51
|
Asymmetric Dimethyl Arginine as a Biomarker of Atherosclerosis in Rheumatoid Arthritis. Mediators Inflamm 2018; 2018:3897295. [PMID: 29576746 PMCID: PMC5822828 DOI: 10.1155/2018/3897295] [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: 09/13/2017] [Accepted: 11/27/2017] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease is the main cause of morbidity and mortality in rheumatoid arthritis (RA). Despite the advent on new drugs targeting the articular manifestations, the burden of cardiovascular disease is still an unmet need in the management of RA. The pathophysiology of accelerated atherosclerosis associated to RA is not yet fully understood, and reliable and specific markers of early cardiovascular involvement are still lacking. Asymmetric dimethylarginine is gaining attention for its implication in the pathogenesis of endothelial dysfunction and as biomarkers of subclinical atherosclerosis. Moreover, the metabolic pathway of methylarginines offers possible targets for therapeutic interventions to decrease the cardiovascular risk. The purpose of this review is to describe the main causes of increased methylarginine levels in RA, their implication in accelerated atherosclerosis, the possible role as biomarkers of cardiovascular risk, and finally the available data on current pharmacological treatment.
Collapse
|
52
|
Endothelial Progenitor Cells for Ischemic Stroke: Update on Basic Research and Application. Stem Cells Int 2017; 2017:2193432. [PMID: 28900446 PMCID: PMC5576438 DOI: 10.1155/2017/2193432] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/03/2017] [Indexed: 01/14/2023] Open
Abstract
Ischemic stroke is one of the leading causes of human death and disability worldwide. So far, ultra-early thrombolytic therapy is the most effective treatment. However, most patients still live with varying degrees of neurological dysfunction due to its narrow therapeutic time window. It has been confirmed in many studies that endothelial progenitor cells (EPCs), as a kind of adult stem cells, can protect the neurovascular unit by repairing the vascular endothelium and its secretory function, which contribute to the recovery of neurological function after an ischemic stroke. This paper reviews the basic researches and clinical trials of EPCs especially in the field of ischemic stroke and addresses the combination of EPC application with new technologies, including neurovascular intervention, synthetic particles, cytokines, and EPC modification, with the aim of shedding some light on the application of EPCs in treating ischemic stroke in the future.
Collapse
|
53
|
Berezin AE. Endothelial progenitor cells dysfunction and impaired tissue reparation: The missed link in diabetes mellitus development. Diabetes Metab Syndr 2017; 11:215-220. [PMID: 27578620 DOI: 10.1016/j.dsx.2016.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/22/2016] [Indexed: 01/08/2023]
Abstract
Diabetes mellitus (DM) is considered a leading cause of premature cardiovascular (CV) mortality and morbidity in general population and in individuals with known CV disease. Recent animal and clinical studies have shown that reduced number and weak function of endothelial progenitor cells (EPCs) may not only indicate to higher CV risk, but contribute to the impaired heart and vessels reparation in patients with DM. Moreover, EPCs having a protective impact on the vasculature may mediate the functioning of other organs and systems. Therefore, EPCs dysfunction is probably promising target for DM treatment strategy, while the role of restoring of EPCs number and functionality in CV risk diminish and reduce of DM-related complications is not fully clear. The aim of the review is summary of knowledge regarding EPCs dysfunction in DM patients.
Collapse
Affiliation(s)
- Alexander E Berezin
- State Medical University of Zaporozhye, 26, Mayakovsky av., Zaporozhye, UA, 69035, Ukraine.
| |
Collapse
|
54
|
The atheroma plaque secretome stimulates the mobilization of endothelial progenitor cells ex vivo. J Mol Cell Cardiol 2017; 105:12-23. [DOI: 10.1016/j.yjmcc.2017.02.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Revised: 02/13/2017] [Accepted: 02/14/2017] [Indexed: 01/28/2023]
|
55
|
Thrombus leukocytes exhibit more endothelial cell-specific angiogenic markers than peripheral blood leukocytes do in acute coronary syndrome patients, suggesting a possibility of trans-differentiation: a comprehensive database mining study. J Hematol Oncol 2017; 10:74. [PMID: 28335793 PMCID: PMC5364721 DOI: 10.1186/s13045-017-0440-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 03/07/2017] [Indexed: 11/10/2022] Open
Abstract
Background Current angiogenic therapies for cancers and cardiovascular diseases have not yet achieved expected benefits, which reflects the need for improved understanding of angiogenesis. In this study, we focused on solving the problem of whether tissues have different angiogenic potentials (APs) in physiological conditions and how angiogenesis is regulated in various disease conditions. Methods In healthy and diseased human and mouse tissues, we profiled the expression of 163 angiogenic genes, including transcription regulators (TRs), growth factors and receptors (GF/Rs), cytokines and chemokines (C/Cs), and proteases and inhibitors (P/Is). TRs were categorized as inflammatory, homeostatic, and endothelial cell-specific TRs, and C/Cs were categorized as pro-angiogenic, anti-angiogenic, and bi-functional C/Cs. Results We made the following findings: (1) the human heart, muscle, eye, pancreas, and lymph node are among the tissues with the highest APs; (2) tissues with high APs have more active angiogenic pathways and angiogenic C/C responses; (3) inflammatory TRs dominate regulation of all angiogenic C/Cs; homeostatic TRs regulate all to a lower extent, while endothelial cell-specific TRs mainly regulate pro-angiogenic and bi-functional C/Cs; (4) tissue AP is positively correlated with the expression of oxygen sensors PHD2 and HIF1B, VEGF pathway gene VEGFB, and stem cell gene SOX2; (5) cancers of the digestive system tend to have increased angiogenesis dominated by endothelial cell-specific pro-angiogenic pathways, while lung cancer and prostate cancer have significantly decreased angiogenesis; and (6) endothelial cell-specific pro-angiogenic pathways are significantly increased in thrombus-derived leukocytes in patients with acute coronary artery disease. Conclusions Our results demonstrate that thrombus-derived leukocytes express more endothelial cell-specific angiogenic markers to directly promote angiogenesis after myocardial infarction and that certain solid tumors may be more sensitive to anti-angiogenic therapies than others. Electronic supplementary material The online version of this article (doi:10.1186/s13045-017-0440-0) contains supplementary material, which is available to authorized users.
