1
|
Lu L, Li J, Jiang X, Bai R. CXCR4/CXCL12 axis: "old" pathway as "novel" target for anti-inflammatory drug discovery. Med Res Rev 2024; 44:1189-1220. [PMID: 38178560 DOI: 10.1002/med.22011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/25/2023] [Accepted: 12/16/2023] [Indexed: 01/06/2024]
Abstract
Inflammation is the body's defense response to exogenous or endogenous stimuli, involving complex regulatory mechanisms. Discovering anti-inflammatory drugs with both effectiveness and long-term use safety is still the direction of researchers' efforts. The inflammatory pathway was initially identified to be involved in tumor metastasis and HIV infection. However, research in recent years has proved that the CXC chemokine receptor type 4 (CXCR4)/CXC motif chemokine ligand 12 (CXCL12) axis plays a critical role in the upstream of the inflammatory pathway due to its chemotaxis to inflammatory cells. Blocking the chemotaxis of inflammatory cells by CXCL12 at the inflammatory site may block and alleviate the inflammatory response. Therefore, developing CXCR4 antagonists has become a novel strategy for anti-inflammatory therapy. This review aimed to systematically summarize and analyze the mechanisms of action of the CXCR4/CXCL12 axis in more than 20 inflammatory diseases, highlighting its crucial role in inflammation. Additionally, the anti-inflammatory activities of CXCR4 antagonists were discussed. The findings might help generate new perspectives for developing anti-inflammatory drugs targeting the CXCR4/CXCL12 axis.
Collapse
Affiliation(s)
- Liuxin Lu
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Junjie Li
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Xiaoying Jiang
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Renren Bai
- Department of Medicinal Chemistry, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, China
- Key Laboratory of Elemene Class Anti-tumor Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Bioactive Compounds (BACs): A Novel Approach to Treat and Prevent Cardiovascular Diseases. Curr Probl Cardiol 2023; 48:101664. [PMID: 36841315 DOI: 10.1016/j.cpcardiol.2023.101664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 02/17/2023] [Indexed: 02/26/2023]
Abstract
Cardiovascular diseases (CVDs) are one of the leading disorders of serious death and cause huge economic loss to patients and society. It is estimated that about 18 million people have a high death ratio due to the incidence of CVDs such as (stroke, coronary heart disease, and non-ischemic heart failure). Bioactive compounds (BACs) are healthy nutritional ingredients providing beneficial effects and nutritional value to the human body. Epidemiological studies strongly shed light on several bioactive compounds that are favorable candidates for CVDs treatment. Globally, the high risk of CVDs and related results on human body parts made them a serious scenario in all communities. In this present review, we intend to collect previously published data concerned over the years concerning green-colored foods and their BACs that aim to work in the prevention, diagnosis, and/or systematic treating CVDs. We also comprehensively discussed the oral delivery of several bioactive compounds derived from fruits and vegetables and their bioavailability and physiological effects on human health. Moreover, their important characteristics, such as anti-inflammatory, lowering blood pressure, anti-obesity, antioxidant, anti-diabetics, lipid-lowering responses, improving atherosclerosis, and cardioprotective properties, will be elaborated further. More precisely, medicinal plants' advantages and multifaceted applications have been reported in this literature to treat CVDs. To the best of our knowledge, this is our first attempt that will open a new window in the area of CVDs with the opportunity to achieve a better prognosis and effective treatment for CVDs.
Collapse
|
3
|
Lee KG, Santos ARMP, Kang YG, Chae YJ, Shah M, Pirzada RH, Song M, Kim J, Choi S, Park Y. Efficacy Evaluation of SDF-1α-Based Polypeptides in an Acute Myocardial Infarction Model Using Structure-Based Drug Design. ACS Biomater Sci Eng 2022; 8:4486-4496. [PMID: 36178141 DOI: 10.1021/acsbiomaterials.2c00766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Stromal cell-derived factor-1 alpha (SDF-1α, CXCL12) mediates the migration of circulating cells to desired sites for tissue development, homeostasis, and regeneration and can be used to promote cardiac regeneration by recruiting stem cells. However, the use of SDF-1α in the injured heart necessitates not only higher binding affinity to its receptor, CXCR4+, but also better robustness against enzymatic degradation than other SDF-1 isoforms. Here, we conduct a screening of SDF-1α analog peptides that were designed by structure-based drug design (SBDD), a type of computer-aided drug design (CADD). We have developed in vitro and in vivo methods that enable us to estimate the effect of peptides on the migration of human mesenchymal stem cells (hMSCs) and cardiac regeneration in acute myocardial infarction (AMI)-induced animals, respectively. We demonstrate that one type of SDF-1α analog peptide, SDP-4, among the four analog peptides preselected by SBDD, is more potent than native SDF-1α for cardiac regeneration in myocardial infarction. It is interesting to note that the migratory effects of SDP-4 determined by a wound healing assay, a Transwell assay, and a 2D migration assay are comparable to those of SDF-1α. These results suggest that in vivo, as well as in vitro, screening of peptides developed by SBDD is a quintessential process to the development of a novel therapeutic compound for cardiac regeneration. Our finding also has an implication that the SDP-4 peptide is an excellent candidate for use in the regeneration of an AMI heart.
Collapse
Affiliation(s)
- Kang-Gon Lee
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Ana Rita M P Santos
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Yong Guk Kang
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Yun Jin Chae
- R&D center, Scholar Foxtrot Co. Ltd., Seoul 02796, Korea
| | - Masaud Shah
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| | | | - Myeongjin Song
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| | - Jongseong Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea.,R&D center, Scholar Foxtrot Co. Ltd., Seoul 02796, Korea
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea
| | - Yongdoo Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Seoul 02841, Korea
| |
Collapse
|
4
|
Oprescu N, Micheu MM, Scafa-Udriste A, Popa-Fotea NM, Dorobantu M. Inflammatory markers in acute myocardial infarction and the correlation with the severity of coronary heart disease. Ann Med 2021; 53:1041-1047. [PMID: 34180324 PMCID: PMC8245096 DOI: 10.1080/07853890.2021.1916070] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 04/07/2021] [Indexed: 11/15/2022] Open
Abstract
INTRODUCTION The inflammatory hypothesis of atherosclerosis is appealing in acute coronary syndromes, but the dynamics and precise role are not established. OBJECTIVES The study investigates the levels of C reactive protein (CRP), interleukin 1β (IL-1β) and stromal-derived factor 1α (SDF-1α) at the time of acute myocardial infarction (AMI) and at 1 and 6 months afterwards, compared with a control group. RESULTS In the acute phase of AMI, CRP and SDF-1α were significantly higher, while IL-1β showed lower levels compared with controls. CRP positively correlated with coronary stenosis severity (rho = 0.3, p=.05) and negatively related with left ventricle ejection fraction (LVEF) at 1 month (rho= -0.43, p=.05). IL-1β weakly correlated with the severity of coronary lesions (rho =0.29, p=.02) and strongly with LVEF (rho= -0.8, p=.05). SDF-1α, slightly correlated with LVEF at 1 month (rho = 0.22, p=.01) and with the severity of coronary atherosclerosis (rho= -0.41, p=.003). CONCLUSIONS CRP, IL-1β and SDF-1α have important dynamic in the first 6 months after AMI and CRP and SDF-1α levels correlated with the severity of coronary lesions and LVEF at 1 month after the acute ischaemic event.
Collapse
Affiliation(s)
- Nicoleta Oprescu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Romania
| | | | - Alexandru Scafa-Udriste
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Romania
- Cardio-thoracic Department, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Nicoleta-Monica Popa-Fotea
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Romania
- Cardio-thoracic Department, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| | - Maria Dorobantu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, Romania
- Cardio-thoracic Department, University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania
| |
Collapse
|
5
|
Sedighi M, Ghorbanzadeh V, Abaszadeh S, Karimi A, Cheraghi M, Rafieian-Kopaei M, Moghimian M, Mohammadi A, Veiskarami S, Mokhayeri Y, Nazari A. Up-regulation of chemokine receptor type 4 expression in the ischemic reperfused heart by alcoholic extract of Cichorium intybus rescue the heart from ischemia injury in male rat. J Pharm Pharmacol 2021; 73:1351-1360. [PMID: 34076244 DOI: 10.1093/jpp/rgab076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 05/11/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Cichorium intybus is used in traditional medicine for various diseases including heart disease. This study aimed at evaluating the chemokine receptor type 4 up-regulation and cardioprotective effects of hydroalcoholic extract of C. intybus in a rat model of ischemic reperfusion. METHODS Animals in four groups of eight rats each received vehicle or one of three doses of C. intybus (50, 100 or 200 mg/kg/d) for 14 days. Then they were subjected to 30 min of ischemia followed by 7 days of reperfusion. At the end of the experiment, blood specimens were prepared for serum assays. The level of myocardium chemokine receptor type 4 was also measured using RT-PCR. KEY FINDINGS Cichorium intybus (CI-50) improved infarct size, episodes of the ventricular ectopic beat, ventricular tachycardia, and duration of ventricular tachycardia, QTc shortening. It also stabilized the ST segment changes and increased heart rate during ischemia. The blood pressure decreased in CI-50 group in comparison to the control and CI-200 group. C. intybus increased serum superoxide dismutase and reduced lactate dehydrogenase activity, Cardiac Troponin I and malondialdehyde levels. C. intybus led to an increase in the expression of chemokine receptor type 4. CONCLUSIONS These findings suggest that C. intybus administration before ischemia is able to induce cardioprotective effect against ischemic reperfusion injury, probably through chemokine receptor type 4 over-expression and antioxidant activity.
Collapse
Affiliation(s)
- Mehrnoosh Sedighi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Vajihe Ghorbanzadeh
- Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Saber Abaszadeh
- Department of clinical biochemistry, Lorestan University of Medical Science, Khorramabad, Iran
| | - Arash Karimi
- Department of Anesthesiology, Anesthesiologist, Faculty of Medicine, Lorestan University of Medical Science, Khorramabad, Iran
| | - Mostafa Cheraghi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Mahmoud Rafieian-Kopaei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Siences, Shahrekord, Iran
| | - Maryam Moghimian
- Department of Physiology, Gonabad University of Medical Science, Gonabad, Iran
| | - Asghar Mohammadi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Saeid Veiskarami
- Lorestan Agricultural and Natural Resources Research and Education Center, Department of animal science, Iran
| | - Yaser Mokhayeri
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | - Afshin Nazari
- Department of Physiology, Lorestan University of Medical Science, Khorramabad, Iran
| |
Collapse
|
6
|
Wu X, Iroegbu CD, Peng J, Guo J, Yang J, Fan C. Cell Death and Exosomes Regulation After Myocardial Infarction and Ischemia-Reperfusion. Front Cell Dev Biol 2021; 9:673677. [PMID: 34179002 PMCID: PMC8220218 DOI: 10.3389/fcell.2021.673677] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 05/18/2021] [Indexed: 12/21/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death in the global population, accounting for about one-third of all deaths each year. Notably, with CVDs, myocardial damages result from myocardial infarction (MI) or cardiac arrhythmias caused by interrupted blood flow. Significantly, in the process of MI or myocardial ischemic-reperfusion (I/R) injury, both regulated and non-regulated cell death methods are involved. The critical factor for patients’ prognosis is the infarct area’s size, which determines the myocardial cells’ survival. Cell therapy for MI has been a research hotspot in recent years; however, exosomes secreted by cells have attracted much attention following shortcomings concerning immunogens. Exosomes are extracellular vesicles containing several biologically active substances such as lipids, nucleic acids, and proteins. New evidence suggests that exosomes play a crucial role in regulating cell death after MI as exosomes of various stem cells can participate in the cell damage process after MI. Hence, in the review herein, we focused on introducing various cell-derived exosomes to reduce cell death after MI by regulating the cell death pathway to understand myocardial repair mechanisms better and provide a reference for clinical treatment.
