 (arg1) and mannose receptors) and concentration of nitrite, IL-1β, TNF-α and IL-10. RESULTS Exenatide significantly increased the level of IL-10 and decreased both TNF-α and IL-1β in LPS-treated monocytes/macrophages. Furthermore exenatide increased the expression of arg1-a marker of classical activation and reduced the LPS-induced expression of iNOS-a marker of classical activation. According to experiments with protein kinases inhibitors we found that proinflammatory markers were protein kinase A dependent, whereas the activation of alternative activation was similarly reliant on protein kinase A and B/Akt. CONCLUSIONS We showed that exenatide skewed the macrophages phenotype toward anti-inflammatory phenotype and this effect is predominantly attributable to protein kinase A and to a less extent to B/Akt activation.
Collapse
|
34
|
Guo L, Qiao Y, Zhang L, Pan Q. Protective Role of Glucagon-Like Peptide-1 Against High-Glucose-Induced Endothelial Oxidative Damage. Medicine (Baltimore) 2015; 94:e2055. [PMID: 26632709 PMCID: PMC5058978 DOI: 10.1097/md.0000000000002055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
To investigate the protective effect of glucagon-like peptide-1 (GLP-1) against cell damage induced by high glucose.Human umbilical vein endothelial cells (HUVECs) were divided into control group (5.5 mmol/L) and high glucose groups (19, 33, or 47 mmol/L), which were cultured with different concentrations of glucose for 48 hours, respectively. Cell viability was measured with MTT assay. Levels of intracellular reactive oxygen species (ROS) were monitored by flow cytometry and apoptotic cell death was measured by staining with Annexin V-FITC and propidium iodide. Cultured cells were detected with intercellular adhesion molecule 1 (ICAM-1), VCAM-1, and JNK on protein.Compared with the control group, cell viability was decreased by 20% and 37%, respectively, when cultured under 33 and 47 mM, while increased in different GLP-1-treated groups (0.01 L, 0.1, 1, and 10 nmol/L). The GLP-1 treatment significantly reduced the ROS level of high glucose treatment group but not impact on the control group. Meanwhile, the level of apoptosis was elevated in the high glucose treatment group. Early apoptosis was significantly reversed in the GLP-1-treated group (0.1, 1, and 10 nmol/L). Late apoptosis was uniquely decreased in the GLP-1 concentrations of 10 nmol/L. Furthermore, GLP-1 could also reduce the protein levels of ICAM-1, VCAM-1, and phospho JNK in the endothelial cells with high glucose treatment.GLP-1 could inhibit cell apoptosis and reduce ROS generation and JNK-Bax signaling pathway activation, which were induced by high glucose treatment.
Collapse
Affiliation(s)
- Lixin Guo
- From the Department of Endocrinology, Beijing Hospital, Beijing, P.R. China
| | | | | | | |
Collapse
|
35
|
Huang C, Yuan L, Cao S. Endogenous GLP-1 as a key self-defense molecule against lipotoxicity in pancreatic islets. Int J Mol Med 2015; 36:173-85. [PMID: 25976560 PMCID: PMC4494597 DOI: 10.3892/ijmm.2015.2207] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 05/07/2015] [Indexed: 01/15/2023] Open
Abstract
The number of pro-α cells is known to increase in response to β cell injury and these cells then generate glucagon-like peptide-1 (GLP-1), thus attenuating the development of diabetes. The aim of the present study was to further examine the role and the mechanisms responsible for intra-islet GLP-1 production as a self-protective response against lipotoxicity. The levels of the key enzyme, prohormone convertase 1/3 (PC1/3), as well as the synthesis and release of GLP-1 in models of lipotoxicity were measured. Furthermore, islet viability, apoptosis, oxidative stress and inflammation, as well as islet structure were assessed after altering GLP-1 receptor signaling. Both prolonged exposure to palmitate and a high-fat diet facilitated PC1/3 expression, as well as the synthesis and release of GLP-1 induced by β cell injury and the generation of pro-α cells. Prolonged exposure to palmitate increased reactive oxygen species (ROS) production, and the antioxidant, N-acetylcysteine (NAC), partially prevented the detrimental effects induced by palmitate on β cells, resulting in decreased GLP-1 levels. Furthermore, the inhibition of GLP-1 receptor (GLP-1R) signaling by treatment with exendin‑(9-39) further decreased cell viability, increased cell apoptosis and caused a stronger inhibition of the β cell-specific transcription factor, pancreatic duodenal homeobox 1 (PDX1). Moreover, treatment with the GLP-1R agonist, liraglutide, normalized islet structure and function, resulting in a decrease in cell death and in the amelioration of β cell marker expression. Importantly, liraglutide maintained the oxidative balance and decreased inflammatory factor and p65 expression. Overall, our data demonstrate that an increase in the number of pro-α cells and the activation of the intra-islet GLP-1 system comprise a self-defense mechanism for enhancing β cell survival to combat lipid overload, which is in part mediated by oxidative stress and inflammation.