Collapse
|
56
|
Huang Q, Yang Z, Zhou JP, Luo Y. HMGB1 induces endothelial progenitor cells apoptosis via RAGE-dependent PERK/eIF2α pathway. Mol Cell Biochem 2017; 431:67-74. [PMID: 28251435 DOI: 10.1007/s11010-017-2976-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 02/16/2017] [Indexed: 02/06/2023]
Abstract
Studies have demonstrated that the high-mobility group 1B protein (HMGB1) could regulate endothelial progenitor cell (EPC) homing, but the effect of HMGB1 on EPC apoptosis and associated mechanisms are still unclear. The aim of this study was to investigate the effects of HMGB1 on EPC apoptosis and the possible involvement of the endoplasmic reticulum (ER) stress pathway. EPC apoptosis was determined by flow cytometry. The expressions of PERK, eIF2α, and CHOP were detected by western blotting. Additionally, the effects of PERK shRNA on the biological behaviors of EPCs were assessed. Our results showed that incubation of EPCs with HMGB1 (0.1-1 μg/ml) for 12-48 h induced apoptosis as well as activated ER stress transducers, as assessed by up-regulating PERK protein expression and eIF2α phosphorylation in a dose or time-dependent manner. Moreover, HMGB1-mediated EPC apoptosis and CHOP expression were dramatically suppressed by PERK shRNA or a specific eIF2α inhibitor (salubrinal). Importantly, a blocking antibody specifically targeted against RAGE (anti-RAGE antibody) markedly inhibited HMGB1-induced EPC apoptosis and ER stress marker protein (PERK, eIF2α, and CHOP) expression levels. Our novel findings suggest that HMGB1 triggered EPC apoptosis in a manner of RAGE-mediated activation of the PERK/eIF2α pathway.
Collapse
Affiliation(s)
- Qun Huang
- Department of Child Health Care, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, Hunan, China
| | - Zhen Yang
- Department of Hypertension and Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, 510080, China
| | - Ji-Peng Zhou
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Xiangya Road 87#, Changsha, 410008, Hunan, China.
| | - Ying Luo
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Xiangya Road 87#, Changsha, 410008, Hunan, China.
| |
Collapse
|
57
|
Reply to: "Endothelial progenitor cell release is usually considered a beneficial effect: Problems in interpreting the acute effects of e-cigarette use". Atherosclerosis 2017; 258:164-165. [PMID: 28159320 DOI: 10.1016/j.atherosclerosis.2017.01.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 01/18/2017] [Indexed: 11/23/2022]
|
58
|
Zhang M, Jiang L. Oxidized low-density lipoprotein decreases VEGFR2 expression in HUVECs and impairs angiogenesis. Exp Ther Med 2016; 12:3742-3748. [PMID: 28105106 DOI: 10.3892/etm.2016.3823] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 09/06/2016] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis (AS), which is triggered by endothelial cell injury, evolves into a chronic inflammatory disease. Oxidized low-density lipoprotein (ox-LDL) is an important risk factor for the development of atherosclerosis; ox-LDL induces atherosclerotic plaque formation via scavenging receptors. The present study used ox-LDL-treated human umbilical vein endothelial cells (HUVECs) to investigate the effect of ox-LDL on angiogenesis. ox-LDL decreased HUVEC proliferation by MTT, induced apoptosis by Annexin V-fluorescein isothiocyanate (FITC) staining and markedly suppressed HUVEC tube formation by the Matrigel assay in a dose-dependent manner. Angiogenesis has been correlated with monocyte invasion, plaque instability and atherosclerotic lesion formation. In addition, ox-LDL induced the overproduction of reactive oxygen species using DCFH-DA staining and increased caspase-3 activity. Vascular endothelial growth factor receptor 2 (VEGFR2) were detected by quantitative polymerase chain reaction and western blot analysis and has previously been observed to have a key role in angiogenesis. Furthermore, the present study demonstrated that the abundance of VEGFR2 was decreased in ox-LDL-treated HUVECs. These results suggested that ox-LDL impairs angiogenesis via VEGFR2 degradation, thus suggesting that VEGFR2 may be involved in adaptation to oxidative stress and AS.
Collapse
Affiliation(s)
- Min Zhang
- Division of Cardiology, Shanghai Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200336, P.R. China
| | - Li Jiang
- Division of Cardiology, Shanghai Tongren Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200336, P.R. China
| |
Collapse
|
59
|
Gupta A, Bhatnagar S. Vasoregression: A Shared Vascular Pathology Underlying Macrovascular And Microvascular Pathologies? OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 19:733-53. [PMID: 26669709 DOI: 10.1089/omi.2015.0128] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vasoregression is a common phenomenon underlying physiological vessel development as well as pathological microvascular diseases leading to peripheral neuropathy, nephropathy, and vascular oculopathies. In this review, we describe the hallmarks and pathways of vasoregression. We argue here that there is a parallel between characteristic features of vasoregression in the ocular microvessels and atherosclerosis in the larger vessels. Shared molecular pathways and molecular effectors in the two conditions are outlined, thus highlighting the possible systemic causes of local vascular diseases. Our review gives us a system-wide insight into factors leading to multiple synchronous vascular diseases. Because shared molecular pathways might usefully address the diagnostic and therapeutic needs of multiple common complex diseases, the literature analysis presented here is of broad interest to readership in integrative biology, rational drug development and systems medicine.
Collapse
Affiliation(s)
- Akanksha Gupta
- 1 Computational and Structural Biology Laboratory, Division of Biotechnology, Netaji Subhas Institute of Technology , Dwarka, New Delhi, India .,2 Department of Biotechnology, IMS Engineering College , Ghaziabad, India
| | - Sonika Bhatnagar
- 1 Computational and Structural Biology Laboratory, Division of Biotechnology, Netaji Subhas Institute of Technology , Dwarka, New Delhi, India
| |
Collapse
|
60
|
Suzuki Y, Tada-Oikawa S, Hayashi Y, Izuoka K, Kataoka M, Ichikawa S, Wu W, Zong C, Ichihara G, Ichihara S. Single- and double-walled carbon nanotubes enhance atherosclerogenesis by promoting monocyte adhesion to endothelial cells and endothelial progenitor cell dysfunction. Part Fibre Toxicol 2016; 13:54. [PMID: 27737702 PMCID: PMC5064793 DOI: 10.1186/s12989-016-0166-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 10/05/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The use of carbon nanotubes has increased lately. However, the cardiovascular effect of exposure to carbon nanotubes remains elusive. The present study investigated the effects of pulmonary exposure to single-walled carbon nanotubes (SWCNTs) and double-walled carbon nanotubes (DWCNTs) on atherosclerogenesis using normal human aortic endothelial cells (HAECs) and apolipoprotein E-deficient (ApoE-/-) mice, a model of human atherosclerosis. METHODS HAECs were cultured and exposed to SWCNTs or DWCNTs for 16 h. ApoE-/- mice were exposed to SWCNTs or DWCNTs (10 or 40 μg/mouse) once every other week for 10 weeks by pharyngeal aspiration. RESULTS Exposure to CNTs increased the expression level of adhesion molecule (ICAM-1) and enhanced THP-1 monocyte adhesion to HAECs. ApoE-/- mice exposed to CNTs showed increased plaque area in the aorta by oil red O staining and up-regulation of ICAM-1 expression in the aorta, compared with vehicle-treated ApoE-/- mice. Endothelial progenitor cells (EPCs) are mobilized from the bone marrow into the circulation and subsequently migrate to the site of endothelial damage and repair. Exposure of ApoE-/- mice to high-dose SWCNTs or DWCNTs reduced the colony-forming units of EPCs in the bone marrow and diminished their migration function. CONCLUSION The results suggested that SWCNTs and DWCNTs enhanced atherosclerogenesis by promoting monocyte adhesion to endothelial cells and inducing EPC dysfunction.