Collapse
Affiliation(s)
- Xun Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chukwuemeka Daniel Iroegbu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jun Peng
- Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha, China
| | - Jianjun Guo
- Hunan Fangsheng Pharmaceutical Co., Ltd., Changsha, China
| | - Jinfu Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Key Laboratory of Cardiovascular Research, Changsha, China.,Hunan Fangsheng Pharmaceutical Co., Ltd., Changsha, China
| |
Collapse
|
7
|
Yuan Z, Lin C, Dai L, He Y, Hu J, Xu K, Tao B, Liu P, Cai K. Near-Infrared Light-Activatable Dual-Action Nanoparticle Combats the Established Biofilms of Methicillin-Resistant Staphylococcus aureus and Its Accompanying Inflammation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007522. [PMID: 33690998 DOI: 10.1002/smll.202007522] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 01/23/2021] [Indexed: 05/22/2023]
Abstract
Clinically, inhibition of both bacterial infection and excessive inflammation is a crucial step for improved wound treatments. Herein, the fabrication of near-infrared-light (NIR)-activatable deoxyribonuclease (DNase)-carbon monoxide (CO)@mesoporous polydopamine nanoparticles (MPDA NPs) is demonstrated for efficient elimination of methicillin-resistant Staphylococcus aureus (MRSA) biofilms and the following anti-inflammatory activity. Specifically, thermosensitive CO-gas-releasing donors (CO releasing molecules, FeCO) are first encapsulated into MPDA NPs, followed by covalently immobilizing deoxyribonuclease I (DNase I) on the surfaces of MPDA NPs. DNase I can degrade the extracellular DNA in biofilms, which site specifically destroys the compactness of the biofilms. With NIR irradiation, DNase-CO@MPDA NPs display great photothermal ability, and further trigger on-demand delivery of bactericidal CO gas that can adequately permeate the impaired biofilms. Eventually, they achieve effective MRSA biofilm elimination in virtue of the synergistic effects of both DNase I participation and CO-gas-potentiated photothermal therapy. Importantly, the inflammatory responses of DNase-CO@MPDA NPs and NIR-treated wounds are simultaneously alleviated owing to the anti-inflammatory features of released CO. Finally, NIR-activatable DNase-CO@MPDA NPs accelerate the healing process of MRSA-biofilm-infected cutaneous wounds. Taken together, this phototherapeutic strategy displays great therapeutic potential in treating the formidable clinical problems caused by MRSA biofilms and the accompanying inflammation.
Collapse
Affiliation(s)
- Zhang Yuan
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, P. R. China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, P. R. China
| | - Chuanchuan Lin
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, P. R. China
| | - Liangliang Dai
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, P. R. China
| | - Ye He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, P. R. China
| | - Jingwei Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, P. R. China
| | - Kun Xu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, P. R. China
| | - Bailong Tao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, P. R. China
| | - Peng Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, P. R. China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education College of Bioengineering, Chongqing University, Chongqing, 400044, P. R. China
| |
Collapse
|
8
|
Analyzing Impetus of Regenerative Cellular Therapeutics in Myocardial Infarction. J Clin Med 2020; 9:jcm9051277. [PMID: 32354170 PMCID: PMC7287592 DOI: 10.3390/jcm9051277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 04/20/2020] [Accepted: 04/26/2020] [Indexed: 02/06/2023] Open
Abstract
Both vasculature and myocardium in the heart are excessively damaged following myocardial infarction (MI), hence therapeutic strategies for treating MI hearts should concurrently aim for true cardiac repair by introducing new cardiomyocytes to replace lost or injured ones. Of them, mesenchymal stem cells (MSCs) have long been considered a promising candidate for cell-based therapy due to their unspecialized, proliferative differentiation potential to specific cell lineage and, most importantly, their capacity of secreting beneficial paracrine factors which further promote neovascularization, angiogenesis, and cell survival. As a consequence, the differentiated MSCs could multiply and replace the damaged tissues to and turn into tissue- or organ-specific cells with specialized functions. These cells are also known to release potent anti-fibrotic factors including matrix metalloproteinases, which inhibit the proliferation of cardiac fibroblasts, thereby attenuating fibrosis. To achieve the highest possible therapeutic efficacy of stem cells, the other interventions, including hydrogels, electrical stimulations, or platelet-derived biomaterials, have been supplemented, which have resulted in a narrow to broad range of outcomes. Therefore, this article comprehensively analyzed the progress made in stem cells and combinatorial therapies to rescue infarcted myocardium.
Collapse
|
9
|
Paschou SA, Siasos G, Bletsa E, Stampouloglou PK, Oikonomou E, Antonopoulos AS, Batzias K, Tsigkou V, Mourouzis K, Vryonidou A, Tentolouris N, Vavouranakis M, Tousoulis D. The Effect of DPP-4i on Endothelial Function and Arterial Stiffness in Patients with Type 2 Diabetes: A Systematic Review of Randomized Placebo-controlled Trials. Curr Pharm Des 2020; 26:5980-5987. [PMID: 32303166 DOI: 10.2174/1381612826666200417153241] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 03/25/2020] [Indexed: 12/31/2022]
Abstract
We systematically reviewed the literature regarding the impact of dipeptidyl peptidase-4 inhibitors (DPP-4i) on vascular function, including endothelial function and arterial stiffness, as predictors of atherosclerosis progression and cardiovascular disease in patients with type 2 diabetes mellitus (T2DM). We searched PubMed in order to identify clinical trials that investigated the effect of DPP-4i on vascular function in patients with T2DM when compared with placebo. Although 168 articles were initially found, only 6 studies (total 324 patients) investigated the effect of DPP-4i in comparison with placebo, specifically linagliptin and sitagliptin, and satisfied the inclusion criteria. There are scarce data to indicate that linagliptin may enhance endothelial function and exert a slight beneficial effect on arterial wall properties. Sitagliptin seems to have a neutral effect on these variables. Further trials are needed to elucidate the topic. The standards of reporting were in accordance with the PRISMA guidelines.
Collapse
Affiliation(s)
- Stavroula A Paschou
- First Department of Cardiology, "Hippokration" General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Gerasimos Siasos
- First Department of Cardiology, "Hippokration" General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evanthia Bletsa
- First Department of Cardiology, "Hippokration" General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiota K Stampouloglou
- First Department of Cardiology, "Hippokration" General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Evangelos Oikonomou
- First Department of Cardiology, "Hippokration" General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexios S Antonopoulos
- First Department of Cardiology, "Hippokration" General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Batzias
- First Department of Cardiology, "Hippokration" General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Vasiliki Tsigkou
- First Department of Cardiology, "Hippokration" General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Mourouzis
- First Department of Cardiology, "Hippokration" General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Andromachi Vryonidou
- Department of Endocrinology and Diabetes, Hellenic Red Cross Hospital, Athens, Greece
| | - Nikolaos Tentolouris
- First Department of Propaedeutic and Internal Medicine, Diabetes Center, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Manolis Vavouranakis
- First Department of Cardiology, "Hippokration" General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitrios Tousoulis
- First Department of Cardiology, "Hippokration" General Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
Huizer K, Sacchetti A, Swagemakers S, van der Spek PJ, Dik W, Mustafa DA, Kros JM. Circulating angiogenic cells in glioblastoma: toward defining crucial functional differences in CAC-induced neoplastic versus reactive neovascularization. Neurooncol Adv 2020; 2:vdaa040. [PMID: 32642695 PMCID: PMC7276933 DOI: 10.1093/noajnl/vdaa040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Background In order to identify suitable therapeutic targets for glioma anti-angiogenic therapy, the process of neovascularization mediated by circulating angiogenic cells (CACs) needs to be scrutinized. Methods In the present study, we compared the expression of neovascularization-related genes by 3 circulating CAC subsets (hematopoietic progenitor cells [HPCs], CD34+, and KDR+ cells; internal controls: peripheral blood mononuclear cells and circulating endothelial cells) of treatment-naïve patients with glioblastoma (GBM) to those of patients undergoing reactive neovascularization (myocardial infarction (MI). CACs from umbilical cord (representing developmental neovascularization) and healthy subjects served as controls. Fluorescent-activated cell sorting was used to isolate CACs, RT-PCR to determine the expression levels of a panel of 48 neovascularization-related genes, and Luminex assays to measure plasma levels of 21 CAC-related circulating molecules. Results We found essential differences in gene expression between GBM and MI CACs. GBM CACs had a higher expression of proangiogenic factors (especially, KITL, CXCL12, and JAG1), growth factor and chemotactic receptors (IGF1R, TGFBR2, CXCR4, and CCR2), adhesion receptor monomers (ITGA5 and ITGA6), and matricellular factor POSTN. In addition, we found major differences in the levels of neovascularization-related plasma factors. A strong positive correlation between plasma MMP9 levels and expression of CXCR4 in the CAC subset of HPCs was found in GBM patients. Conclusions Our findings indicate that CAC-mediated neovascularization in GBM is characterized by more efficient CAC homing to target tissue and a more potent proangiogenic response than in physiologic tissue repair in MI. Our findings can aid in selecting targets for therapeutic strategies acting against GBM-specific CACs.
Collapse
Affiliation(s)
- Karin Huizer
- Laboratory for Tumor Immuno-Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Andrea Sacchetti
- Laboratory for Tumor Immuno-Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sigrid Swagemakers
- Department of Pathology and Clinical Bio-Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Peter J van der Spek
- Department of Pathology and Clinical Bio-Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Wim Dik
- Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Dana A Mustafa
- Laboratory for Tumor Immuno-Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Johan M Kros
- Laboratory for Tumor Immuno-Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
11
|
Xu W, Zhang L, Ma S, Zhang Y, Cai Z, Zhang K, Jin D. TRAF5 protects against myocardial ischemia reperfusion injury via AKT signaling. Eur J Pharmacol 2020; 878:173092. [PMID: 32234528 DOI: 10.1016/j.ejphar.2020.173092] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 03/13/2020] [Accepted: 03/26/2020] [Indexed: 12/22/2022]
Abstract
During the processes of myocardial ischemia reperfusion (I/R) injury, inflammation and apoptosis play an important role. I/R and its induced acute myocardial infarction (AMI) with high morbidity and mortality, and there is no effective treatment for it so far. TRAF5 has been shown to regulate inflammation and apoptosis in atherosclerosis, steatosis and melanoma cells, but its function in myocardial I/R injury is still unclear. This study demonstrates that the expression of TRAF5 is significant up-regulation in heart tissues of I/R injury mice and hypoxia/reoxygenation (H/R)-stimulated cardiomyocytes. TRAF5 knockout mice exhibites heavier heart damage, inflammatory response and cell death after myocardial I/R injury. Further, TRAF5 overexpression inhibites inflammation and apoptosis of H/R-stimulated cardiomyocytes. Mechanistically, we prove that TRAF5 promotes the activation of AKT. Overall, our study indicates that TRAF5 can regulate the processes of myocardial I/R injury. TRAF5 can be a new therapy target for myocardial I/R injury.