Collapse
Affiliation(s)
- Chenghu Huang
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shuyi Cao
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
36
|
Eriksson L, Saxelin R, Röhl S, Roy J, Caidahl K, Nyström T, Hedin U, Razuvaev A. Glucagon-Like Peptide-1 Receptor Activation Does not Affect Re-Endothelialization but Reduces Intimal Hyperplasia via Direct Effects on Smooth Muscle Cells in a Nondiabetic Model of Arterial Injury. J Vasc Res 2015; 52:41-52. [PMID: 25966620 DOI: 10.1159/000381097] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2014] [Accepted: 02/15/2015] [Indexed: 11/19/2022] Open
Abstract
UNLABELLED Diabetic patients have an increased risk of restenosis and late stent thrombosis after angioplasty, i.e. complications that are related to a defective re-endothelialization. Exendin-4, a stable glucagon-like peptide (GLP)-1 receptor agonist, has been suggested to influence the formation of intimal hyperplasia and to increase endothelial cell proliferation in vitro. Thus, the aim of this study was to investigate the mechanisms by which treatment with exendin-4 could influence re-endothelialization and intimal hyperplasia after vascular injury. METHODS Sprague-Dawley rats were subjected to balloon injury of the left common carotid artery and treated for 4 weeks with exendin-4 or vehicle. Intimal hyperplasia and vessel wall elasticity were monitored noninvasively by high-frequency ultrasound, and re-endothelialization was evaluated upon sacrifice using Evans blue dye. RESULTS AND CONCLUSION Exendin-4 selectively reduced the proliferation of smooth muscle cells (SMCs) and intimal hyperplasia in vivo without affecting the re-endothelialization process, but treatment with exendin-4 improved arterial wall elasticity. Our data also show that exendin-4 significantly decreased the proliferation and increased the apoptosis of SMCs in vitro, effects that appear to be mediated through cAMP signaling and endothelial nitric oxide synthase following GLP-1 receptor activation. Together, these effects of exendin-4 are highly desirable and may lead to an improved outcome for patients undergoing vascular interventions.
Collapse
Affiliation(s)
- Linnea Eriksson
- Department of Clinical Science and Education, Södersjukhuset, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Sathanoori R, Swärd K, Olde B, Erlinge D. The ATP Receptors P2X7 and P2X4 Modulate High Glucose and Palmitate-Induced Inflammatory Responses in Endothelial Cells. PLoS One 2015; 10:e0125111. [PMID: 25938443 PMCID: PMC4418812 DOI: 10.1371/journal.pone.0125111] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/20/2015] [Indexed: 12/20/2022] Open
Abstract
Endothelial cells lining the blood vessels are principal players in vascular inflammatory responses. Dysregulation of endothelial cell function caused by hyperglycemia, dyslipidemia, and hyperinsulinemia often result in impaired vasoregulation, oxidative stress, inflammation, and altered barrier function. Various stressors including high glucose stimulate the release of nucleotides thus initiating signaling via purinergic receptors. However, purinergic modulation of inflammatory responses in endothelial cells caused by high glucose and palmitate remains unclear. In the present study, we investigated whether the effect of high glucose and palmitate is mediated by P2X7 and P2X4 and if they play a role in endothelial cell dysfunction. Transcript and protein levels of inflammatory genes as well as reactive oxygen species production, endothelial-leukocyte adhesion, and cell permeability were investigated in human umbilical vein endothelial cells exposed to high glucose and palmitate. We report high glucose and palmitate to increase levels of extracellular ATP, expression of P2X7 and P2X4, and inflammatory markers. Both P2X7 and P2X4 antagonists inhibited high glucose and palmitate-induced interleukin-6 levels with the former having a significant effect on interleukin-8 and cyclooxygenase-2. The effect of the antagonists was confirmed with siRNA knockdown of the receptors. In addition, P2X7 mediated both high glucose and palmitate-induced increase in reactive oxygen species levels and decrease in endothelial nitric oxide synthase. Blocking P2X7 inhibited high glucose and palmitate-induced expression of intercellular adhesion molecule-1 and vascular cell adhesion molecule-1 as well as leukocyte-endothelial cell adhesion. Interestingly, high glucose and palmitate enhanced endothelial cell permeability that was dependent on both P2X7 and P2X4. Furthermore, antagonizing the P2X7 inhibited high glucose and palmitate-mediated activation of p38-mitogen activated protein kinase. These findings support a novel role for P2X7 and P2X4 coupled to induction of inflammatory molecules in modulating high glucose and palmitate-induced endothelial cell activation and dysfunction.