Collapse
Affiliation(s)
- Yuka Suzuki
- Graduate School of Regional Innovation Studies, Mie University, 1577 Kurimamachiya-cho, Tsu, 514-8507, Japan
| | - Saeko Tada-Oikawa
- Graduate School of Regional Innovation Studies, Mie University, 1577 Kurimamachiya-cho, Tsu, 514-8507, Japan
| | - Yasuhiko Hayashi
- Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Kiyora Izuoka
- Graduate School of Regional Innovation Studies, Mie University, 1577 Kurimamachiya-cho, Tsu, 514-8507, Japan
| | - Misa Kataoka
- Graduate School of Regional Innovation Studies, Mie University, 1577 Kurimamachiya-cho, Tsu, 514-8507, Japan
| | - Shunsuke Ichikawa
- Graduate School of Regional Innovation Studies, Mie University, 1577 Kurimamachiya-cho, Tsu, 514-8507, Japan
| | - Wenting Wu
- Department of Occupational and Environmental Health, Nagoya Univeristy Graduate School of Medicine, Nagoya, Japan
| | - Cai Zong
- Department of Occupational and Environmental Health, Nagoya Univeristy Graduate School of Medicine, Nagoya, Japan
| | - Gaku Ichihara
- Department of Occupational and Environmental Health, Tokyo Univeristy of Science, Noda, Japan
| | - Sahoko Ichihara
- Graduate School of Regional Innovation Studies, Mie University, 1577 Kurimamachiya-cho, Tsu, 514-8507, Japan.
| |
Collapse
|
61
|
Addition of aspirin to a fish oil-rich diet decreases inflammation and atherosclerosis in ApoE-null mice. J Nutr Biochem 2016; 35:58-65. [PMID: 27394692 DOI: 10.1016/j.jnutbio.2016.05.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/25/2016] [Accepted: 05/25/2016] [Indexed: 12/31/2022]
Abstract
Aspirin (ASA) is known to alter the production of potent inflammatory lipid mediators, but whether it interacts with omega-3 fatty acids (FAs) from fish oil to affect atherosclerosis has not been determined. The goal was to investigate the impact of a fish oil-enriched diet alone and in combination with ASA on the production of lipid mediators and atherosclerosis. ApoE(-/-) female mice were fed for 13weeks one of the four following diets: omega-3 FA deficient (OD), omega-3 FA rich (OR) (1.8g omega-3 FAs/kg·diet per day), omega-3 FA rich plus ASA (ORA) (0.1g ASA/kg·diet per day) or an omega-3 FA deficient plus ASA (ODA) with supplement levels equivalent to human doses. Plasma lipids, atherosclerosis, markers of inflammation, hepatic gene expression and aortic lipid mediators were determined. Hepatic omega-3 FAs were markedly higher in OR (9.9-fold) and ORA (7-fold) groups. Mice in both OR and ORA groups had 40% less plasma cholesterol in very low-density lipoprotein-cholesterol and low-density lipoprotein fractions, but aortic plaque area formation was only significantly lower in the ORA group (5.5%) compared to the OD group (2.5%). Plasma PCSK9 protein levels were approximately 70% lower in the OR and ORA groups. Proinflammatory aortic lipid mediators were 50%-70% lower in the ODA group than in the OD group and more than 50% lower in the ORA group. In summary, less aortic plaque lesions and aortic proinflammatory lipid mediators were observed in mice on the fish oil diet plus ASA vs. just the fish oil diet.