Collapse
Affiliation(s)
- Weipan Xu
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huang Shi, 435000, China
| | - Li Zhang
- Center for Animal Experiment, Wuhan University, Wuhan, 430000, China
| | - Shanxue Ma
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huang Shi, 435000, China
| | - Yi Zhang
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huang Shi, 435000, China
| | - Zhenxuan Cai
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huang Shi, 435000, China
| | - Kai Zhang
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huang Shi, 435000, China
| | - Daoqun Jin
- Department of Cardiology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Edong Healthcare Group, Huang Shi, 435000, China.
| |
Collapse
|
12
|
Mousavi SH, Dormanesh B, Shahidi S, Johari Moghadam A, Kazemi M, Abediny A. Stromal Cell-derived Factor-1 and CXC Chemokine Receptor Type-4 are Associated with Cardiovascular Disease in Patients under Hemodialysis. Int J Prev Med 2020; 10:219. [PMID: 31929866 PMCID: PMC6941382 DOI: 10.4103/ijpvm.ijpvm_69_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/22/2018] [Indexed: 11/04/2022] Open
Abstract
Background Cardiovascular disease (CVD) is the most common cause of death among patients with end-stage renal disease especially whom under hemodialysis (HD). Stromal cell-derived factor-1 (SDF-1) and its receptor CXC chemokine receptor type-4 (CXCR4) could contribute to CVD. The main aim of this study was to evaluate the association between SDF-1 and CXCR4 with CVD and its related risk factors in patients under HD. Methods Sixty patients under HD and 29 healthy subjects were recruited in the study. The serum levels and relative messenger RNA (mRNA) expressions of SDF-1 and CXCR4 were measured using enzyme-linked immunosorbent assay and real-time polymerase chain reaction in patients and controls, respectively. CVD history of the patients was obtained. Results Twenty patients (33.3%) had a history of CVD. The mean levels of serum and relative mRNA expressions of SDF-1 and CXCR4 were higher in patients than controls and also in patients with a history of CVD than patients without it. The serum levels and relative expressions of SDF-1 and CXCR4 were positively correlated with blood urea nitrogen, parathyroid hormone, and high-sensitivity C-reactive protein and inversely correlated with hemoglobin. The history of CVD was the independent predictor of serum levels of SDF-1 and CXCR4 and also relative mRNA expression of CXCR4. Conclusions The higher levels of serum and relative mRNA expressions of SDF-1 and CXCR4 were associated with CVD in patients under HD. Furthermore, SDF-1 and CXCR4 were associated with several traditional and uremia-related CVD risk factors in such patients.
Collapse
Affiliation(s)
| | - Banafsheh Dormanesh
- Department of Pediatric Nephrology, AJA University of Medical Sciences, Tehran, Iran
| | - Shahrzad Shahidi
- Department of Internal Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.,Isfahan Kidney Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Mohammad Kazemi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amin Abediny
- Department of Pediatric Nephrology, AJA University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Lin YS, Wang FZ, Lei XJ, He JM. [Comparative study with the effect of stromal cell derived factor-1 on osteogenic differentiation of human healthy and inflammatory periodontal ligament stem cells]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2019; 37:469-475. [PMID: 31721491 DOI: 10.7518/hxkq.2019.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
OBJECTIVE This study aims to compare the osteogenic differentiation capability of stem cells derived from human inflammatory periodontal ligament tissues (iPDLSCs) with those of stem cells derived from healthy periodontal ligament tissues (hPDLSCs). Both types of tissues were induced by stromal cell derived factor (SDF-1) in vitro. METHODS iPDLSCs and hPDLSCs were primarily cultured by tissue digestion method and purified by limited dilution cloning. The cells were passaged and identified by stem cell surface marker expression through flow cytometry. Then, we used thiazolyl blue tetrazolium bromide to detect and compare the proliferation capabilities of the iPDLSCs and hPDLSCs. Express of bone volumes were detected by alizarin red staining after SDF-1 was added to the cells. Using alkaline phosphatase, we evaluated the osteogenic differentiation capability of the cells induced by SDF-1. The expression levels of the osteogenesis-related genes of the cells induced by SDF-1 were determined by reverse transcription-polymerase chain reaction. RESULTS After purification, both iPDLSCs and hPDLSCs expressed stem cell markers. hPDLCSs had a higher proliferation capability than iPDLSCs. Osteogenesis-related genes had higher expression levels in the cells induced by SDF-1 than in those without induction (P<0.05). SDF-1 at 50 and 200 ng·mL⁻¹ concentration greatly affected the differen-tiation capabilities of iPDLSCs and hPDLSCs respectively. CONCLUSIONS iPDLSCs and hPDLSCs had osteogenic differentia-tion capability. The level of osteogenic differentiation in normal and inflamed periodontal ligament stem cells increases after SDF-1 induction.
Collapse
Affiliation(s)
- Yong-Sheng Lin
- Key Laboratory of Oral Diseases of Gansu Provincial, Key Laboratory of Stomatology of State Ethnic Affairs Commission, Northwest Minzu University, Lanzhou 730030, China
| | - Feng-Zhi Wang
- Dept. of Oral Medicine, Hainan Stomatological Hospital, Hainan 570100, China
| | - Xiao-Jing Lei
- Dept. of Oral Medicine, Hainan Stomatological Hospital, Hainan 570100, China
| | - Jian-Min He
- Dept. of Stomatology, Gansu Provincial Hospital, Lanzhou 730000, China
| |
Collapse
|
14
|
Bromage DI, Taferner S, He Z, Ziff OJ, Yellon DM, Davidson SM. Stromal cell-derived factor-1α signals via the endothelium to protect the heart against ischaemia-reperfusion injury. J Mol Cell Cardiol 2019; 128:187-197. [PMID: 30738798 PMCID: PMC6408335 DOI: 10.1016/j.yjmcc.2019.02.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 01/18/2019] [Accepted: 02/06/2019] [Indexed: 12/31/2022]
Abstract
AIMS The chemokine stromal derived factor-1α (SDF-1α) is known to protect the heart acutely from ischaemia-reperfusion injury via its cognate receptor, CXCR4. However, the timing and cellular location of this effect, remains controversial. METHODS AND RESULTS Wild type male and female mice were subjected to 40 min LAD territory ischaemia in vivo and injected with either saline (control) or SDF-1α prior to 2 h reperfusion. Infarct size as a proportion of area at risk was assessed histologically using Evans blue and triphenyltetrazolium chloride. Our results confirm the cardioprotective effect of exogenous SDF-1α in mouse ischaemia-reperfusion injury and, for the first time, show protection when SDF-1α is delivered just prior to reperfusion, which has important therapeutic implications. The role of cell type was examined using the same in vivo ischaemia-reperfusion protocol in cardiomyocyte- and endothelial-specific CXCR4-null mice, and by Western blot analysis of endothelial cells treated in vitro. These experiments demonstrated that the acute infarct-sparing effect is mediated by endothelial cells, possibly via the signalling kinases Erk1/2 and PI3K/Akt. Unexpectedly, cardiomyocyte-specific deletion of CXCR4 was found to be cardioprotective per se. RNAseq analysis indicated altered expression of the mitochondrial protein co-enzyme Q10b in these mice. CONCLUSIONS Administration of SDF-1α is cardioprotective when administered prior to reperfusion and may, therefore, have clinical utility. SDF-1α-CXCR4-mediated cardioprotection from ischaemia-reperfusion injury is contingent on the cellular location of CXCR4 activation. Specifically, cardioprotection is mediated by endothelial signalling, while cardiomyocyte-specific deletion of CXCR4 has an infarct-sparing effect per se.
Collapse
Affiliation(s)
- Daniel I Bromage
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - Stasa Taferner
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - Zhenhe He
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - Oliver J Ziff
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK.
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| |
Collapse
|
15
|
Jia C, Chen H, Zhang J, Zhou K, Zhuge Y, Niu C, Qiu J, Rong X, Shi Z, Xiao J, Shi Y, Chu M. Role of pyroptosis in cardiovascular diseases. Int Immunopharmacol 2018; 67:311-318. [PMID: 30572256 DOI: 10.1016/j.intimp.2018.12.028] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/02/2018] [Accepted: 12/11/2018] [Indexed: 01/01/2023]
Abstract
Pyroptosis is a form of programmed necrosis, and is morphologically and mechanistically unique form of programmed cell death compared to others, such as apoptosis and autophagic cell death. More specifically, pyroptosis features gasdermin family-mediated membrane pore formation and subsequent cell lysis, as well as release of pro-inflammatory intracellular contents including IL-1β, IL-18 and HMGB1. Mechanistically, pyroptosis is driven by two main signaling pathways - one mediated by caspase-1 and the other by caspase-4/5/11. Recent studies show that pyroptosis is implicated in several cardiovascular diseases. In this review, we summarize recent scientific discoveries of pyroptosis's involvement in atherosclerosis, myocardial infarction, diabetic cardiomyopathy, reperfusion injury and myocarditis. We also organized new and emerging evidence suggesting that pyroptosis signaling pathways may be potential therapeutic targets in cardiovascular diseases.
Collapse
Affiliation(s)
- Chang Jia
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Huanwen Chen
- University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jian Zhang
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yingzhi Zhuge
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Chao Niu
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Jianxin Qiu
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Xing Rong
- Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhewei Shi
- Department of Cardiology, Zhuji People's Hospital of Zhejiang Province, Shaoxing 311800, China
| | - Jian Xiao
- Pharmacology, School of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Yong Shi
- Comprehensive Breast Health Center, Department of Thyroid and Breast Surgery, Lishui People's Hospital, The Six Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang 323000, China.
| | - Maoping Chu
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Children's Heart Center, Institute of Cardiovascular Development and Translational Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
16
|
He Q, Wang F, Honda T, James J, Li J, Redington A. Loss of miR-144 signaling interrupts extracellular matrix remodeling after myocardial infarction leading to worsened cardiac function. Sci Rep 2018; 8:16886. [PMID: 30443020 PMCID: PMC6237773 DOI: 10.1038/s41598-018-35314-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023] Open
Abstract
We have previously shown that MicroRNA (miR) -144 is a key modulator of the acute cardioprotection associated with remote ischemic preconditioning and post myocardial infarction (MI) remodeling. In this study we examine the biology of the remodeling response after permanent ligation of the left anterior descending coronary artery in male miR-144 KO mice, and wild-type littermates (WT). Collagen content and cross linking were determined by hydroxyproline and pyridinoline assays, MI size and scar thickness were measured post PicoSirius Red staining, and cardiac function was evaluated by echocardiography. miR-144 KO mice developed normally with normal cardiac function, however after MI, infarction size was greater and scar thickness was reduced in miR-144 KO mice compared with WT littermates. miR-144 KO mice had a lower incidence of acute cardiac rupture compared with WT littermates early after MI but there was impaired late remodeling, reflected by increased total cardiac collagen content and collagen cross-linkage associated with changes in Zeb1/LOX1 axis, and decreased left ventricular ejection fraction. We conclude that miR-144 is involved in extracellular matrix remodeling post MI and its loss leads to increased myocardial fibrosis and impaired functional recovery.