Collapse
Affiliation(s)
- Ramasri Sathanoori
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
- * E-mail:
| | - Karl Swärd
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Björn Olde
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| | - David Erlinge
- Department of Cardiology, Clinical Sciences, Lund University, Lund, Sweden
| |
Collapse
|
38
|
Lu K, Chang G, Ye L, Zhang P, Li Y, Zhang D. Protective effects of extendin‑4 on hypoxia/reoxygenation‑induced injury in H9c2 cells. Mol Med Rep 2015; 12:3007-16. [PMID: 25936390 DOI: 10.3892/mmr.2015.3682] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 02/24/2015] [Indexed: 11/05/2022] Open
Abstract
Glucagon-like peptide-1 (GLP-1) analogues are likely to exert cardioprotective effects via balancing the energy metabolism in cardiomyocytes following ischemic or hypoxic insults. The present study aimed to explore the protective effects and mechanism of exendin-4, a GLP-1 analogue, on cardiomyocyte glucose uptake using an in vitro model of hypoxia/reoxygenation (H/R) of H9c2 cardiomyocyte cells. Pre-treatment with exendin-4 (200 nM) prior to H/R increased the cell viability, decreased cell apoptosis, enhanced cardiomyocyte glucose uptake and increased the production of adenosine triphosphate. Exendin-4 also decreased the levels of lactate dehydrogenase and creatine kinase-MB in the culture medium. Furthermore, the activity of carnitine palmitoyltransferase-1 in the H9c2 cells was decreased, while the activity of phosphofructokinase-1 was increased following exendin-4 treatment. Moreover, pre-treatment with exendin-4 increased the expression of p38 mitogen-activated protein kinase (p38MAPK) γ and translocation of glucose transporter-1 in H9c2 cells subjected to H/R. However, these effects were attenuated by the p38MAPK inhibitors BIRB796 and SB203580. The results suggested that exendin-4 exerted significant cardioprotective effects against H/R-induced cell injury and restored the metabolic imbalance of cardiomyocytes by activating the p38MAPK signaling pathway in the H9c2 cell model. Importantly, p38MAPKγ, one subunit of p38MAPK, may have the most important function in this process. The results of the present study may be helpful in the development of novel drugs to treat patients with coronary heart disease.
Collapse
Affiliation(s)
- Kai Lu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Guanglei Chang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Lin Ye
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Peng Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yong Li
- Center for Perinatal Biology, Division of Pharmacology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Dongying Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
39
|
Eriksson L, Nyström T. Antidiabetic agents and endothelial dysfunction - beyond glucose control. Basic Clin Pharmacol Toxicol 2015; 117:15-25. [PMID: 25827165 DOI: 10.1111/bcpt.12402] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 03/24/2015] [Indexed: 12/15/2022]
Abstract
Diabetes is rapidly increasing worldwide, and the number of patients suffering from diabetes is projected to rise by 50% over the next 25 years, then affecting almost 600 million adults. Type 2 diabetes comprises 90-95% of all people with diabetes, and they constitute a patient group that carries a high burden of cardiovascular disease. The relationship between hyperglycaemia and macrovascular complications is still uncertain, at least in terms of the possibility of reducing cardiovascular events solely by improving glycaemic control. This MiniReview has thus focused on the effect of common antidiabetic agents, with emphasis on glucagon-like peptide-1, on the endothelial cells of the vasculature. Patients with type 2 diabetes suffer a two to four times higher risk of myocardial infarction and stroke than healthy persons. In addition to this, patients with diabetes have an increased atherosclerotic burden. Endothelial dysfunction is thought to be an early and important predictor of atherosclerosis and cardiovascular disease, and in people with type 2 diabetes, endothelial dysfunction is a common finding. It is therefore of importance to consider whether drugs used within the clinical management of Type 2 diabetes mellitus (T2DM) exert direct and positive effects on the vasculature independent of their glucose-lowering ability. This might serve to limit the adverse consequences of the macrovascular complications of T2DM, as dysfunction of endothelial cells is believed to contribute to a premature development of atherosclerosis.
Collapse
Affiliation(s)
- Linnéa Eriksson
- Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Centre for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Thomas Nyström
- Department of Clinical Science and Education, Karolinska Institutet, Stockholm South Hospital, Stockholm, Sweden
| |
Collapse
|
40
|
Balestrieri ML, Rizzo MR, Barbieri M, Paolisso P, D'Onofrio N, Giovane A, Siniscalchi M, Minicucci F, Sardu C, D'Andrea D, Mauro C, Ferraraccio F, Servillo L, Chirico F, Caiazzo P, Paolisso G, Marfella R. Sirtuin 6 expression and inflammatory activity in diabetic atherosclerotic plaques: effects of incretin treatment. Diabetes 2015; 64:1395-406. [PMID: 25325735 DOI: 10.2337/db14-1149] [Citation(s) in RCA: 148] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The role of sirtuin 6 (SIRT6) in atherosclerotic progression of diabetic patients is unknown. We evaluated SIRT6 expression and the effect of incretin-based therapies in carotid plaques of asymptomatic diabetic and nondiabetic patients. Plaques were obtained from 52 type 2 diabetic and 30 nondiabetic patients undergoing carotid endarterectomy. Twenty-two diabetic patients were treated with drugs that work on the incretin system, GLP-1 receptor agonists, and dipeptidyl peptidase-4 inhibitors for 26 ± 8 months before undergoing the endarterectomy. Compared with nondiabetic plaques, diabetic plaques had more inflammation and oxidative stress, along with a lesser SIRT6 expression and collagen content. Compared with non-GLP-1 therapy-treated plaques, GLP-1 therapy-treated plaques presented greater SIRT6 expression and collagen content, and less inflammation and oxidative stress, indicating a more stable plaque phenotype. These results were supported by in vitro observations on endothelial progenitor cells (EPCs) and endothelial cells (ECs). Indeed, both EPCs and ECs treated with high glucose (25 mmol/L) in the presence of GLP-1 (100 nmol/L liraglutide) presented a greater SIRT6 and lower nuclear factor-κB expression compared with cells treated only with high glucose. These findings establish the involvement of SIRT6 in the inflammatory pathways of diabetic atherosclerotic lesions and suggest its possible positive modulation by incretin, the effect of which is associated with morphological and compositional characteristics of a potential stable plaque phenotype.