Collapse
|
62
|
Koller L, Hohensinner P, Sulzgruber P, Blum S, Maurer G, Wojta J, Hülsmann M, Niessner A. Prognostic relevance of circulating endothelial progenitor cells in patients with chronic heart failure. Thromb Haemost 2016; 116:309-16. [PMID: 27412580 DOI: 10.1160/th16-01-0051] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 04/28/2016] [Indexed: 12/20/2022]
Abstract
Novel strategies for a tailored risk prediction in chronic heart failure (CHF) are crucial to identify patients at very high risk for an improved patient management and to specify treatment regimens. Endothelial progenitor cells (EPCs) are an important endogenous repair mechanism with the ability to counteract endothelial injury and the possibility of new vessel formation. We hypothesised that exhaustion of circulating EPCs may be a suitable prognostic biomarker in patients with CHF. EPCs, defined as CD34+CD45dimKDR+ cells, were analysed using fluorescence-activated cell sorting. EPCs were measured in 185 patients with CHF including 87 (47 %) patients with ischaemic aetiology and 98 (53 %) patients with non-ischaemic CHF and followed for a median time of 2.7 years. During this period, 34.7 % of patients experienced the primary study endpoint all-cause mortality. EPC count was a significant and independent inverse predictor of mortality with an hazard ratio hazard ratio (HR) per increase of one standard deviation (1-SD) of 0.47 (95 % confidence interval [CI]: 0.35-0.61; p<0.001) and remained significant after multivariable adjustment for a comprehensive set of cardiovascular risk factors and potential confounders with a HR per 1-SD of 0.54 (95 % CI: 0.4-0.73; p<0.001). EPCs further demonstrated additional prognostic information indicated by improvements in C-statistic, net reclassification index and integrated discrimination increment. In conclusion, in our study circulating EPCs turned out as strong and independent inverse predictors of mortality underlining the importance of an impaired endothelial repair mechanism in the pathophysiology and progression of CHF.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Alexander Niessner
- Alexander Niessner, MD, MSc, Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria, Tel.: +43 1 404004614, Fax: +43 1 404004216, E-mail:
| |
Collapse
|
63
|
Li X, Fang P, Li Y, Kuo YM, Andrews AJ, Nanayakkara G, Johnson C, Fu H, Shan H, Du F, Hoffman NE, Yu D, Eguchi S, Madesh M, Koch WJ, Sun J, Jiang X, Wang H, Yang X. Mitochondrial Reactive Oxygen Species Mediate Lysophosphatidylcholine-Induced Endothelial Cell Activation. Arterioscler Thromb Vasc Biol 2016; 36:1090-100. [PMID: 27127201 DOI: 10.1161/atvbaha.115.306964] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 04/15/2016] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Hyperlipidemia-induced endothelial cell (EC) activation is considered as an initial event responsible for monocyte recruitment in atherogenesis. However, it remains poorly defined what is the mechanism underlying hyperlipidemia-induced EC activation. Here, we tested a novel hypothesis that mitochondrial reactive oxygen species (mtROS) serve as signaling mediators for EC activation in early atherosclerosis. APPROACH AND RESULTS Metabolomics and transcriptomics analyses revealed that several lysophosphatidylcholine (LPC) species, such as 16:0, 18:0, and 18:1, and their processing enzymes, including Pla2g7 and Pla2g4c, were significantly induced in the aortas of apolipoprotein E knockout mice during early atherosclerosis. Using electron spin resonance and flow cytometry, we found that LPC 16:0, 18:0, and 18:1 induced mtROS in primary human aortic ECs, independently of the activities of nicotinamide adenine dinucleotide phosphate oxidase. Mechanistically, using confocal microscopy and Seahorse XF mitochondrial analyzer, we showed that LPC induced mtROS via unique calcium entry-mediated increase of proton leak and mitochondrial O2 reduction. In addition, we found that mtROS contributed to LPC-induced EC activation by regulating nuclear binding of activator protein-1 and inducing intercellular adhesion molecule-1 gene expression in vitro. Furthermore, we showed that mtROS inhibitor MitoTEMPO suppressed EC activation and aortic monocyte recruitment in apolipoprotein E knockout mice using intravital microscopy and flow cytometry methods. CONCLUSIONS ATP synthesis-uncoupled, but proton leak-coupled, mtROS increase mediates LPC-induced EC activation during early atherosclerosis. These results indicate that mitochondrial antioxidants are promising therapies for vascular inflammation and cardiovascular diseases.
Collapse
Affiliation(s)
- Xinyuan Li
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Pu Fang
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Yafeng Li
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Yin-Ming Kuo
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Andrew J Andrews
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Gayani Nanayakkara
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Candice Johnson
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Hangfei Fu
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Huimin Shan
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Fuyong Du
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Nicholas E Hoffman
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Daohai Yu
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Satoru Eguchi
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Muniswamy Madesh
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Walter J Koch
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Jianxin Sun
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Xiaohua Jiang
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Hong Wang
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.)
| | - Xiaofeng Yang
- From the Centers for Metabolic Disease Research, Cardiovascular Research, Thrombosis Research (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., S.E., X.J., H.W., X.Y.), Department of Pharmacology (X.L., P.F., Y.L., G.N., C.J., H.F., H.S., F.D., W.J.K., X.J., H.W., X.Y.), Department of Biochemistry (N.E.H., M.M.), Department of Physiology (S.E.), Center for Translational Medicine (N.E.H., M.M., W.J.K.), and Department of Clinical Sciences (D.Y.), Temple University School of Medicine, Philadelphia, PA; Department of Cancer Biology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA (Y.-M.K., A.J.A.); and Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA (J.S.).
| |
Collapse
|
64
|
Kim BS, Jacobs D, Emontzpohl C, Goetzenich A, Soppert J, Jarchow M, Schindler L, Averdunk L, Kraemer S, Marx G, Bernhagen J, Pallua N, Schlemmer HP, Simons D, Stoppe C. Myocardial Ischemia Induces SDF-1α Release in Cardiac Surgery Patients. J Cardiovasc Transl Res 2016; 9:230-238. [PMID: 27055858 DOI: 10.1007/s12265-016-9689-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 03/22/2016] [Indexed: 01/07/2023]
Abstract
In the present observational study, we measured serum levels of the chemokine stromal cell-derived factor-1α (SDF-1α) in 100 patients undergoing cardiac surgery with cardiopulmonary bypass at seven distinct time points including preoperative values, myocardial ischemia, reperfusion, and the postoperative course. Myocardial ischemia triggered a marked increase of SDF-1α serum levels whereas cardiac reperfusion had no significant influence. Perioperative SDF-1α serum levels were influenced by patients' characteristics (e.g., age, gender, aspirin intake). In an explorative analysis, we observed an inverse association between SDF-1α serum levels and the incidence of organ dysfunction. In conclusion, time of myocardial ischemia was identified as the key stimulus for a significant upregulation of SDF-1α, indicating its role as a marker of myocardial injury. The inverse association between SDF-1α levels and organ dysfunction association encourages further studies to evaluate its organoprotective properties in cardiac surgery patients.
Collapse
Affiliation(s)
- Bong-Sung Kim
- Department of Plastic Surgery, Hand Surgery, Burn Center, RWTH Aachen University, Aachen, Germany
| | - Denise Jacobs
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Christoph Emontzpohl
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Andreas Goetzenich
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Josefin Soppert
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Mareike Jarchow
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Lisa Schindler
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany.,Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany
| | - Luisa Averdunk
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Sandra Kraemer
- Department of Thoracic, Cardiac and Vascular Surgery, University Hospital, RWTH Aachen, Aachen, Germany
| | - Gernot Marx
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany
| | - Jürgen Bernhagen
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Ludwig-Maximilians-University, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Norbert Pallua
- Department of Plastic Surgery, Hand Surgery, Burn Center, RWTH Aachen University, Aachen, Germany
| | | | - David Simons
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Christian Stoppe
- Department of Intensive Care Medicine, University Hospital, RWTH Aachen, Aachen, Germany.