Collapse
Affiliation(s)
- Quan He
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Fangfei Wang
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Takashi Honda
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jeanne James
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Jing Li
- Division of Cardiology, Labatt Family Heart Center, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Andrew Redington
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.
| |
Collapse
|
17
|
microRNA-212-induced protection of the heart against myocardial infarction occurs via the interplay between AQP9 and PI3K/Akt signaling pathway. Exp Cell Res 2018; 370:531-541. [DOI: 10.1016/j.yexcr.2018.07.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/09/2018] [Accepted: 07/10/2018] [Indexed: 01/09/2023]
|
18
|
Induced Expression of Endogenous CXCR4 in iPSCs by Targeted CpG Demethylation Enhances Cell Migration Toward the Ligand CXCL12. Inflammation 2018; 42:20-34. [DOI: 10.1007/s10753-018-0869-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
19
|
Champeau M, Póvoa V, Militão L, Cabrini FM, Picheth GF, Meneau F, Jara CP, de Araujo EP, de Oliveira MG. Supramolecular poly(acrylic acid)/F127 hydrogel with hydration-controlled nitric oxide release for enhancing wound healing. Acta Biomater 2018; 74:312-325. [PMID: 29777958 DOI: 10.1016/j.actbio.2018.05.025] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 05/07/2018] [Accepted: 05/15/2018] [Indexed: 01/20/2023]
Abstract
Topical nitric oxide (NO) delivery has been shown to accelerate wound healing. However, delivering NO to wounds at appropriate rates and doses requires new biomaterial-based strategies. Here, we describe the development of supramolecular interpolymer complex hydrogels comprising PEO-PPO-PEO (F127) micelles embedded in a poly(acrylic acid) (PAA) matrix, with S-nitrosoglutathione (GSNO) molecules dissolved in the hydrophilic domain. We show that PAA:F127/GSNO hydrogels start releasing NO upon hydration at rates controlled by their rates of water absorption. SAXS measurements indicate that the supramolecular structure of the hydrogels retains long-range order domains of F127 micelles. The PAA/F1227 hydrogels displayed dense morphologies and reduced rates of hydration. The NO release rates remain constant over the first 200 min, are directly correlated with the hydration rates of the PAA:F127/GSNO hydrogels, and can be modulated in the range of 40 nmol/g h to 1.5 μmol/g h by changing the PAA:F127 mass ratio. Long-term NO-release profiles over 5 days are governed by the first-order exponential decay of GSNO, with half-lives in the range of 0.5-3.4 days. A preliminary in vivo study on full-thickness excisional wounds in mice showed that topical NO release from the PAA:F127/GSNO hydrogels is triggered by exudate absorption and leads to increased angiogenesis and collagen fiber organization, as well as TGF-β, IGF-1, SDF-1, and IL-10 gene expressions in the cicatricial tissue. In summary, these results suggest that hydration-controlled NO release from topical PAA:F127/GSNO hydrogels is a potential strategy for enhancing wound healing. STATEMENT OF SIGNIFICANCE The topical delivery of nitric oxide (NO) to wounds may provide significant beneficial results and represent a promising strategy to treat chronic wounds. However, wound dressings capable of releasing NO after application and allowing the modulation of NO release rates, demand new platforms. Here, we describe a novel strategy to overcome these challenges, based on the use of supramolecular poly(acrylic acid) (PAA):F127 hydrogels charged with the NO donor S-nitrosoglutathione (GSNO) from whereby the NO release can be triggered by exudate absorption and delivered to the wound at rates controlled by the PAA:F127 mass ratio. Preliminary in vivo results offer a proof of concept for this strategy by demonstrating increased angiogenesis; collagen fibers organization; and TGF-β, IGF-1, SDF-1, and IL-10 gene expressions in the cicatricial tissue after topical treatment with a PAA:F127/GSNO hydrogel.
Collapse
|
20
|
Zhang H, Yu S, Zhao X, Mao Z, Gao C. Stromal cell-derived factor-1α-encapsulated albumin/heparin nanoparticles for induced stem cell migration and intervertebral disc regeneration in vivo. Acta Biomater 2018; 72:217-227. [PMID: 29597025 DOI: 10.1016/j.actbio.2018.03.032] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/13/2018] [Accepted: 03/16/2018] [Indexed: 12/30/2022]
Abstract
Intervertebral disc (IVD) degeneration may cause many diseases and pain. Stem cell migration toward the site of IVD degeneration is a key factor for IVD regeneration. In the current study, we prepared albumin/heparin nanoparticles (BHNPs) as injectable carriers of stromal cell-derived factor-1α (SDF-1α, also known as C-X-C motif chemokine 12), a powerful chemoattractant for the homing of bone marrow resident mesenchymal stem cells (MSCs), for protection of the molecule against degradation for a sustained release. The NPs have relatively uniform small size, with a diameter of about 110 nm. The NPs possess a high loading capacity of SDF-1α with a sustained release profile. The bioactivity of the obtained BHNPs/SDF was then studied in vitro and in vivo. The BHNPs/SDF can induce migration of MSCs in a dose-dependent manner in vitro. After injected into the damaged disc, BHNPs/SDF induce much better regeneration of annulus fibrosus and nucleus pulposus, compared to SDF-1α and BHNPs alone, evidenced with better histological grade scores and higher expression of SOX9, Aggrecan, and Collagen type II at the level of both mRNA and protein. This study provides a simple nanoplatform to load SDF-1α and protect it against degradation, with potential application in inductive tissue regeneration in vivo. STATEMENT OF SIGNIFICANCE Stem cell migration toward the site of IVD degeneration is a key event to promote IVD regeneration. In the current study, we prepared albumin/heparin nanoparticles (BHNPs) as injectable carriers to protect SDF-1α against degradation and for the sustained release of the molecule. After injected into the damaged disc, BHNPs/SDF induced much better regeneration of IVD, compared to SDF-1α and BHNPs alone. This study provides a simple nanoplatform to load SDF-1α and protect it from degradation, with potential application in inductive tissue regeneration in vivo.
Collapse
|
21
|
Liu D, Zeng X, Li X, Mehta JL, Wang X. Role of NLRP3 inflammasome in the pathogenesis of cardiovascular diseases. Basic Res Cardiol 2017; 113:5. [PMID: 29224086 DOI: 10.1007/s00395-017-0663-9] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 12/04/2017] [Indexed: 12/21/2022]
Abstract
NLRP3 inflammasome is a key multiprotein signaling platform that tightly controls inflammatory responses and coordinates antimicrobial host defenses by activating caspase-1 for the subsequent maturation of pro-inflammatory cytokines, IL-1β and IL-18, and induces pyroptosis. The assembly and activation of NLRP3 inflammasome are linked to the pathogenesis of several cardiovascular disease risk factors, such as hypertension and diabetes, and their major consequences-myocardial remodeling. The study of the NLRP3 inflammasome in these cardiovascular disease states may uncover important triggers and endogenous modulators of the disease, and lead to new treatment strategies. This review outlines current insights into NLRP3 inflammasome research associated with cardiovascular diseases and discusses the questions that remain in this field.
Collapse
Affiliation(s)
- Dongling Liu
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xiang Zeng
- Department of Epidemiology and Health Statistics, School of Public Health, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xiao Li
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jawahar L Mehta
- Central Arkansas Veterans Healthcare System and the Division of Cardiology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China.
| |
Collapse
|
22
|
Suffee N, Le Visage C, Hlawaty H, Aid-Launais R, Vanneaux V, Larghero J, Haddad O, Oudar O, Charnaux N, Sutton A. Pro-angiogenic effect of RANTES-loaded polysaccharide-based microparticles for a mouse ischemia therapy. Sci Rep 2017; 7:13294. [PMID: 29038476 PMCID: PMC5643514 DOI: 10.1038/s41598-017-13444-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 08/09/2017] [Indexed: 12/20/2022] Open
Abstract
Peripheral arterial disease results from the chronic obstruction of arteries leading to critical hindlimb ischemia. The aim was to develop a new therapeutic strategy of revascularization by using biodegradable and biocompatible polysaccharides-based microparticles (MP) to treat the mouse hindlimb ischemia. For this purpose, we deliver the pro-angiogenic chemokine Regulated upon Activation, Normal T-cell Expressed and Secreted (RANTES)/CCL5 in the mouse ischemic hindlimb, in solution or incorporated into polysaccharide-based microparticles. We demonstrate that RANTES-loaded microparticles improve the clinical score, induce the revascularization and the muscle regeneration in injured mice limb. To decipher the mechanisms underlying RANTES effects in vivo, we demonstrate that RANTES increases the spreading, the migration of human endothelial progenitor cells (EPC) and the formation of vascular network. The main receptors of RANTES i.e. CCR5, syndecan-4 and CD44 expressed at endothelial progenitor cell surface are involved in RANTES-induced in vitro biological effects on EPC. By using two RANTES mutants, [E66A]-RANTES with impaired ability to oligomerize, and [44AANA47]-RANTES mutated in the main RANTES-glycosaminoglycan binding site, we demonstrate that both chemokine oligomerization and binding site to glycosaminoglycans are essential for RANTES-induced angiogenesis in vitro. Herein we improved the muscle regeneration and revascularization after RANTES-loaded MP local injection in mice hindlimb ischemia.
Collapse
Affiliation(s)
- N Suffee
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - C Le Visage
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
| | - H Hlawaty
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - R Aid-Launais
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - V Vanneaux
- APHP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, F-75475, Paris, France.,Inserm UMR1160 et CIC de Biothérapies, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - J Larghero
- APHP, Hôpital Saint-Louis, Unité de Thérapie Cellulaire, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, F-75475, Paris, France.,Inserm UMR1160 et CIC de Biothérapies, Institut Universitaire d'Hématologie, Hôpital Saint-Louis, Paris, France
| | - O Haddad
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - O Oudar
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France
| | - N Charnaux
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France.,Laboratoire de Biochimie, Hôpital Jean Verdier, AP-HP, Bondy, France
| | - A Sutton
- INSERM, U1148, Laboratory for Vascular Translational Science, UFR SMBH, Université Paris 13, Sorbonne Paris Cité, Bobigny, France. .,Laboratoire de Biochimie, Hôpital Jean Verdier, AP-HP, Bondy, France.
| |
Collapse
|
23
|
Danshensu accelerates angiogenesis after myocardial infarction in rats and promotes the functions of endothelial progenitor cells through SDF-1α/CXCR4 axis. Eur J Pharmacol 2017; 814:274-282. [PMID: 28864209 DOI: 10.1016/j.ejphar.2017.08.035] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 01/16/2023]
Abstract
The present study was performed to investigate the potential role of Danshensu in therapeutic angiogenesis in ischemic myocardium and endothelial progenitor cells (EPCs) function. The rat model of myocardial infarction (MI) injury was induced by left anterior descending coronary artery ligation for 14 days. Danshensu significantly alleviated myocardial ischemia injury by ameliorating left ventricular function and reducing infarct size. Furthermore, Danshensu potentiated post-ischemia neovascularization as evidenced by increased microvessel density in infarction boundary zone, as well as the expression of marker proteins vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF). Moreover, Danshensu notably promoted stromal cell-derived factor-1α (SDF-1α) level in plasma and C-X-C chemokine receptor type 4 (CXCR4) expression in peri-infarction myocardium, and AMD3100 (CXCR4 antagonist) could reverse the angiogenic and cardioprotective effects of Danshensu. For in vitro study, EPCs were isolated from bone marrow of rats. On the one hand, Danshensu provided significant cytoprotection against hypoxia insult by boosting EPCs viability and inhibiting apoptosis, and upregulated Akt phosphorylation. On the other hand, Danshensu enhanced proangiogenic functions of EPCs on cell migration and tube formation, and increased SDF-1α and CXCR4 expression. Likewise, the cytoprotection and proangiogenic functions of Danshensu on EPCs were partly negated by LY294002 (PI3K antagonist) and CXCR4 siRNA, respectively. Taken together, our results suggested that the cardioprotection of Danshensu in MI rats may be related to promoting myocardial neovascularization. The possible mechanisms may involve improving EPCs survival in hypoxia condition through Akt phosphorylation, and accelerating EPCs proangiogenic functions through SDF-1α/CXCR4 axis.