Collapse
Affiliation(s)
- Maria Luisa Balestrieri
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Maria Rosaria Rizzo
- Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Second University of Naples, Naples, Italy
| | - Michelangela Barbieri
- Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Second University of Naples, Naples, Italy
| | - Pasquale Paolisso
- Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Second University of Naples, Naples, Italy
| | - Nunzia D'Onofrio
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Alfonso Giovane
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | | | - Fabio Minicucci
- Department of Cardiology, Cardarelli Hospital, Naples, Italy
| | - Celestino Sardu
- Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Second University of Naples, Naples, Italy
| | - Davide D'Andrea
- Department of Cardiology, Cardarelli Hospital, Naples, Italy
| | - Ciro Mauro
- Department of Cardiology, Cardarelli Hospital, Naples, Italy
| | - Franca Ferraraccio
- Department of Clinical, Public and Preventive Medicine, Second University of Naples, Naples, Italy
| | - Luigi Servillo
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Fabio Chirico
- Department of Neurosurgery, Cardarelli Hospital, Naples, Italy
| | | | - Giuseppe Paolisso
- Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Second University of Naples, Naples, Italy
| | - Raffaele Marfella
- Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Second University of Naples, Naples, Italy
| |
Collapse
|
41
|
Salheen SM, Panchapakesan U, Pollock CA, Woodman OL. The DPP-4 inhibitor linagliptin and the GLP-1 receptor agonist exendin-4 improve endothelium-dependent relaxation of rat mesenteric arteries in the presence of high glucose. Pharmacol Res 2015; 94:26-33. [PMID: 25697548 DOI: 10.1016/j.phrs.2015.02.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 02/05/2015] [Accepted: 02/06/2015] [Indexed: 10/24/2022]
Abstract
The aim of the study was to investigate the effects of the DPP-4 inhibitors and GLP-1R agonist, exendin-4 on the mechanism(s) of endothelium-dependent relaxation in rat mesenteric arteries exposed to high glucose concentration (40 mM). Organ bath techniques were employed to investigate vascular endothelial function in rat mesenteric arteries in the presence of normal (11 mM) or high (40 mM) glucose concentrations. Pharmacological tools (1μM TRAM-34, 1μM apamin, 100 nM Ibtx, 100 μM l-NNA, 10 μM ODQ) were used to distinguish between NO and EDHF-mediated relaxation. Superoxide anion levels were assessed by L-012 and lucigenin enhanced-chemiluminescence techniques. Incubation of mesenteric rings with high glucose for 2 h caused a significant increase in superoxide anion generation and a significant impairment of endothelium-dependent relaxation. Exendin-4 and DPP-4 inhibitor linagliptin, but not sitagliptin or vildagliptin, significantly reduced vascular superoxide and improved endothelium-dependent relaxation in the presence of high glucose. The beneficial actions of exendin-4, but not linagliptin, were attenuated by the GLP-1R antagonist exendin fragment (9-39). Further experiments demonstrated that the presence of high glucose impaired the contribution of both nitric oxide and endothelium-dependent hyperpolarisation to relaxation and that linagliptin improved both mechanisms involved in endothelium-dependent relaxation. These findings demonstrate that high glucose impaired endothelium-dependent relaxation can be improved by exendin-4 and linagliptin, likely due to their antioxidant activity and independently of any glucose lowering effect.
Collapse
Affiliation(s)
- S M Salheen
- School of Medical Sciences, RMIT University, Melbourne, Australia
| | - U Panchapakesan
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, New South Wales, Australia
| | - C A Pollock
- Kolling Institute of Medical Research, Royal North Shore Hospital, University of Sydney, New South Wales, Australia
| | - O L Woodman
- School of Medical Sciences, RMIT University, Melbourne, Australia.
| |
Collapse
|
42
|
Osto E, Doytcheva P, Corteville C, Bueter M, Dörig C, Stivala S, Buhmann H, Colin S, Rohrer L, Hasballa R, Tailleux A, Wolfrum C, Tona F, Manz J, Vetter D, Spliethoff K, Vanhoutte PM, Landmesser U, Pattou F, Staels B, Matter CM, Lutz TA, Lüscher TF. Rapid and body weight-independent improvement of endothelial and high-density lipoprotein function after Roux-en-Y gastric bypass: role of glucagon-like peptide-1. Circulation 2015; 131:871-81. [PMID: 25673670 DOI: 10.1161/circulationaha.114.011791] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Roux-en-Y gastric bypass (RYGB) reduces body weight and cardiovascular mortality in morbidly obese patients. Glucagon-like peptide-1 (GLP-1) seems to mediate the metabolic benefits of RYGB partly in a weight loss-independent manner. The present study investigated in rats and patients whether obesity-induced endothelial and high-density lipoprotein (HDL) dysfunction is rapidly improved after RYGB via a GLP-1-dependent mechanism. METHODS AND RESULTS Eight days after RYGB in diet-induced obese rats, higher plasma levels of bile acids and GLP-1 were associated with improved endothelium-dependent relaxation compared with sham-operated controls fed ad libitum and sham-operated rats that were weight matched to those undergoing RYGB. Compared with the sham-operated rats, RYGB improved nitric oxide (NO) bioavailability resulting from higher endothelial Akt/NO synthase activation, reduced c-Jun amino terminal kinase phosphorylation, and decreased oxidative stress. The protective effects of RYGB were prevented by the GLP-1 receptor antagonist exendin9-39 (10 μg·kg(-1)·h(-1)). Furthermore, in patients and rats, RYGB rapidly reversed HDL dysfunction and restored the endothelium-protective properties of the lipoprotein, including endothelial NO synthase activation, NO production, and anti-inflammatory, antiapoptotic, and antioxidant effects. Finally, RYGB restored HDL-mediated cholesterol efflux capacity. To demonstrate the role of increased GLP-1 signaling, sham-operated control rats were treated for 8 days with the GLP-1 analog liraglutide (0.2 mg/kg twice daily), which restored NO bioavailability and improved endothelium-dependent relaxations and HDL endothelium-protective properties, mimicking the effects of RYGB. CONCLUSIONS RYGB rapidly reverses obesity-induced endothelial dysfunction and restores the endothelium-protective properties of HDL via a GLP-1-mediated mechanism. The present translational findings in rats and patients unmask novel, weight-independent mechanisms of cardiovascular protection in morbid obesity.