| |
Collapse
|
65
|
Zeng H, Jiang Y, Tang H, Ren Z, Zeng G, Yang Z. Abnormal phosphorylation of Tie2/Akt/eNOS signaling pathway and decreased number or function of circulating endothelial progenitor cells in prehypertensive premenopausal women with diabetes mellitus. BMC Endocr Disord 2016; 16:13. [PMID: 26936372 PMCID: PMC4776390 DOI: 10.1186/s12902-016-0093-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 02/25/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUNDS The number and activity of circulating endothelial progenitor cells (EPCs) in prehypertension is preserved in premenopausal women. However, whether this favorable effect still exists in prehypertensive premenopausal women with diabetes is not clear. METHODS This study compared the number and functional activity of circulating EPCs in normotensive or prehypertensive premenopausal women without diabetes mellitus and normotensive or prehypertensive premenopausal women with diabetes mellitus, evaluated the vascular endothelial function in each groups, and investigated the possible underlying mechanism. RESULTS We found that compared with normotensive premenopausal women, the number and function of circulating EPCs, as well as endothelial function evaluated by flow-mediated dilatation (FMD) in prehypertensive premenopausal women were preserved. In parallel, the Tie2/Akt/eNOS signaling pathway and the plasma NO level or NO secretion of circulating EPCs in prehypertensive premenopausal women was also retained. However, in presence of normotension or prehypertension with diabetes mellitus, the number or function of circulating EPCs and FMD in premenopausal women decreased. Similarly, the phosphorylation of Tie2/Akt/eNOS signaling pathway and the plasma NO level or NO secretion of circulating EPCs was reduced in prehypertension premenopausal with diabetes mellitus. CONCLUSION The present findings firstly demonstrate that the unfavorable effects of diabetes mellitus on number and activity of circulating EPCs in prehypertension premenopausal women, which is at least partially related to the abnormal phosphorylation of Tie2/Akt/eNOS signaling pathway and subsequently reduced nitric oxide bioavailability. The Tie2/Akt/eNOS signaling pathway may be a potential target of vascular protection in prehypertensive premenopausal women with diabetes mellitus.
Collapse
Affiliation(s)
- Haitao Zeng
- Center for Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Yanping Jiang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China
| | - Hailin Tang
- Cancer Center, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Zi Ren
- Center for Reproductive Medicine, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, People's Republic of China
| | - Gaofeng Zeng
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China.
| | - Zhen Yang
- Department of Hypertension & Vascular Disease, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China.
| |
Collapse
|
66
|
Vazquez G, Solanki S, Dube P, Smedlund K, Ampem P. On the Roles of the Transient Receptor Potential Canonical 3 (TRPC3) Channel in Endothelium and Macrophages: Implications in Atherosclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:185-99. [PMID: 27161230 DOI: 10.1007/978-3-319-26974-0_9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the cardiovascular and hematopoietic systems the Transient Receptor Potential Canonical 3 (TRPC3) channel has a well-recognized role in a number of signaling mechanisms that impact the function of diverse cells and tissues in physiology and disease. The latter includes, but is not limited to, molecular and cellular mechanisms associated to the pathogenesis of cardiac hypertrophy, hypertension and endothelial dysfunction. Despite several of these functions being closely related to atherorelevant mechanisms, the potential roles of TRPC3 in atherosclerosis, the major cause of coronary artery disease, have remained largely unexplored. Over recent years, a series of studies from the authors' laboratory revealed novel functions of TRPC3 in mechanisms related to endothelial inflammation, monocyte adhesion to endothelium and survival and apoptosis of macrophages. The relevance of these new TRPC3 functions to atherogenesis has recently began to receive validation through studies in mouse models of atherosclerosis with conditional gain or loss of TRPC3 function. This chapter summarizes these novel findings and provides a discussion of their impact in the context of atherosclerosis, in an attempt to delineate a framework for further exploration of this terra incognita in the TRPC field.
Collapse
Affiliation(s)
- Guillermo Vazquez
- Department of Physiology and Pharmacology, and Center for Hypertension and Personalized Medicine, University of Toledo College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Transverse Dr., UTHSC Mail stop 1008, Toledo, OH, 43614, USA.
| | - Sumeet Solanki
- Department of Physiology and Pharmacology, and Center for Hypertension and Personalized Medicine, University of Toledo College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Transverse Dr., UTHSC Mail stop 1008, Toledo, OH, 43614, USA
| | - Prabhatachandra Dube
- Department of Physiology and Pharmacology, and Center for Hypertension and Personalized Medicine, University of Toledo College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Transverse Dr., UTHSC Mail stop 1008, Toledo, OH, 43614, USA
| | - Kathryn Smedlund
- Department of Physiology and Pharmacology, and Center for Hypertension and Personalized Medicine, University of Toledo College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Transverse Dr., UTHSC Mail stop 1008, Toledo, OH, 43614, USA
| | - Prince Ampem
- Department of Physiology and Pharmacology, and Center for Hypertension and Personalized Medicine, University of Toledo College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Transverse Dr., UTHSC Mail stop 1008, Toledo, OH, 43614, USA
| |
Collapse
|
67
|
Pitha J, Králová Lesná I, Hubáček JA, Sekerková A, Lánská V, Adámková V, Dorobantu M, Nicolescu R, Steiner R, Ivić V, Borbely A, Papp Z, Vari SG. Smoking impairs and circulating stem cells favour the protective effect of the T allele of the connexin37 gene in ischemic heart disease--A multinational study. Atherosclerosis 2016; 244:73-8. [PMID: 26588185 DOI: 10.1016/j.atherosclerosis.2015.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 11/04/2015] [Accepted: 11/04/2015] [Indexed: 01/26/2023]
Abstract
BACKGROUND The connexin 37 (Cx37) gene is considered to be a candidate gene for ischemic heart disease (IHD). We analyzed the association between the C1019 > T (Pro319 > Ser) variant of the Cx37 gene and IHD in patients in the Czech Republic, Croatia, Hungary and Romania with regard to the presence/absence of selected cardiovascular risk factors (RF). In a complementary study, we analyzed the association between the Cx37 gene and circulating stem and endothelial progenitor cells in healthy women. METHODS The study population comprised 2396 patients (663 women) with IHD. The control population comprised 2476 subjects (1, 337 women). Additionally, in 662 healthy women, the association between the Cx37 gene and circulating stem and endothelial progenitor cells was analyzed. RESULTS The strongest protective effect of the Cx37 T allele was detected in non-smoking patients without diabetes mellitus and hypertension (OR 0.610, 95% CI 0.377-0.990); a similar effect was found in non-smoking men (OR 0.781, 95% CI 0.628-0.971); weaker effect was found in non-smoking women (OR 0.768, 95% CI 0.560-1.050). In non-smoking healthy women, stem cells were significantly higher in TT than in CT and CC carriers (p for trend 0.011). Additionally, non-smoking TT carriers had significantly higher number of stem cells than past and current smoking TT carriers (p for trend = 0.006); no such trend was found in CT and CC carriers. CONCLUSIONS The protective effect of the T allele of the Cx37 gene might be strongly modified by smoking; in women, this effect could be mediated through stem cells.