Collapse
|
24
|
Ling L, Gu S, Cheng Y. Resveratrol activates endogenous cardiac stem cells and improves myocardial regeneration following acute myocardial infarction. Mol Med Rep 2017; 15:1188-1194. [PMID: 28138705 PMCID: PMC5367360 DOI: 10.3892/mmr.2017.6143] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 10/13/2016] [Indexed: 11/06/2022] Open
Abstract
Stem cell antigen-1-positive (Sca-1+) cardiac stem cells (CSCs) therapy for myocardial regeneration following acute myocardial infarction (AMI) is limited by insufficient cell viability and a high rate of apoptosis, due to the poor regional microenvironment. Resveratrol, which is a compound extracted from red wine, has been reported to protect myocardial tissue post‑AMI by increasing the expression of angiogenic and chemotactic factors. The present study aimed to investigate the effects of resveratrol on Sca‑1+ CSCs, and to optimize Sca‑1+ CSCs therapy for myocardial regeneration post‑AMI. C57/BL6 mice (age, 6 weeks) were divided into two groups, which received intragastric administration of PBS or 2.5 mg/kg.d resveratrol. The endogenous expression of Sca‑1+ CSCs in the heart was assessed on day 7. Furthermore, C57/BL6 mice underwent left anterior descending coronary artery ligation for the construction of an AMI model, and received an injection of 1x106 CSCs into the peri‑ischemic area (n=8/group). Mice received intragastric administration of PBS or resveratrol (2.5 mg/kg.d) for 4 weeks after cell transplantation. Echocardiography was used to evaluate cardiac function 4 weeks after cell transplantation. Capillary density and cardiomyocyte apoptosis in the peri‑ischemic myocardium were assessed by cluster of differentiation 31 immunofluorescent staining and terminal deoxynucleotidyl transferase‑mediated dUTP nick end labeling assay, respectively. Western blot analysis was conducted to detect the protein expression levels of vascular endothelial growth factor (VEGF) and stromal cell‑derived factor (SDF)‑1α in the myocardium. Treatment with resveratrol increased the number of endogenous Sca‑1+ CSCs in heart tissue after 7 days (PBS vs. Res, 1.85±0.41/field vs. 3.14±0.26/field, P<0.05). Furthermore, intragastric administration of resveratrol significantly increased left ventricle (LV) function 4 weeks after AMI, as determined by an increase in LV fractional shortening (CSCs vs. Res + CSCs, 28.82±1.58% vs. 31.18±2.02%, P<0.05), reduced LV end‑diastolic diameter (CSCs vs. Res + CSCs, 0.37±0.01 mm vs. 0.35±0.02 mm, P<0.05), and reduced LV end‑systolic diameter (CSCs vs. Res + CSCs, 0.26±0.01 mm vs. 0.23±0.02 mm, P<0.05). These protective effects were predominantly achieved via an increase in capillary density (CSCs vs. Res + CSCs, 281.02±24.08/field vs. 329.75±36.69/field, P<0.05) and a reduction in cardiomyocyte apoptosis (CSCs vs. Res + CSCs, 1.5±0.54/field vs. 0.83±0.40/field, P<0.05) in peri‑ischemic myocardium. Western blot analysis indicated that VEGF and SDF‑1α were upregulated in resveratrol‑treated myocardium after a 7 day treatment or 4 weeks after AMI (7 days VEGF PBS vs. Res, 0.89±0.07 vs. 1.21±0.02, P<0.05; SDF‑1α PBS vs. Res, 0.66±0.04 vs. 1.33±0.04, P<0.05; 4 weeks VEGF CSCs vs. Res + CSCs, 0.54±0.03 vs. 0.93±0.13, P<0.05; SDF‑1α CSCs vs. Res + CSCs, 0.53±0.03 vs. 0.93±0.03, P<0.05). Resveratrol activated endogenous CSCs, increased capillary density and decreased cardiomyocyte apoptosis in the peri‑ischemic myocardium, and augmented the effects of CSCs transplantation. These effects may be caused by the upregulation of VEGF and SDF‑1α.
Collapse
Affiliation(s)
- Lin Ling
- Department of Cardiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Shaohua Gu
- Department of Nephrology, The Third People's Hospital of Kunshan, Wuxi, Jiangsu 214000, P.R. China
| | - Yan Cheng
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214000, P.R. China
| |
Collapse
|
25
|
Testosterone and Voluntary Exercise Promote Angiogenesis in Hearts of Rats with Diabetes by Enhancing Expression of VEGF-A and SDF-1a. Can J Diabetes 2016; 40:436-441. [PMID: 27444229 DOI: 10.1016/j.jcjd.2016.03.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/28/2016] [Accepted: 03/08/2016] [Indexed: 11/21/2022]
Abstract
OBJECTIVES Impaired angiogenesis in cardiac tissue is a major complication of diabetes. This study was aimed to evaluate the effects of testosterone and voluntary exercise on vascular endothelial growth factor-A (VEGF-A), stromal cell-derived factor 1a (SDF-1a) and myocardial capillary density in heart of rats with diabetes. METHODS Type 1 diabetes was induced by intraperitoneal injection of 55 mg/kg of streptozotocin in 80 male Wistar rats. After 42 days of treatment with testosterone (2 mg/kg/day) or voluntary exercise alone or in combination, angiogenesis was determined in the hearts by immunostaining for PECAM-1/CD31. The expressions of VEGF-A and SDF-1a levels in heart were also determined by the ELISA method. RESULTS Our results showed that capillary density, VEGF-A and SDF-1a levels in the heart were significantly decreased in castrated rats with diabetes, whereas these effects were reversed by testosterone and exercise. Furthermore, simultaneous treatment of castrated rats with diabetes with testosterone and exercise had a synergistic effect on capillary density, VEGF-A and SDF-1a levels in the heart. In the group with diabetes, either testosterone or exercise increased capillary density, VEGF-A and SDF-1a protein levels in heart tissue. However, the effects of combination therapy in rats with diabetes with testosterone and exercise on capillary density, VEGF-A and SDF-1a levels in the heart was synergistic. CONCLUSIONS Our findings suggest that testosterone and exercise can promote neoangiogenesis in rats with diabetes and in castrated rats with diabetes. The proangiogenesis effect of testosterone and exercise is associated with the enhanced expression of VEGF-A and SDF-1a in heart tissue.
Collapse
|
26
|
Autologous bone marrow concentrate enriched in progenitor cells — An adjuvant in the treatment of acute myocardial infarction. INTERNATIONAL JOURNAL OF THE CARDIOVASCULAR ACADEMY 2016. [DOI: 10.1016/j.ijcac.2016.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
27
|
Sun J, Zhao Y, Li Q, Chen B, Hou X, Xiao Z, Dai J. Controlled Release of Collagen-Binding SDF-1α Improves Cardiac Function after Myocardial Infarction by Recruiting Endogenous Stem Cells. Sci Rep 2016; 6:26683. [PMID: 27226084 PMCID: PMC4881239 DOI: 10.1038/srep26683] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 05/06/2016] [Indexed: 11/14/2022] Open
Abstract
Stromal cell-derived factor-1α (SDF-1α) is a well-characterized chemokine that mobilizes stem cells homing to the ischemic heart, which is beneficial for cardiac regeneration. However, clinically administered native SDF-1α diffuses quickly, thus decreasing its local concentration, and results in side effects. Thus, a controlled release system for SDF-1α is required to produce an effective local concentration in the ischemic heart. In this study, we developed a recombinant chemokine, consisting of SDF-1α and a collagen-binding domain, which retains both the SDF-1α and collagen-binding activity (CBD-SDF-1α). In an in vitro assay, CBD-SDF-1α could specifically bind to a collagen gel and achieve sustained release. An intramyocardial injection of CBD-SDF-1α after acute myocardial infarction demonstrated that the protein was largely tethered in the ischemic area and that controlled release had been achieved. Furthermore, CBD-SDF-1α enhanced the recruitment of c-kit positive (c-kit+) stem cells, increased capillary density and improved cardiac function, whereas NAT-SDF-1α had no such beneficial effects. Our findings demonstrate that CBD-SDF-1α can specifically bind to collagen and achieve controlled release both in vitro and in vivo. Local delivery of this protein could mobilize endogenous stem cells homing to the ischemic heart and improve cardiac function after myocardial infarction.
Collapse
Affiliation(s)
- Jie Sun
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Yannan Zhao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Qingguo Li
- Department of Cardiothoracic Surgery, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Bing Chen
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Xianglin Hou
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Zhifeng Xiao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Jianwu Dai
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| |
Collapse
|
28
|
de Almeida Salles T, Zogbi C, de Lima TM, de Godoi Carneiro C, Garcez AT, Barbeiro HV, Antonio EL, Dos Santos L, da Costa Pereira A, Tucci PJF, de Paula Faria D, Soriano FG, Girardi ACC. The contributions of dipeptidyl peptidase IV to inflammation in heart failure. Am J Physiol Heart Circ Physiol 2016; 310:H1760-72. [PMID: 27199127 DOI: 10.1152/ajpheart.00735.2015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 04/12/2016] [Indexed: 01/14/2023]
Abstract
Circulating dipeptidyl peptidase IV (DPPIV) activity correlates with cardiac dysfunction in humans and experimental heart failure (HF) models. Similarly, inflammatory markers are associated with poorer outcomes in HF patients. However, the contributions of DPPIV to inflammation in HF remain elusive. Therefore, this study aimed to investigate whether the cardioprotective effects of DPPIV inhibition after myocardial injury are accompanied by reduced cardiac inflammation, whether circulating DPPIV activity correlates with the levels of systemic inflammatory markers in HF patients, and whether leukocytes and/or splenocytes may be one of the sources of circulating DPPIV in HF. Experimental HF was induced in male Wistar rats by left ventricular myocardial injury after radiofrequency catheter ablation. The rats were divided into three groups: sham, HF, and HF + DPPIV inhibitor (sitagliptin). Six weeks after surgery, cardiac function, perfusion and inflammatory status were evaluated. Sitagliptin treatment improved cardiac function and perfusion, reduced macrophage infiltration, and diminished the levels of inflammatory biomarkers including TNF-α, IL-1β, and CCL2. In HF patients, serum DPPIV activity correlated with CCL2, suggesting that leukocytes may be the source of circulating DPPIV in HF. Unexpectedly, DPPIV release was higher in splenocytes from HF rats and similar in HF circulating mononuclear cells compared with those from sham, suggesting an organ-specific modulation of DPPIV in HF. Collectively, our data provide new evidence that the cardioprotective effects of DPPIV inhibition in HF may be due to suppression of inflammatory cytokines. Moreover, they suggest that a vicious circle between DPPIV and inflammation may contribute to HF development and progression.
Collapse
Affiliation(s)
| | - Camila Zogbi
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Thais Martins de Lima
- Department of Clinical Medicine,University of São Paulo Medical School, São Paulo, Brazil
| | - Camila de Godoi Carneiro
- Laboratory of Nuclear Medicine (LIM 43), University of São Paulo Medical School, São Paulo, Brazil
| | - Alexandre Teles Garcez
- Laboratory of Nuclear Medicine (LIM 43), University of São Paulo Medical School, São Paulo, Brazil
| | - Hermes Vieira Barbeiro
- Department of Clinical Medicine,University of São Paulo Medical School, São Paulo, Brazil
| | - Ednei Luiz Antonio
- Department of Physiology, Federal University of São Paulo, São Paulo, Brazil; and
| | - Leonardo Dos Santos
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitoria, Espírito Santo, Brazil
| | | | | | - Daniele de Paula Faria
- Laboratory of Nuclear Medicine (LIM 43), University of São Paulo Medical School, São Paulo, Brazil
| | | | | |
Collapse
|
29
|
Connelly KA, Advani A, Zhang Y, Advani SL, Kabir G, Abadeh A, Desjardins JF, Mitchell M, Thai K, Gilbert RE. Dipeptidyl peptidase-4 inhibition improves cardiac function in experimental myocardial infarction: Role of stromal cell-derived factor-1α. J Diabetes 2016; 8:63-75. [PMID: 25565455 DOI: 10.1111/1753-0407.12258] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 11/21/2014] [Accepted: 12/11/2014] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND In addition to degrading glucagon-like peptide-1 (GLP-1), dipeptidyl peptidase-4 (DPP-4) inactivates several chemokines, including stromal cell-derived factor-1α (SDF-1α), a pro-angiogenic and cardiomyocyte protective protein. We hypothesized that DPP-4 inhibition may confer benefit following myocardial infarction (MI) in the diabetic setting as a consequence of enhanced SDF-1α availability rather than potentiating GLP-1. To test this we compared the effects of saxagliptin with those of liraglutide and used the SDF-1α receptor (CXCR4) antagonist plerixafor. METHODS Studies were conducted in streptozotocin-diabetic rats. Rats were randomized to receive saxagliptin (10 mg/kg per day), liraglutide (0.2 mg/kg, s.c., b.i.d.), plerixafor (1 mg/kg per day, s.c.), saxagliptin plus plerixafor or vehicle (1% phosphate-buffered saline). Two weeks later, rats underwent experimental MI, with cardiac function examined 4 weeks after MI. RESULTS Glycemic control and MI size were similar in all groups. Four weeks after MI, mortality was reduced in saxagliptin-treated rats compared with vehicle treatment (P < 0.05). Furthermore, rats receiving saxagliptin had improved cardiac function compared with vehicle-treated rats (P < 0.05). Antagonism of CXCR4 prevented the improvement in cardiac function in saxagliptin-treated rats and was associated with increased mortality (P < 0.05). CONCLUSION Saxagliptin-mediated DPP-4 inhibition, but not liraglutide-mediated GLP-1R agonism, improved cardiac function after MI independent of glucose lowering. These findings suggest that non-GLP-1 actions of DPP-4 inhibition, such as SDF-1α potentiation, mediate biological effects.