Collapse
Affiliation(s)
- Elena Osto
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.).
| | - Petia Doytcheva
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Caroline Corteville
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Marco Bueter
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Claudia Dörig
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Simona Stivala
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Helena Buhmann
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Sophie Colin
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Lucia Rohrer
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Reda Hasballa
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Anne Tailleux
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Christian Wolfrum
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Francesco Tona
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Jasmin Manz
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Diana Vetter
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Kerstin Spliethoff
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Paul M Vanhoutte
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Ulf Landmesser
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Francois Pattou
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Bart Staels
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Christian M Matter
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Thomas A Lutz
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| | - Thomas F Lüscher
- From Centre for Molecular Cardiology, University of Zurich and University Heart Center, Cardiology, University Hospital Zurich, Switzerland (E.O., P.D., S.S., J.M., U.L., C.M.M., T.F.L.); Institute of Veterinary Physiology, University of Zurich, Switzerland (P.D., C.C., C.D., H.B., K.S., T.A.L.); Department of Surgery (M.B., D.V.) and Institute of Clinical Chemistry (L.R., R.H.), University Hospital Zurich, Switzerland; Université Lille 2, INSERM UMR1011, EGID, Institut Pasteur de Lille, France (S.C., A.T., B.S.); Department of Health Sciences and Technology, ETH Zurich, Switzerland (C.W.); Department of Cardiac, Thoracic and Vascular Sciences, University of Padua, Italy (F.T.); State Key Laboratory for Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, University of Hong Kong, SAR (P.M.V.); and Department of Endocrine Surgery, Lille University Hospital, France (F.P.)
| |
Collapse
|
43
|
Eriksson L, Nyström T. Activation of AMP-activated protein kinase by metformin protects human coronary artery endothelial cells against diabetic lipoapoptosis. Cardiovasc Diabetol 2014; 13:152. [PMID: 25391818 PMCID: PMC4234893 DOI: 10.1186/s12933-014-0152-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 10/28/2014] [Indexed: 12/02/2022] Open
Abstract
Background The prevalence of type 2 diabetes (T2D) among adults worldwide is rapidly increasing, and in patients with diabetes the major cause of death is macrovascular disease. Endothelial cells play an important role in maintaining vascular homeostasis. Free fatty acids, which are elevated in T2D, have previously been shown to induce endothelial dysfunction and apoptosis of endothelial cells, which is considered as an important and early factor in the onset of atherosclerosis and cardiovascular disease. Metformin, which is used as first line treatment of T2D patients, is believed to exert its pharmacological effects through activation of AMP-activated protein kinase, which has emerged as a new potential target in reversing endothelial dysfunction. Methods Here we studied the protective effect of metformin against free fatty acid-induced apoptosis of human coronary artery endothelial cells (HCAECs) by assessing DNA fragmentation and cleaved caspase 3 levels. We also attempted to elucidate the underlying mechanisms by investigating the involvement of AMP-activated protein kinase, p38 MAPK and eNOS. Generation of reactive oxygen species by free fatty acid exposure was also examined. Results Our results suggest that metformin protects HCAECs from lipoapoptosis, an effect that involves eNOS and p38 MAPK, downstream of AMPK signaling, but not as previously suggested through suppression of reactive oxygen species. Conclusion The protective effect of metformin against free fatty acid induced apoptosis is potentially clinically relevant as metformin is first line treatment for patients with T2D, a patient group which is rapidly increasing and carries a high burden of cardiovascular disease.
Collapse
Affiliation(s)
- Linnéa Eriksson
- Department of Clinical Science and Education, Section of Endocrinology and Diabetology, Karolinska Institutet, Södersjukhuset AB, Stockholm, Sweden. .,Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, Center for Molecular Medicine, Stockholm, Sweden.
| | - Thomas Nyström
- Department of Clinical Science and Education, Section of Endocrinology and Diabetology, Karolinska Institutet, Södersjukhuset AB, Stockholm, Sweden.