Collapse
Affiliation(s)
- Jan Pitha
- Center for Experimental Medicine, Laboratory for Atherosclerosis Research, Institute for Clinical & Experimental Medicine, Prague, Czech Republic.
| | - Ivana Králová Lesná
- Center for Experimental Medicine, Laboratory for Atherosclerosis Research, Institute for Clinical & Experimental Medicine, Prague, Czech Republic
| | - Jaroslav A Hubáček
- Center for Experimental Medicine, Laboratory for Atherosclerosis Research, Institute for Clinical & Experimental Medicine, Prague, Czech Republic
| | - Alena Sekerková
- Department of Clinical and Transplant Immunology, Institute of Clinical & Experimental Medicine, Prague, Czech Republic
| | - Věra Lánská
- Medicine Statistic Unit, Institute of Clinical & Experimental Medicine, Prague, Czech Republic
| | - Věra Adámková
- Department of Preventive Cardiology, Institute of Clinical & Experimental Medicine, Prague, Czech Republic
| | - Maria Dorobantu
- University of Medicine and Pharmacy "Carol Davila" Bucharest, Romania Emergency Clinical Hospital of Bucharest, Cardiology Dept., Romania
| | - Rodica Nicolescu
- Emergency Clinical Hospital of Bucharest, Cardiology Dept., Romania
| | - Robert Steiner
- Department of Cardiovascular Disease and Intensive Care, Osijek University Hospital, Osijek, Croatia
| | - Vedrana Ivić
- Department of Medical Biology and Genetics, Faculty of Medicine, University of Osijek, Osijek, Croatia
| | - Attila Borbely
- Regional Cooperation for Health, Science and Technology (RECOOP HST) Association, Debrecen, Hungary
| | - Zoltan Papp
- Regional Cooperation for Health, Science and Technology (RECOOP HST) Association, Debrecen, Hungary
| | - Sandor G Vari
- Regional Cooperation for Health, Science and Technology (RECOOP HST) Association, Debrecen, Hungary; International Research and Innovation Management Program, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
68
|
Li YF, Huang X, Li X, Gong R, Yin Y, Nelson J, Gao E, Zhang H, Hoffman NE, Houser SR, Madesh M, Tilley DG, Choi ET, Jiang X, Huang CX, Wang H, Yang XF. Caspase-1 mediates hyperlipidemia-weakened progenitor cell vessel repair. Front Biosci (Landmark Ed) 2016; 21:178-91. [PMID: 26709768 DOI: 10.2741/4383] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Caspase-1 activation senses metabolic danger-associated molecular patterns (DAMPs) and mediates the initiation of inflammation in endothelial cells. Here, we examined whether the caspase-1 pathway is responsible for sensing hyperlipidemia as a DAMP in bone marrow (BM)-derived Stem cell antigen-1 positive (Sca-(1+)) stem/progenitor cells and weakening their angiogenic ability. Using biochemical methods, gene knockout, cell therapy and myocardial infarction (MI) models, we had the following findings: 1) Hyperlipidemia induces caspase-1 activity in mouse Sca-(1+) progenitor cells in vivo; 2) Caspase-1 contributes to hyperlipidemia-induced modulation of vascular cell death-related gene expression in vivo; 3) Injection of Sca-1+ progenitor cells from caspase-1(-/-) mice improves endothelial capillary density in heart and decreases cardiomyocyte death in a mouse model of MI; and 4) Caspase-1(-/-) Sca-(1+) progenitor cell therapy improves mouse cardiac function after MI. Our results provide insight on how hyperlipidemia activates caspase-1 in Sca-(1+) progenitor cells, which subsequently weakens Sca-(1+) progenitor cell repair of vasculature injury. These results demonstrate the therapeutic potential of caspase-1 inhibition in improving progenitor cell therapy for MI.
Collapse
Affiliation(s)
- Ya-Feng Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China
| | - Xiao Huang
- Department of Cardiology, The Second Affiliated Hospital to Nanchang University, Nanchang, JiangXi 330006, China
| | - Xinyuan Li
- Center for Metabolic Disease Research, Department of Pharmacology, Thrombosis Research Center
| | - Ren Gong
- Department of Cardiology, The Second Affiliated Hospital to Nanchang University, Nanchang, JiangXi 330006, China
| | - Ying Yin
- Center for Metabolic Disease Research, Department of Pharmacology, Thrombosis Research Center
| | - Jun Nelson
- Center for Metabolic Disease Research, Department of Pharmacology, Thrombosis Research Center
| | - Erhe Gao
- Center for Translational Medicine, Department of Surgery, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Hongyu Zhang
- Center for Metabolic Disease Research, Department of Pharmacology, Thrombosis Research Center
| | - Nicholas E Hoffman
- Center for Translational Medicine, Department of Surgery, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | - Muniswamy Madesh
- Center for Translational Medicine, Department of Surgery, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Douglas G Tilley
- Center for Translational Medicine, Department of Surgery, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | - Xiaohua Jiang
- Center for Metabolic Disease Research, Department of Pharmacology, Thrombosis Research Center
| | - Cong-Xin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430060, China,
| | - Hong Wang
- Department of Lung Cancer, Affiliated Hospital of Academy of Military Medical Sciences(307 Hospital, PLA), No.8 DongDa Road, FengTai Area, Beijing, P. R. China
| | - Xiao-Feng Yang
- Department of Pharmacology, Cardiovascular Research Center
| |
Collapse
|
69
|
Huang Z, Zhang X, Zhang H, Ye R, Xiong Y, Sun W, Li Y, Liu X. Reduced endothelial progenitor cells in extracranial arterial stenosis but not intracranial arterial stenosis. J Vasc Surg 2015; 62:1539-45. [DOI: 10.1016/j.jvs.2015.07.085] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 07/23/2015] [Indexed: 11/27/2022]
|
70
|
Ye L, Poh KK. Enhancing endothelial progenitor cell for clinical use. World J Stem Cells 2015; 7:894-898. [PMID: 26240678 PMCID: PMC4515434 DOI: 10.4252/wjsc.v7.i6.894] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/03/2015] [Accepted: 05/18/2015] [Indexed: 02/06/2023] Open
Abstract
Circulating endothelial progenitor cells (EPCs) have been demonstrated to correlate negatively with vascular endothelial dysfunction and cardiovascular risk factors. However, translation of basic research into the clinical practice has been limited by the lack of unambiguous and consistent definitions of EPCs and reduced EPC cell number and function in subjects requiring them for clinical use. This article critically reviews the definition of EPCs based on commonly used protocols, their value as a biomarker of cardiovascular risk factor in subjects with cardiovascular disease, and strategies to enhance EPCs for treatment of ischemic diseases.