Collapse
Affiliation(s)
- Kim A Connelly
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Andrew Advani
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Yanling Zhang
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Suzanne L Advani
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Golam Kabir
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Armin Abadeh
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Jean-Francois Desjardins
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Melissa Mitchell
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Kerri Thai
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Richard E Gilbert
- Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Crosstalk between SDF-1/CXCR4 and SDF-1/CXCR7 in cardiac stem cell migration. Sci Rep 2015; 5:16813. [PMID: 26578388 PMCID: PMC4649491 DOI: 10.1038/srep16813] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 10/20/2015] [Indexed: 12/20/2022] Open
Abstract
Stromal cell-derived factor 1 (SDF-1) is a chemokine that can be expressed in injured cardiomyocytes after myocardial infarction (MI). By combining with its receptor CXCR4, SDF-1 induced stem and progenitor cells migration. CXCR7, a novel receptor for SDF-1, has been identified recently. We aimed to explore the roles of SDF-1/CXCR4 and SDF-1/CXCR7 pathway and their crosstalk in CSCs migration. In the present study, CXCR4 and CXCR7 expression were identified in CSCs. Transwell assay showed that SDF-1 caused CSCs migration in a dose- and time-dependent manner, which could be significantly suppressed by CXCR4 or CXCR7 siRNA. Phospho-ERK, phospho-Akt and Raf-1 significantly elevated in CSCs with SDF-1 stimulation. Knockdown of CXCR4 or CXCR7 significantly decreased phospho-ERK or phospho-Akt, respectively, and eventually resulted in the inhibition of CSCs migration. Moreover, western blot showed that MK2206 (Akt inhibitor) increased the expression of phospho-ERK and Raf-1, whereas PD98059 (ERK inhibitor) had no effect on phospho-Akt and Raf-1. GW5074 (Raf-1 inhibitor) upregulated the expression of phospho-ERK, but had no effect on phospho-Akt. The present study indicated that SDF-1/CXCR7/Akt and SDF-1/CXCR4/ERK pathway played important roles in CSCs migration. Akt phosphorylation inhibited Raf-1 activity, which in turn dephosphorylated ERK and negatively regulated CSCs migration.
Collapse
|
31
|
Periyasamy-Thandavan S, Herberg S, Arounleut P, Upadhyay S, Dukes A, Davis C, Johnson M, McGee-Lawrence M, Hamrick MW, Isales CM, Hill WD. Caloric restriction and the adipokine leptin alter the SDF-1 signaling axis in bone marrow and in bone marrow derived mesenchymal stem cells. Mol Cell Endocrinol 2015; 410:64-72. [PMID: 25779533 PMCID: PMC4706462 DOI: 10.1016/j.mce.2015.03.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/27/2015] [Accepted: 03/01/2015] [Indexed: 12/24/2022]
Abstract
Growing evidence suggests that the chemokine stromal cell-derived factor-1 (SDF-1) is essential in regulating bone marrow (BM) derived mesenchymal stromal/stem cell (BMSC) survival, and differentiation to either a pro-osteogenic or pro-adipogenic fate. This study investigates the effects of caloric restriction (CR) and leptin on the SDF-1/CXCR4 axis in bone and BM tissues in the context of age-associated bone loss. For in vivo studies, we collected bone, BM cells and BM interstitial fluid from 12 and 20 month-old C57Bl6 mice fed ad-libitum (AL), and 20-month-old mice on long-term CR with, or without, intraperitoneal injection of leptin for 10 days (10 mg/kg). To mimic conditions of CR in vitro, 18 month murine BMSCs were treated with (1) control (Ctrl): normal proliferation medium, (2) nutrient restriction (NR): low glucose, low serum medium, or (3) NR + leptin: NR medium + 100 ng/ml leptin for 6-48 h. In BMSCs both protein and mRNA expression of SDF-1 and CXCR4 were increased by CR and CR + leptin. In contrast, the alternate SDF-1 receptor CXCR7 was decreased, suggesting a nutrient signaling mediated change in SDF-1 axis signaling in BMSCs. However, in bone SDF-1, CXCR4 and 7 gene expression increase with age and this is reversed with CR, while addition of leptin returns this to the "aged" level. Histologically bone formation was lower in the calorically restricted mice and BM adipogenesis increased, both effects were reversed with the 10 day leptin treatment. This suggests that in bone CR and leptin alter the nutrient signaling pathways in different ways to affect the local action of the osteogenic cytokine SDF-1. Studies focusing on the molecular interaction between nutrient signaling by CR, leptin and SDF-1 axis may help to address age-related musculoskeletal changes.
Collapse
Affiliation(s)
| | | | - Phonepasong Arounleut
- Department of Cellular Biology & Anatomy, Georgia Regents University, Augusta, GA, USA
| | - Sunil Upadhyay
- Department of Cellular Biology & Anatomy, Georgia Regents University, Augusta, GA, USA
| | - Amy Dukes
- Department of Cellular Biology & Anatomy, Georgia Regents University, Augusta, GA, USA
| | - Colleen Davis
- Department of Cellular Biology & Anatomy, Georgia Regents University, Augusta, GA, USA
| | - Maribeth Johnson
- Department of Biostatistics, Georgia Regents University, Augusta, GA, USA
| | - Meghan McGee-Lawrence
- Department of Cellular Biology & Anatomy, Georgia Regents University, Augusta, GA, USA; Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, USA
| | - Mark W Hamrick
- Department of Cellular Biology & Anatomy, Georgia Regents University, Augusta, GA, USA; Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, USA; Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA, USA
| | - Carlos M Isales
- Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, USA; Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA, USA; Department of Neuroscience and Regenerative Medicine, Georgia Regents University, Augusta, GA, USA
| | - William D Hill
- Department of Cellular Biology & Anatomy, Georgia Regents University, Augusta, GA, USA; Institute for Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA, USA; Department of Orthopaedic Surgery, Georgia Regents University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
32
|
Takahashi A, Ihara M, Yamazaki S, Asanuma H, Asakura M, Kitakaze M. Impact of Either GLP-1 Agonists or DPP-4 Inhibitors on Pathophysiology of Heart Failure. Int Heart J 2015; 56:372-6. [PMID: 26104180 DOI: 10.1536/ihj.15-028] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Since diabetes mellitus (DM) is the most common cause of heart failure (HF), it is critically important to clarify whether incretin hormones including glucagon-like peptide-1 (GLP-1), which play an important role in blood glucose control, mediate cardioprotection. There are many lines of basic research evidence indicating that GLP-1 improves the pathophysiology of HF: In murine and canine HF models, either GLP-1 analogues or DPP-IV inhibitors improved cardiac functions. The first question that arises is how either GLP-1 analogues or DPP-IV inhibitors mediate cardioprotection. Cardiovascular diseases are tightly linked to impaired glucose tolerance (IGT): IGT is not only one of the causes of cardiovascular events but also the result of HF. Indeed, the treatment of IGT improved HF, showing that one of the mechanisms attributable to DPP-IV inhibitors is related to the improvement of IGT. Intriguingly, either DPP-IV inhibitors or GLP-1 analogues mediate cardioprotection even without IGT, suggesting two possible explanations: One is that GLP-1 analogues directly activate the prosurvival kinases, such as Akt and Erk1/2, and another is that DPP-IV inhibition increases cardioprotective peptides such as BNP and SDF-1α. The next question is whether cardioprotection is translated to clinical medicine. Small scale clinical trials proved their cardioprotective effects; however, several large scale clinical trials have not proved the beneficial effects of DPP-IV inhibitors. Taken together, GLP-1 analogues or DPP-IV inhibitors can mediate cardioprotection, however, what needs to be clarified is who mainly receives their benefits among the patients with cardiovascular diseases and/or DM.
Collapse
Affiliation(s)
- Ayako Takahashi
- Department of Clinical Medicine and Development, National Cerebral and Cardiovascular Center
| | | | | | | | | | | |
Collapse
|
33
|
Boucek RJ, Steele J, Jacobs JP, Steele P, Asante-Korang A, Quintessenza J, Steele A. Ex vivo paracrine properties of cardiac tissue: Effects of chronic heart failure. J Heart Lung Transplant 2015; 34:839-48. [DOI: 10.1016/j.healun.2014.07.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 06/11/2014] [Accepted: 07/10/2014] [Indexed: 12/15/2022] Open
|
34
|
Gu XL, Liu L, Lu XD, Liu ZR. Serum CXCL12 Levels as a Novel Predictor of Future Stroke Recurrence in Patients with Acute Ischemic Stroke. Mol Neurobiol 2015; 53:2807-2814. [PMID: 25833101 DOI: 10.1007/s12035-015-9151-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 03/19/2015] [Indexed: 12/28/2022]
Abstract
Previous studies had shown that CXC chemokine ligand-12 (CXCL12) plays a significant role in animal models of ischemic stroke, but its role in human stroke is unclear. The aim of this study was to test the relationship between elevated serum circulating CXCL12 levels and the 1-year stroke recurrence in Chinese patients with acute ischemic stroke (AIS). All consecutive patients with first-ever acute ischemic stroke from January 2011 to September 2013 were recruited to participate in the study. Serum levels of CXCL12 and National Institute of Health Stroke Scale (NIHSS) were measured at the time of admission. Logistic regression analysis was used to evaluate the stroke recurrence according to serum CXCL12 levels. Receiver operating characteristic (ROC) curve was used to evaluate the accuracy of serum CXCL12 in predicting stroke recurrence. Clinical follow-up was performed at 1 year. In our study, 248 patients finished the 1-year follow-up. At 1-year follow-up, 31 patients had a recurrence ischemic stroke. The median CXCL12 levels were significantly higher in those who sustained a recurrence ischemic stroke compared with those who did not [24.2 ng/mL (IQR 15.4-33.7) vs 6.5 ng/mL (IQR 3.4-10.2); Z = 8.258, P < 0.0001]. In multivariate analysis, there was an increased risk of stroke recurrence associated with serum CXCL12 levels ≥12.15 ng/mL (OR 9.122, 95 % CI 6.103-15.104) after adjusting for above possible confounders. The time to recurrence stroke distribution between patients with baseline CXCL12 levels ≥12.15 ng/mL and those with baseline CXCL12 levels <12.15 ng/mL were significantly different (P < 0.0001, log-rank test). Elevated circulating CXCL12 levels at admission are strongly associated with the future recurrence of ischemic stroke in Chinese patients with AIS. Further studies are warranted to confirm this association and define the role for CXCL12 as a novel predictor biomarker for stroke recurrence.