| |
Collapse
|
44
|
DeNicola M, Du J, Wang Z, Yano N, Zhang L, Wang Y, Qin G, Zhuang S, Zhao TC. Stimulation of glucagon-like peptide-1 receptor through exendin-4 preserves myocardial performance and prevents cardiac remodeling in infarcted myocardium. Am J Physiol Endocrinol Metab 2014; 307:E630-43. [PMID: 25117407 PMCID: PMC4200306 DOI: 10.1152/ajpendo.00109.2014] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 08/04/2014] [Indexed: 12/21/2022]
Abstract
We have demonstrated that GLP-1 improved myocardial functional recovery in acute myocardial ischemic injury. However, whether stimulation of the GLP-1 receptor (GLP-1R) with exendin-4, a selective GLP-1R agonist, could initiate a protective effect in the heart remains to be determined. Mouse myocardial infarction (MI) was created by ligation of the left descending artery. After 48 h of MI, animals were divided into the following groups (n = 5-7/group): 1) sham (animals that underwent thoracotomy without ligation), 2) MI [animals that underwent MI and received a daily dose of intraperitoneal injection (ip) of saline]; and 3) MI + exendin-4 [infarcted mice that received injections of exendin-4 (0.1 mg/kg ip)]. Two weeks later, cardiac function was assessed by echocardiography and an isovolumetrically perfused heart. Compared with control MI hearts, stimulation of GLP-1R improved cardiac function, which was associated with attenuation of myocardial hypertrophy, the mitigation of interstitial fibrosis, and an increase in survival rate in post-MI hearts. Furthermore, H9c2 cardiomyoblasts were preconditioned with exendin-4 at a dose of 100 nmol/l and then subjected to hydrogen peroxide exposure at concentrations of 50 and 100 μmol/l. The exendin-4 treatment decreased lactate dehydrogenase leakage and increased cell survival. Notably, this event was also associated with the reduction of cleaved caspase-3 and caspase-9 and attenuation of reactive oxygen species production. Exendin-4 treatments improved mitochondrial respiration and suppressed the opening of mitochondrial permeability transition pore and protected mitochondria function. Our results indicate that GLP-1R serves as a novel approach to eliciting cardioprotection and mitigating oxidative stress-induced injury.
Collapse
MESH Headings
- Animals
- Antioxidants/pharmacology
- Antioxidants/therapeutic use
- Apoptosis/drug effects
- Cardiotonic Agents/pharmacology
- Cardiotonic Agents/therapeutic use
- Cell Line
- Cell Survival/drug effects
- Disease Models, Animal
- Exenatide
- Glucagon-Like Peptide-1 Receptor
- Heart Ventricles/diagnostic imaging
- Heart Ventricles/drug effects
- Heart Ventricles/physiopathology
- Hypertrophy, Left Ventricular/diagnostic imaging
- Hypertrophy, Left Ventricular/etiology
- Hypertrophy, Left Ventricular/prevention & control
- Kaplan-Meier Estimate
- Male
- Mice, Inbred ICR
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Molecular Targeted Therapy
- Myoblasts, Cardiac/drug effects
- Myoblasts, Cardiac/metabolism
- Myocardial Infarction/drug therapy
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Oxidative Stress/drug effects
- Peptides/pharmacology
- Peptides/therapeutic use
- Rats
- Receptors, Glucagon/agonists
- Receptors, Glucagon/metabolism
- Ultrasonography
- Venoms/pharmacology
- Venoms/therapeutic use
- Ventricular Dysfunction, Left/diagnostic imaging
- Ventricular Dysfunction, Left/etiology
- Ventricular Dysfunction, Left/prevention & control
- Ventricular Remodeling/drug effects
Collapse
Affiliation(s)
- Megan DeNicola
- Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, Rhode Island
| | - Jianfeng Du
- Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, Rhode Island
| | - Zhengke Wang
- Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, Rhode Island
| | - Naohiro Yano
- Department of Obstetrics and Gynecology, Women and Infants' Hospital of Rhode Island, Providence, Rhode Island
| | | | - Yigang Wang
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio; and
| | - Gangjian Qin
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Shougang Zhuang
- Department of Medicine, Rhode Island Hospital, Alpert Medical School, Brown University, Providence, Rhode Island
| | - Ting C Zhao
- Department of Surgery, Roger Williams Medical Center, Boston University Medical School, Boston University, Providence, Rhode Island;
| |
Collapse
|
45
|
Darsalia V, Nathanson D, Nyström T, Klein T, Sjöholm Å, Patrone C. GLP-1R activation for the treatment of stroke: updating and future perspectives. Rev Endocr Metab Disord 2014; 15:233-42. [PMID: 24777909 DOI: 10.1007/s11154-014-9285-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Stroke is the leading cause of adult disability in Westernized societies with increased incidence along ageing and it represents a major health and economical threat. Inactive lifestyle, smoking, hypertension, atherosclerosis, obesity and diabetes all dramatically increase the risk of stroke. While preventive strategies based on lifestyle changes and risk factor management can delay or decrease the likelihood of having a stroke, post stroke pharmacological strategies aimed at minimizing stroke-induced brain damage are highly needed. Unfortunately, several candidate drugs that have shown significant preclinical neuroprotective efficacy, have failed in clinical trials and no treatment for stroke based on neuroprotection is available today. Glucagon-like peptide 1 (GLP-1) is a peptide originating in the enteroendocrine L-cells of the intestine and secreted upon nutrient ingestion. The activation of the GLP-1R by GLP-1 enhances glucose-dependent insulin secretion, suppresses glucagon secretion and exerts multifarious extrapancreatic effects. Stable GLP-1 analogues and inhibitors of the proteolytic enzyme dipeptidyl peptidase 4 (DPP-4) (which counteract endogenous GLP-1 degradation) have been developed clinically for the treatment of type 2 diabetes. Besides their antidiabetic properties, experimental evidence has shown neurotrophic and neuroprotective effects of GLP-1R agonists and DPP-4 inhibitors in animal models of neurological disorders. Herein, we review recent experimental data on the neuroprotective effects mediated by GLP-1R activation in stroke. Due to the good safety profile of the drugs targeting the GLP-1R, we also discuss the high potential of GLP-1R stimulation in view of developing a safe clinical treatment against stroke based on neuroprotection in both diabetic and non-diabetic patients.