Collapse
|
71
|
van der Vorst EPC, Döring Y, Weber C. MIF and CXCL12 in Cardiovascular Diseases: Functional Differences and Similarities. Front Immunol 2015; 6:373. [PMID: 26257740 PMCID: PMC4508925 DOI: 10.3389/fimmu.2015.00373] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Accepted: 07/07/2015] [Indexed: 12/11/2022] Open
Abstract
Coronary artery disease (CAD) as part of the cardiovascular diseases is a pathology caused by atherosclerosis, a chronic inflammatory disease of the vessel wall characterized by a massive invasion of lipids and inflammatory cells into the inner vessel layer (intima) leading to the formation of atherosclerotic lesions; their constant growth may cause complications such as flow-limiting stenosis and plaque rupture, the latter triggering vessel occlusion through thrombus formation. Pathophysiology of CAD is complex and over the last years many players have entered the picture. One of the latter being chemokines (small 8-12 kDa cytokines) and their receptors, known to orchestrate cell chemotaxis and arrest. Here, we will focus on the chemokine CXCL12, also known as stromal cell-derived factor 1 (SDF-1) and the chemokine-like function chemokine, macrophage migration-inhibitory factor (MIF). Both are ubiquitously expressed and highly conserved proteins and play an important role in cell homeostasis, recruitment, and arrest through binding to their corresponding chemokine receptors CXCR4 (CXCL12 and MIF), ACKR3 (CXCL12), and CXCR2 (MIF). In addition, MIF also binds to the receptor CD44 and the co-receptor CD74. CXCL12 has mostly been studied for its crucial role in the homing of (hematopoietic) progenitor cells in the bone marrow and their mobilization into the periphery. In contrast to CXCL12, MIF is secreted in response to diverse inflammatory stimuli, and has been associated with a clear pro-inflammatory and pro-atherogenic role in multiple studies of patients and animal models. Ongoing research on CXCL12 points at a protective function of this chemokine in atherosclerotic lesion development. This review will focus on the role of CXCL12 and MIF and their differences and similarities in CAD of high risk patients.
Collapse
Affiliation(s)
- Emiel P C van der Vorst
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich , Munich , Germany
| | - Yvonne Döring
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich , Munich , Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich , Munich , Germany ; German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance , Munich , Germany ; Cardiovascular Research Institute Maastricht (CARIM), Maastricht University , Maastricht , Netherlands
| |
Collapse
|
72
|
Abstract
Endothelial progenitor cells (EPCs) play a critical role in maintenance of the endothelial integrity and vascular homeostasis, as well as in neovascularization. Dysfunctional EPCs are believed to contribute to the endothelial dysfunction and are closely related to the development of various cardiovascular diseases, such as hypertension, hyperlipidemia, and stroke. However, the underlying mechanisms of EPC dysfunction are complicated and remain largely elusive. Recent studies have demonstrated that reactive oxygen species (ROS) are key factors that involve in modulation of stem and progenitor cell function under various physiologic and pathologic conditions. It has been shown that NADPH oxidase (NOX)-derived ROS are the major sources of ROS in cardiovascular system. Accumulating evidence suggests that NOX-mediated oxidative stress can modulate EPC bioactivities, such as mobilization, migration, and neovascularization, and that inhibition of NOX has been shown to improve EPC functions. This review summarized recent progress in the studies on the correlation between NOX-mediated EPC dysfunction and cardiovascular diseases.
Collapse
|
73
|
Alexandru N, Andrei E, Dragan E, Georgescu A. Interaction of platelets with endothelial progenitor cells in the experimental atherosclerosis: Role of transplanted endothelial progenitor cells and platelet microparticles. Biol Cell 2015; 107:189-204. [DOI: 10.1111/boc.201400071] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 03/06/2015] [Indexed: 11/29/2022]
Affiliation(s)
- Nicoleta Alexandru
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| | - Eugen Andrei
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| | - Emanuel Dragan
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| | - Adriana Georgescu
- Institute of Cellular Biology and Pathology ‘Nicolae Simionescu’ of the Romanian Academy; Bucharest Romania
| |
Collapse
|
74
|
Gong X, Shao L, Fu YM, Zou Y. Effects of olmesartan on endothelial progenitor cell mobilization and function in carotid atherosclerosis. Med Sci Monit 2015; 21:1189-93. [PMID: 25913171 PMCID: PMC4422112 DOI: 10.12659/msm.892996] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Olmesartan is a type of angiotensin II receptor inhibitor that can reduce the incidence of cardiovascular events. However, its role in the function of endothelial progenitor cells in atherosclerosis patients is still unclear. Our study aimed to explore the effects and mechanism of olmesartan on endothelial progenitor cell mobilization and function in carotid atherosclerosis. MATERIAL/METHODS Forty carotid atherosclerosis patients were enrolled. Patients were administrated olmesartan 20 mg/day for 3 months. Flow cytometry was used for counting circulating endothelial progenitor cells; colorimetric method was used to measure the serum levels of endothelial nitric oxide synthase and nitric oxide. Cell migration, adhesion, and proliferation capacity, and related signaling pathway were also analyzed. Spearman rank correlation analysis was used to investigate the influence of olmesartan on endothelial progenitor cells and clinical characteristics (e.g., sex, age, blood pressure). RESULTS Compared with the control group, the number of circulating endothelial progenitor cells was significantly decreased. Olmesartan can increase circulating endothelial progenitor cells number and the serum levels of eNOS and NO. Furthermore, it can improve cell migration, adhesion, and proliferation capacities. Spearman rank correlation analysis showed there is no relationship between olmesartan promotion effects on endothelial progenitor cell mobilization and the clinical characteristics (P>0.05). P-eNOS and P-Akt expression can be unregulated by RNH-6270 treatment and blocked by LY294002. CONCLUSIONS Olmesartan can effectively promote the endothelial progenitor cells mobilization and improve their function in patients with carotid atherosclerosis, independent of basic characteristics. This process relies on the PI3K/Akt/eNOS signaling pathway.