Collapse
Affiliation(s)
- Xiao-Lin Gu
- Department of Neurology, The People's Hospital of Laiwu City, No. 1, Xuehudajie, Changshao North Road, Laiwu, 271100, Shandong Province, China.
| | - Lin Liu
- Department of Neurology, The People's Hospital of Laiwu City, No. 1, Xuehudajie, Changshao North Road, Laiwu, 271100, Shandong Province, China
| | - Xiang-Dong Lu
- Department of Neurosurgery, The People's Hospital of Laiwu City, Laiwu, 271100, Shandong Province, China
| | - Zhen-Rui Liu
- Department of Neurosurgery, The People's Hospital of Laiwu City, Laiwu, 271100, Shandong Province, China
| |
Collapse
|
35
|
Abstract
OBJECTIVE Evaluate the risk of female breast cancer associated with HIV-CXCR4 (X4) tropism as determined by various genotypic measures. METHODS A breast cancer case-control study, with pairwise comparisons of tropism determination methods, was conducted. From the Women's Interagency HIV Study repository, one stored plasma specimen was selected from 25 HIV-infected cases near the breast cancer diagnosis date and 75 HIV-infected control women matched for age and calendar date. HIV-gp120 V3 sequences were derived by Sanger population sequencing (PS) and 454-pyro deep sequencing (DS). Sequencing-based HIV-X4 tropism was defined using the geno2pheno algorithm, with both high-stringency DS [false-positive rate (3.5) and 2% X4 cutoff], and lower stringency DS (false-positive rate, 5.75 and 15% X4 cutoff). Concordance of tropism results by PS, DS, and previously performed phenotyping was assessed with kappa (κ) statistics. Case-control comparisons used exact P values and conditional logistic regression. RESULTS In 74 women (19 cases, 55 controls) with complete results, prevalence of HIV-X4 by PS was 5% in cases vs 29% in controls (P = 0.06; odds ratio, 0.14; confidence interval: 0.003 to 1.03). Smaller case-control prevalence differences were found with high-stringency DS (21% vs 36%, P = 0.32), lower stringency DS (16% vs 35%, P = 0.18), and phenotyping (11% vs 31%, P = 0.10). HIV-X4 tropism concordance was best between PS and lower stringency DS (93%, κ = 0.83). Other pairwise concordances were 82%-92% (κ = 0.56-0.81). Concordance was similar among cases and controls. CONCLUSIONS HIV-X4 defined by population sequencing (PS) had good agreement with lower stringency DS and was significantly associated with lower odds of breast cancer.
Collapse
|
36
|
Salles TA, dos Santos L, Barauna VG, Girardi ACC. Potential role of dipeptidyl peptidase IV in the pathophysiology of heart failure. Int J Mol Sci 2015; 16:4226-49. [PMID: 25690036 PMCID: PMC4346954 DOI: 10.3390/ijms16024226] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 02/05/2015] [Accepted: 02/09/2015] [Indexed: 12/14/2022] Open
Abstract
Dipeptidyl peptidase IV (DPPIV) is a widely expressed multifunctional serine peptidase that exists as a membrane-anchored cell surface protein or in a soluble form in the plasma and other body fluids. Numerous substrates are cleaved at the penultimate amino acid by DPPIV, including glucagon-like peptide-1 (GLP-1), brain natriuretic peptide (BNP) and stromal cell-derived factor-1 (SDF-α), all of which play important roles in the cardiovascular system. In this regard, recent reports have documented that circulating DPPIV activity correlates with poorer cardiovascular outcomes in human and experimental heart failure (HF). Moreover, emerging evidence indicates that DPPIV inhibitors exert cardioprotective and renoprotective actions in a variety of experimental models of cardiac dysfunction. On the other hand, conflicting results have been found when translating these promising findings from preclinical animal models to clinical therapy. In this review, we discuss how DPPIV might be involved in the cardio-renal axis in HF. In addition, the potential role for DPPIV inhibitors in ameliorating heart disease is revised, focusing on the effects of the main DPPIV substrates on cardiac remodeling and renal handling of salt and water.
Collapse
Affiliation(s)
- Thiago A Salles
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 05403-000, SP, Brazil.
| | - Leonardo dos Santos
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitoria 29043-900, ES, Brazil.
| | - Valério G Barauna
- Department of Physiological Sciences, Federal University of Espírito Santo, Vitoria 29043-900, ES, Brazil.
| | - Adriana C C Girardi
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo 05403-000, SP, Brazil.
| |
Collapse
|
37
|
Iliodromitis EK, Cohen MV, Dagres N, Andreadou I, Kremastinos DT, Downey JM. What is Wrong With Cardiac Conditioning? We May be Shooting at Moving Targets. J Cardiovasc Pharmacol Ther 2015; 20:357-69. [DOI: 10.1177/1074248414566459] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/02/2014] [Indexed: 12/29/2022]
Abstract
Early recanalization of the occluded culprit coronary artery clearly reduces infarct size in both animal models and patients and improves clinical outcomes. Unfortunately, reperfusion can seldom be accomplished before some myocardium infarcts. As a result there has been an intensive search for interventions that will make the heart resistant to infarction so that reperfusion could salvage more myocardium. A number of interventions have been identified in animal models, foremost being ischemic preconditioning. It protects by activating signaling pathways that prevent lethal permeability transition pores from forming in the heart’s mitochondria at reperfusion. Such conditioning can be accomplished in a clinically relevant manner either by staccato reperfusion (ischemic postconditioning) or by pharmacological activation of the conditioning signaling pathways prior to reperfusion. Unfortunately, clinical trials of ischemic postconditioning and pharmacologic conditioning have been largely disappointing. We suggest that this may be caused by inappropriate use as models intended to mimic the clinical scenario of young healthy animals that receive none of the many drugs currently given to our patients. Patients may be resistant to some forms of conditioning because of comorbidities, for example, diabetes, or they may already be conditioned by adjunct medications, for example, P2Y12 inhibitors or opioids. Incremental technological improvements in patient care may render some approaches to cardioprotection redundant, and thus the clinical target may be continually changing, while our animal models have not kept pace. In remote conditioning, a limb is subjected to ischemia/reperfusion prior to or during coronary reperfusion. Its mechanism is not as well understood as that of ischemic preconditioning, but the results have been very encouraging. In the present article, we will review ischemic, remote, and pharmacologic conditioning and possible confounders that could interfere with their efficacy in clinical trials in 2 settings of myocardial ischemia: (1) primary angioplasty in acute myocardial infarction and (2) elective angioplasty.
Collapse
Affiliation(s)
- Efstathios K. Iliodromitis
- Second University Department of Cardiology, Medical School, Attikon General Hospital, University of Athens, Athens, Greece
| | - Michael V. Cohen
- Department of Physiology, University of South Alabama College of Medicine, Mobile, AL, USA
- Department of Medicine, University of South Alabama College of Medicine, Mobile, AL, USA
| | - Nikolaos Dagres
- Second University Department of Cardiology, Medical School, Attikon General Hospital, University of Athens, Athens, Greece
| | - Ioanna Andreadou
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Athens, Athens, Greece
| | - Dimitrios Th. Kremastinos
- Second University Department of Cardiology, Medical School, Attikon General Hospital, University of Athens, Athens, Greece
| | - James M. Downey
- Second University Department of Cardiology, Medical School, Attikon General Hospital, University of Athens, Athens, Greece
| |
Collapse
|
38
|
Heusch G, Bøtker HE, Przyklenk K, Redington A, Yellon D. Remote ischemic conditioning. J Am Coll Cardiol 2015; 65:177-95. [PMID: 25593060 PMCID: PMC4297315 DOI: 10.1016/j.jacc.2014.10.031] [Citation(s) in RCA: 490] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 10/16/2014] [Accepted: 10/22/2014] [Indexed: 12/12/2022]
Abstract
In remote ischemic conditioning (RIC), brief, reversible episodes of ischemia with reperfusion in one vascular bed, tissue, or organ confer a global protective phenotype and render remote tissues and organs resistant to ischemia/reperfusion injury. The peripheral stimulus can be chemical, mechanical, or electrical and involves activation of peripheral sensory nerves. The signal transfer to the heart or other organs is through neuronal and humoral communications. Protection can be transferred, even across species, with plasma-derived dialysate and involves nitric oxide, stromal derived factor-1α, microribonucleic acid-144, but also other, not yet identified factors. Intracardiac signal transduction involves: adenosine, bradykinin, cytokines, and chemokines, which activate specific receptors; intracellular kinases; and mitochondrial function. RIC by repeated brief inflation/deflation of a blood pressure cuff protects against endothelial dysfunction and myocardial injury in percutaneous coronary interventions, coronary artery bypass grafting, and reperfused acute myocardial infarction. RIC is safe and effective, noninvasive, easily feasible, and inexpensive.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Centre Essen, University of Essen Medical School, Essen, Germany.
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital Skejby, Aarhus, Denmark
| | - Karin Przyklenk
- Cardiovascular Research Institute, Wayne State University School of Medicine, Detroit, Michigan
| | - Andrew Redington
- Hospital for Sick Children and University of Toronto, Toronto, Ontario, Canada
| | - Derek Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| |
Collapse
|
39
|
Schesny MK, Monaghan M, Bindermann AH, Freund D, Seifert M, Eble JA, Vogel S, Gawaz MP, Hinderer S, Schenke-Layland K. Preserved bioactivity and tunable release of a SDF1-GPVI bi-specific protein using photo-crosslinked PEGda hydrogels. Biomaterials 2014; 35:7180-7. [DOI: 10.1016/j.biomaterials.2014.04.116] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 04/29/2014] [Indexed: 11/29/2022]
|
40
|
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors represent 2 distinct classes of incretin-based therapies used for the treatment of type 2 diabetes mellitus. Activation of GLP-1R signaling or inhibition of DPP-4 activity produces a broad range of overlapping and unique cardiovascular actions. Native GLP-1 regulates cardiovascular biology via activation of the classical GLP-1R, or through GLP-1(9-36), a cardioactive metabolite generated by DPP-4-mediated cleavage. In contrast, clinically approved GLP-1R agonists are not cleaved to GLP-1(9-36) and produce the majority of their actions through the classical GLP-1R. The cardiovascular mechanisms engaged by DPP-4 inhibition are more complex, encompassing increased levels of intact GLP-1, reduced levels of GLP-1(9-36), and changes in levels of numerous cardioactive peptides. Herein we review recent experimental and clinical advances that reveal how GLP-1R agonists and DPP-4 inhibitors affect the normal and diabetic heart and coronary vasculature, often independent of changes in blood glucose. Improved understanding of the complex science of incretin-based therapies is required to optimize the selection of these therapeutic agents for the treatment of diabetic patients with cardiovascular disease.
Collapse
Affiliation(s)
- John R Ussher
- From the Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, Canada
| | - Daniel J Drucker
- From the Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Ontario, Canada.
| |
Collapse
|
41
|
Connelly KA, Zhang Y, Advani A, Advani SL, Thai K, Yuen DA, Gilbert RE. DPP-4 inhibition attenuates cardiac dysfunction and adverse remodeling following myocardial infarction in rats with experimental diabetes. Cardiovasc Ther 2014; 31:259-67. [PMID: 22963483 DOI: 10.1111/1755-5922.12005] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
AIMS Following myocardial infarction (MI), individuals with diabetes have a two-fold increase in the risk of heart failure, due in part to excessive loss of cardiac microvasculature. Endothelial integrity and restitution are mediated in part by stromal cell-derived factor-1α (SDF-1α), a chemokine that is elaborated by ischemic tissue but rapidly degraded by dipeptidyl peptidase-4 (DPP-4). Accordingly, we hypothesized that inhibiting this enzyme may confer benefit following myocardial infarction in the diabetic setting beyond its effect on glycemia. METHODS AND RESULTS Fischer F344 rats with streptozotocin (STZ)-diabetes were randomized to receive vehicle or the DPP-4 inhibitor, sitagliptin (300 mg/kg/day). Two weeks later, animals underwent experimental MI, induced by ligation of the left anterior descending coronary artery. Cardiac function was assessed by conductance catheterization and echocardiography along with cardiac structure 4 weeks post-MI. Following MI, untreated diabetic rats developed both systolic and diastolic cardiac dysfunction, in association with endothelial cell loss, fibrosis, and myocyte hypertrophy. Without affecting plasma glucose, sitagliptin treatment led to an improvement in passive left ventricular compliance, increased endothelial cell density, reduced myocyte hypertrophy, and a reduction in the abundance of collagen 1 (all P < 0.05). Systolic function was unchanged. CONCLUSIONS This study shows that DPP-4 inhibition attenuates several, but not all, aspects of cardiac dysfunction and adverse remodeling in the post-MI setting.