Collapse
Affiliation(s)
- Vladimer Darsalia
- Department of Clinical Science and Education, Södersjukhuset, Internal Medicine, Karolinska Institutet, SE-118 83, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
46
|
Du X, Hu X, Wei J. Anti-inflammatory effect of exendin-4 postconditioning during myocardial ischemia and reperfusion. Mol Biol Rep 2014; 41:3853-7. [PMID: 24549718 DOI: 10.1007/s11033-014-3252-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Accepted: 02/08/2014] [Indexed: 12/30/2022]
Abstract
High mobility group box 1 protein (HMGB1) plays an important role in myocardial ischemia and reperfusion (I/R) injury. Preconditioning of exendin-4 (Ex), a glucagon-like peptide-1 receptor agonist, has been reported to attenuate myocardial I/R injury. The current study investigated whether Ex postconditioning also attenuated myocardial I/R injury and the potential mechanisms. Anesthetized male rats were subjected to ischemia for 30 min and treated with Ex (5 μg/kg, i.v.) 5 min before reperfusion, in the absence and/or presence of exendin (9-39) (an antagonist of glucagon-like peptide-1 receptor, 5 μg/kg, i.v.), followed by reperfusion for 4 h. Lactate dehydrogenase (LDH), creatine kinase (CK), tumor necrosis factor-α, interleukin-6, and infarct size were measured. HMGB1 expression was assessed by immunoblotting. Postconditioning with Ex significantly decreased infarct size and levels of LDH and CK after 4 h reperfusion (all p < 0.05). Ex also significantly inhibited the increase in malondialdehyde level and decreased the level of superoxide dismutase (both p < 0.05). In addition, the increase in HMGB1 expression induced by I/R was significantly attenuated by Ex postconditioning. Administration of exendin (9-39) abolished the protective effect of Ex postconditioning (all p < 0.05). The present study suggests that Ex postconditioning may attenuate myocardial I/R injury, which may in turn be associated with inhibiting inflammation.
Collapse
Affiliation(s)
- Xianjin Du
- Department of Emergency, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuchang, Wuhan, 430060, China
| | | | | |
Collapse
|
47
|
Zeng Y, Li C, Guan M, Zheng Z, Li J, Xu W, Wang L, He F, Xue Y. The DPP-4 inhibitor sitagliptin attenuates the progress of atherosclerosis in apolipoprotein-E-knockout mice via AMPK- and MAPK-dependent mechanisms. Cardiovasc Diabetol 2014; 13:32. [PMID: 24490809 PMCID: PMC3916068 DOI: 10.1186/1475-2840-13-32] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 01/30/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The dipeptidyl peptidase-4 inhibitor sitagliptin, a new anti-diabetic medicine, is effective in treating type 2 diabetes mellitus by increasing the activation and duration of action of glucagon-like peptide-1. Since atherosclerosis is the main pathological feature of diabetic cardiovascular complications, it is important to investigate the anti-atherosclerotic effect of sitagliptin and explore the relevant mechanisms. METHODS Male apolipoprotein-E-knockout mice were randomly divided into two groups and fed either high-fat diet (HFD) or HFD plus sitagliptin at a concentration of 0.3% for 16 weeks. Body weight, food intake, blood glucose, serum lipids and adhesion molecules were measured. The atherosclerotic plaque area and its histological composition were analyzed using Sudan staining and immunohistochemistry. The expression of inflammatory cytokines (monocyte chemoattractant protein (MCP)-1 and interleukin (IL)-6) and the activation of AMP-activated protein kinase (AMPK) and mitogen-activated protein kinase (MAPK) in the aortas were determined using quantitative polymerase chain reaction and western blot, respectively. RESULTS Mice treated with sitagliptin developed fewer atherosclerotic plaques than the control group (7.64 ± 1.98% vs 12.91 ± 1.15%, p < 0.001), particularly in the aortic arch and abdominal aorta, where plaques were decreased 1.92- and 2.74-fold, respectively (p < 0.05 and p < 0.01). Sitagliptin significantly reduced the content of collagen fiber in plaques 1.2-fold (p < 0.05). Moreover, sitagliptin significantly reduced the expression of monocyte chemoattractant protein-1 and interleukin-6 in the aorta (p < 0.01 and p < 0.05), as well as the serum levels of soluble vascular cell adhesion molecule-1 and P-selectin (both p < 0.05). In addition, Sitagliptin induced phosphorylation of AMPK and Akt (p < 0.05 and p < 0.01), while suppressed phosphorylation of p38 and extracellular signal-regulated kinase (Erk) 1/2 (p < 0.05 and p < 0.01) in aortas. CONCLUSIONS Our present study indicates that sitagliptin can reduce the area of the atherosclerotic lesion, possibly by regulating the AMPK and MAPK pathways and then reducing leukocyte -endothelial cell interaction and inflammation reactions. These actions are independent of weight loss and glucose-reducing effects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yaoming Xue
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510150, China.