Collapse
Affiliation(s)
- Kim Alexander Connelly
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
42
|
Long MY, Li HH, Pen XZ, Huang MQ, Luo DY, Wang PS. Expression of chemokine receptor-4 in bone marrow mesenchymal stem cells on experimental rat abdominal aortic aneurysms and the migration of bone marrow mesenchymal stem cells with stromal-derived factor-1. Kaohsiung J Med Sci 2014; 30:224-8. [DOI: 10.1016/j.kjms.2013.12.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 12/03/2013] [Indexed: 10/25/2022] Open
|
43
|
Dipeptidyl peptidase-4 inhibitors are associated with improved left ventricular diastolic function after acute myocardial infarction in diabetic patients. Heart Vessels 2014; 30:696-701. [PMID: 24736945 DOI: 10.1007/s00380-014-0509-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Accepted: 03/28/2014] [Indexed: 12/30/2022]
Abstract
Dipeptidyl peptidase-4 (DPP4) is an integral membrane glycoprotein that modulates the pathological state of diabetes mellitus (DM), and DPP4 inhibitors are a new class of anti-type-2 DM drugs. Recent preclinical studies have associated DPP4 inhibition with improved myocardial systolic and diastolic function. Based on preclinical findings, we investigated associations between the administration of DPP4 inhibitors and cardiac function after acute myocardial infarction (AMI) in a clinical setting. We enrolled 34 patients with diabetes who were treated for acute myocardial infarction at our hospital between January 2010 and December 2012. We retrospectively compared changes in cardiac parameters determined by trans-thoracic echocardiography between patients treated with (DPP4-I group; n = 13) or without (non-DPP4-I group; n = 21) a DPP4 inhibitor during follow-up. The values of E/e' and of e'/a' significantly decreased and increased, respectively, in the DPP4-I, compared with the non-DPP4-I group (-2.53 ± 5.53 vs. 2.58 ± 5.68, p = 0.038 and 0.08 ± 0.23 vs. -0.12 ± 0.21, p = 0.036, respectively). We concluded that DPP4 inhibitors could improve E/e' and e'/a' in patients with DM and AMI and thus might be effective for treating left ventricular diastolic failure.
Collapse
|
44
|
Bromage DI, Davidson SM, Yellon DM. Stromal derived factor 1α: a chemokine that delivers a two-pronged defence of the myocardium. Pharmacol Ther 2014; 143:305-15. [PMID: 24704323 PMCID: PMC4127789 DOI: 10.1016/j.pharmthera.2014.03.009] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 03/20/2014] [Indexed: 01/03/2023]
Abstract
Alleviating myocardial injury associated with ST elevation myocardial infarction is central to improving the global burden of coronary heart disease. The chemokine stromal cell-derived factor 1α (SDF-1α) has dual potential benefit in this regard. Firstly, SDF-1α is up-regulated in experimental and clinical studies of acute myocardial infarction (AMI) and regulates stem cell migration to sites of injury. SDF-1α delivery to the myocardium after AMI is associated with improved stem cell homing, angiogenesis, and left ventricular function in animal models, and improvements in heart failure and quality of life in humans. Secondly, SDF-1α may have a role in remote ischaemic conditioning (RIC), the phenomenon whereby non-lethal ischaemia–reperfusion applied to an organ or tissue remote from the heart protects the myocardium from lethal ischaemia–reperfusion injury (IRI). SDF-1α is increased in the serum of rats subjected to RIC and protects against myocardial IRI in ex vivo studies. Despite these potential pleiotropic effects, a limitation of SDF-1α is its short plasma half-life due to cleavage by dipeptidyl peptidase-4 (DPP-4). However, DPP-4 inhibitors increase the half-life of SDF-1α by preventing its degradation and are also protective against lethal IRI. In summary, SDF-1 potentially delivers a ‘two-pronged’ defence of the myocardium: acutely protecting it from IRI while simultaneously stimulating repair by recruiting stem cells to the site of injury. In this article we examine the evidence for acute and chronic cardioprotective roles of SDF-1α and discuss potential therapeutic manipulations of this mechanism with DPP-4 inhibitors to protect against lethal tissue injury in the clinical setting.
Collapse
Affiliation(s)
- Daniel I Bromage
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London WC1E 6HX, United Kingdom
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London WC1E 6HX, United Kingdom
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, 67 Chenies Mews, London WC1E 6HX, United Kingdom
| |
Collapse
|
45
|
Gong Y, Zhao Y, Li Y, Fan Y, Hoover-Plow J. Plasminogen regulates cardiac repair after myocardial infarction through its noncanonical function in stem cell homing to the infarcted heart. J Am Coll Cardiol 2014; 63:2862-72. [PMID: 24681141 DOI: 10.1016/j.jacc.2013.11.070] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 11/05/2013] [Accepted: 11/26/2013] [Indexed: 10/25/2022]
Abstract
OBJECTIVES The purpose of this study was to investigate the role of plasminogen (Plg) in stem cell-mediated cardiac repair and regeneration after myocardial infarction (MI). BACKGROUND An MI induces irreversible tissue damage, eventually leading to heart failure. Bone marrow (BM)-derived stem cells promote tissue repair and regeneration after MI. Thrombolytic treatment with Plg activators significantly improves the clinical outcome in MI by restoring cardiac perfusion. However, the role of Plg in stem cell-mediated cardiac repair remains unclear. METHODS An MI was induced in Plg-deficient (Plg(-/-)) and wild-type (Plg(+/+)) mice by ligation of the left anterior descending coronary artery. Stem cells were visualized by in vivo tracking of green fluorescent protein (GFP)-expressing BM cells after BM transplantation. Cardiac function, stem cell homing, and signaling pathways downstream of Plg were examined. RESULTS Granulocyte colony-stimulating factor, a stem cell mobilizer, significantly promoted BM-derived stem cell (GFP(+)c-kit(+) cell) recruitment into the infarcted heart and stem cell-mediated cardiac repair in Plg(+/+) mice. However, Plg deficiency markedly inhibited stem cell homing and cardiac repair, suggesting that Plg is critical for stem cell-mediated cardiac repair. Moreover, Plg regulated C-X-C chemokine receptor type 4 (CXCR4) expression in stem cells in vivo and in vitro through matrix metalloproteinase-9. Lentiviral reconstitution of CXCR4 expression in BM cells successfully rescued stem cell homing to the infarcted heart in Plg-deficient mice, indicating that CXCR4 has a critical role in Plg-mediated stem cell homing after MI. CONCLUSIONS These findings have identified a novel role for Plg in stem cell-mediated cardiac repair after MI. Thus, targeting Plg may offer a new therapeutic strategy for stem cell-mediated cardiac repair after MI.
Collapse
Affiliation(s)
- Yanqing Gong
- Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Departments of Cardiovascular Medicine and Molecular Cardiology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio; Division of Translational Medicine and Human Genetics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania.
| | - Yujing Zhao
- Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Departments of Cardiovascular Medicine and Molecular Cardiology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Ying Li
- Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Departments of Cardiovascular Medicine and Molecular Cardiology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| | - Yi Fan
- Department of Radiation Oncology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Jane Hoover-Plow
- Joseph J. Jacobs Center for Thrombosis and Vascular Biology, Departments of Cardiovascular Medicine and Molecular Cardiology, Cleveland Clinic Lerner Research Institute, Cleveland, Ohio
| |
Collapse
|
46
|
Abstract
Inflammasomes are multiple protein complexes that serve as molecular platforms to activate caspase-1 and regulate maturation of a potent proinflammatory cytokine, interleukin (IL)-1β, as well as proinflammatory cell death, pyroptosis. Although several types of inflammasomes have been reported so far, recent investigations indicate that the NLRP3 inflammasome recognizes non-microbial danger signals and leads to sterile inflammatory responses in various disease conditions. Sterile inflammatory responses are also implicated in the development of myocardial infarction (MI). In particular, IL-1β is an early and prominent mediator of inflammatory responses in MI, suggesting the pathophysiologic role of NLRP3 inflammasomes in MI. This review highlights the current state of knowledge regarding the role of NLRP3 inflammasomes in MI.
Collapse
Affiliation(s)
- Masafumi Takahashi
- Division of Inflammation Research, Center for Molecular Medicine, Jichi Medical University
| |
Collapse
|
47
|
Abstract
Despite their small size and anucleate status, platelets have diverse roles in vascular biology. Not only are platelets the cellular mediator of thrombosis, but platelets are also immune cells that initiate and accelerate many vascular inflammatory conditions. Platelets are linked to the pathogenesis of inflammatory diseases such as atherosclerosis, malaria infection, transplant rejection, and rheumatoid arthritis. In some contexts, platelet immune functions are protective, whereas in others platelets contribute to adverse inflammatory outcomes. In this review, we will discuss platelet and platelet-derived mediator interactions with the innate and acquired arms of the immune system and platelet-vessel wall interactions that drive inflammatory disease. There have been many recent publications indicating both important protective and adverse roles for platelets in infectious disease. Because of this new accumulating data, and the fact that infectious disease continues to be a leading cause of death globally, we will also focus on new and emerging concepts related to platelet immune and inflammatory functions in the context of infectious disease.
Collapse
|
48
|
Gilbert RE. Augmenting Endothelial Repair in Diabetes: Role of Bone Marrow-Derived Cells. Can J Diabetes 2013; 37:315-8. [DOI: 10.1016/j.jcjd.2013.06.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 06/23/2013] [Accepted: 06/24/2013] [Indexed: 01/09/2023]
|
49
|
Gilbert RE. Potential for cardiac protection with dipeptidyl peptidase-4 inhibitors: the stromal cell-derived factor-1α hypothesis. J Diabetes 2013; 5:258-60. [PMID: 23586735 DOI: 10.1111/1753-0407.12053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Richard E Gilbert
- Keenan Research Centre of the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada.
| |
Collapse
|
50
|
Davidson SM, Selvaraj P, He D, Boi-Doku C, Yellon RL, Vicencio JM, Yellon DM. Remote ischaemic preconditioning involves signalling through the SDF-1α/CXCR4 signalling axis. Basic Res Cardiol 2013; 108:377. [PMID: 23917520 DOI: 10.1007/s00395-013-0377-6] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 06/26/2013] [Accepted: 07/29/2013] [Indexed: 01/04/2023]
Abstract
Ischaemic preconditioning is one of the most potent experimental modalities known to decrease infarct size after ischaemia and reperfusion. Much interest has been stimulated by the phenomenon of remote ischaemic conditioning (RIC), in which the preconditioning stimulus is applied to a limb remote from the heart to stimulate cardioprotection via an unidentified humoral factor, believed to be a protein between 3.5 and 15 kDa. Stromal cell-derived factor-1 (SDF-1α or CXCL12) is a chemokine of 10 kDa that is induced by hypoxia and recruits stem cells, but also exerts direct, acute, cardioprotection via its receptor, CXCR4. The serum dipeptidase DPPIV cleaves and inactivates SDF-1α. We measured SDF-1α in rat plasma and found it was significantly increased by RIC. DPPIV activity was unchanged after RIC, suggesting that increased synthesis or release or SDF-1α caused the increase in plasma levels. AMD3100, a highly specific inhibitor of CXCR4, was used to investigate the hypothesis that SDF-1α is involved in RIC. RIC in rats, which decreased infarct size from 53 ± 3 % to 27 ± 3 % (n = 6, P < 0.05), was blocked in rats treated with AMD3100 (40 ± 4 %). RIC also improved functional recovery of cardiac papillary muscle, and this, too, was blocked by AMD3100. Direct application of SDF-1α was confirmed to be protective in this model and was blocked by AMD3100. RIC stimulates SDF-1α release, and this 10-kDa peptide appears to be required for the mechanism of RIC.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | | | | | | | | | | | | |
Collapse
|