| |
Collapse
|
48
|
He X, Song W, Liu C, Chen S, Hua J. Rapamycin inhibits acrolein-induced apoptosis by alleviating ROS-driven mitochondrial dysfunction in male germ cells. Cell Prolif 2014; 47:161-71. [PMID: 24483236 DOI: 10.1111/cpr.12091] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 10/20/2013] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Acrolein (Acr) is a highly reactive α, β-unsaturated aldehyde, which can induce reactive oxygen species (ROS) generation. Several factors, including lipid peroxidation, clinical use of cyclophosphamide, fried foods, automobile exhausts, smoking and aging can increase its concentration in blood serum. Mounting evidence has suggested that Acr-induced ROS might reduce quality of sperm. Thus, the aim of this study was to examine reproductive toxicity of Acr-caused ROS in vitro and find a means to alleviate it. MATERIALS AND METHODS We investigated the effects of Acr on male germ cell (MGC)-derived GC-1 cells in vitro. Dihydroethidium and DCFH-DA fluorescent dyes were used to determine generation of intracellular ROS. RESULTS We found that Acr induced ROS generation, which was accompanied by reduced Bcl2/Bax ratio, substantial decline in mitochondrial membrane potential, and further promoted apoptosis of MGCs. Furthermore, Rapamycin was capable of alleviating Acr-induced ROS, reducing ROS-induced apoptosis by increasing ratio of Bcl2/Bax mRNA and proteins, and protecting MGC mitochondrial membranes. CONCLUSION Rapamycin inhibited Acr-induced apoptosis by alleviating ROS-driven mitochondrial dysfunction in MGCs.
Collapse
Affiliation(s)
- X He
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering & Technology, Key Lab for Animal Biotechnology of Agriculture Ministry of China, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | | | | | | | | |
Collapse
|
49
|
Iwai T, Sawabe T, Tanimitsu K, Suzuki M, Sasaki-Hamada S, Oka JI. Glucagon-like peptide-1 protects synaptic and learning functions from neuroinflammation in rodents. J Neurosci Res 2014; 92:446-54. [PMID: 24464856 DOI: 10.1002/jnr.23335] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Revised: 10/18/2013] [Accepted: 10/29/2013] [Indexed: 12/24/2022]
Abstract
Glucagon-like peptide-1 (GLP-1) is derived from the processing of proglucagon in intestinal L-cells and releases insulin from pancreatic β-cells as an incretin. The GLP-1 receptor has been proposed as a possible therapeutic target for the treatment of Alzheimer's disease, in which neuroinflammation is critical in the pathogenesis. The present study investigates whether GLP-1 (7-36) amide, an active fragment of GLP-1, protected against synaptic impairments induced by inflammation-related injurious agents (lipopolysaccharide [LPS], interleukin-1β [IL-1β], and H2 O2). In the Y-maze test, LPS (10 μg/mouse, i.c.v) significantly decreased the percentage alternation. Pretreatment with GLP-1 (7-36) amide (0.09-0.9 nmol/mouse, i.c.v.) prevented an impairment in spontaneous alternation performance. Pretreatment with LPS (10 μg/ml, 2 hr) impaired LTP induction but not paired-pulse facilitation in the CA1 region of rat hippocampal slices. This impairment was prevented by cotreatment with GLP-1 (7-36) amide (50 nM). IL-1β (0.57 nM) or H2 O2 (50 μM) also impaired LTP induction. This impairment was prevented by GLP-1 (7-36) amide (50 nM). These results suggest that GLP-1 (7-36) amide improves the synaptic impairments induced by inflammation-related injurious agents in the CA1 region of the hippocampus.
Collapse
Affiliation(s)
- Takashi Iwai
- Laboratory of Pharmacology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | | | | | | | | | | |
Collapse
|
50
|
Oyama JI, Higashi Y, Node K. Do incretins improve endothelial function? Cardiovasc Diabetol 2014; 13:21. [PMID: 24428883 PMCID: PMC3898564 DOI: 10.1186/1475-2840-13-21] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Accepted: 01/10/2014] [Indexed: 12/11/2022] Open
Abstract
An impaired endothelial function has been recognized in the early stage of atherosclerosis, and is a major factor affecting the future development of cardiovascular events. Type 2 diabetes mellitus (T2DM) is widely prevalent, and is one of the most important risk factors for cardiovascular disease. T2DM is associated with increases in both morbidity and mortality, particularly from cardiovascular disease.New therapies based on the incretin hormone and its actions are now becoming widely used, and appear to offer advantages over conventional therapies by keeping the body weight steady and limiting hypoglycemia, while also achieving attractive glycemic control. However, there is little data available about the effects of incretins on the cardiovascular system.This review will focus on the effects of incretin therapies, including glucagon-like peptide-1 (GLP-1) analogs and dipeptidyl peptidase (DPP)-4 inhibitors, on the endothelial function, and will discuss the potential mechanisms underlying these effects.
Collapse
Affiliation(s)
- Jun-Ichi Oyama
- Department of Cardiovascular Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan.
| | | | | |
Collapse
|