Collapse
Affiliation(s)
- Xin Gong
- Department of Health Care, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Li Shao
- Department of Health Care, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Yi-Min Fu
- Department of Health Care, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| | - Yong Zou
- Department of Health Care, Yantai Yuhuangding Hospital, Yantai, Shandong, China (mainland)
| |
Collapse
|
75
|
Martí-Fàbregas J, Delgado-Mederos R, Crespo J, Peña E, Marín R, Jiménez-Xarrié E, Fernández-Arcos A, Pérez-Pérez J, Martínez-Domeño A, Camps-Renom P, Prats-Sánchez L, Casoni F, Badimon L. Circulating endothelial progenitor cells and the risk of vascular events after ischemic stroke. PLoS One 2015; 10:e0124895. [PMID: 25874380 PMCID: PMC4395144 DOI: 10.1371/journal.pone.0124895] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2014] [Accepted: 03/07/2015] [Indexed: 11/18/2022] Open
Abstract
Background and Purpose We evaluated the hypothesis that the number of circulating EPC could be associated with the risk of stroke recurrence (SR) or vascular events (VE) after an ischemic stroke. Methods We studied prospectively consecutive patients with cerebral infarction within the first 48 hours after the onset. We recorded demographic factors, vascular risk factors, previous Rankin scale (RS) score, and etiology. We analyzed EPC counts by flow cytometry in blood collected at day 7 and defined EPC as CD34+/CD133+/KDR+ cells. Mean follow-up was 29.3 ± 16 months. We evaluated SR as well as VE. Patients were classified as to the presence or absence of EPC in the circulation (either EPC+ or EPC-). Bivariate analyses, Kaplan-Meier survival curves and Cox regression models were used. Results We included 121 patients (mean age 70.1±12.6 years; 65% were men). The percentage of EPC+ patients was 47.1%. SR occurred in 12 (9.9%) and VE in 18 (14.9%) patients. SR was associated significantly with a worse prior RS score, previous stroke and etiology, but not with EPC count. VE were associated significantly with EPC-, worse prior RS score, previous stroke, high age, peripheral artery disease and etiology. Cox regression model showed that EPC- (HR 7.07, p=0.003), age (HR 1.08, p=0.004) and a worse prior RS score (HR 5.8, p=0.004) were associated significantly with an increased risk of VE. Conclusions The absence of circulating EPC is not associated with the risk of stroke recurrence, but is associated with an increased risk of future vascular events.
Collapse
Affiliation(s)
- Joan Martí-Fàbregas
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
- * E-mail:
| | - Raquel Delgado-Mederos
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Javier Crespo
- Cardiovascular Research Center, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Esther Peña
- Cardiovascular Research Center, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Rebeca Marín
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Elena Jiménez-Xarrié
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Ana Fernández-Arcos
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Jesús Pérez-Pérez
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | | | - Pol Camps-Renom
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Luís Prats-Sánchez
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Francesca Casoni
- Department of Neurology, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Research Center, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
| |
Collapse
|
76
|
Li YF, Ren LN, Guo G, Cannella LA, Chernaya V, Samuel S, Liu SX, Wang H, Yang XF. Endothelial progenitor cells in ischemic stroke: an exploration from hypothesis to therapy. J Hematol Oncol 2015; 8:33. [PMID: 25888494 PMCID: PMC4446087 DOI: 10.1186/s13045-015-0130-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 03/24/2015] [Indexed: 12/29/2022] Open
Abstract
As the population ages and lifestyles change in concordance, the number of patients suffering from ischemic stroke and its associated disabilities is increasing. Studies on determining the relationship between endothelial progenitor cells (EPCs) and ischemic stroke have become a new hot spot and have reported that EPCs may protect the brain against ischemic injury, promote neurovascular repair, and improve long-term neurobehavioral outcomes. More importantly, they introduce a new perspective for prognosis assessment and therapy of ischemic stroke. However, EPCs’ origin, function, influence factors, injury repair mechanisms, and cell-based therapy strategies remain controversial. Particularly, research conducted to date has less clinical studies than pre-clinical experiments on animals. In this review, we summarized and analyzed the current understanding of basic characteristics, influence factors, functions, therapeutic strategies, and disadvantages of EPCs as well as the regulation of inflammatory factors involved in the function and survival of EPCs after ischemic stroke. Identifying potential therapeutic effects of EPCs in ischemic stroke will be a challenging but an incredibly important breakthrough in neurology, which may bring promise for patients with ischemic stroke.
Collapse
Affiliation(s)
- Ya-Feng Li
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA. .,Department of Nephrology and Hemodialysis Center, The Second Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China.
| | - Li-Na Ren
- The First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China.
| | - Geng Guo
- Department of Neurosurgery, The First Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China.
| | - Lee Anne Cannella
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA.
| | - Valeria Chernaya
- Department of Biology, College of Science and Technology, Temple University, 1801 N. Broad St., Philadelphia, PA, 19122, USA.
| | - Sonia Samuel
- Department of Biology, College of Science and Technology, Temple University, 1801 N. Broad St., Philadelphia, PA, 19122, USA.
| | - Su-Xuan Liu
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA.
| | - Hong Wang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA.
| | - Xiao-Feng Yang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Temple University School of Medicine, Philadelphia, PA, 19140, USA.
| |
Collapse
|
77
|
Wang LY, Zhang JH, Yu J, Yang J, Deng MY, Kang HL, Huang L. Reduction of Store-Operated Ca(2+) Entry Correlates with Endothelial Progenitor Cell Dysfunction in Atherosclerotic Mice. Stem Cells Dev 2015; 24:1582-90. [PMID: 25753987 DOI: 10.1089/scd.2014.0538] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The dysfunction of endothelial progenitor cells (EPCs) has been shown to prevent endothelial repair during the development of atherosclerosis (AS). Previous studies have revealed that store-operated calcium entry (SOCE) is an important factor in regulating EPC functions. However, whether this is also the mechanism in AS has not been elucidated. Therefore, we evaluated the role of SOCE in EPCs isolated from an atherosclerotic mouse model. Atheromatous plaques were more frequent in the aortas of ApoE(-/-) mice fed a high-fat diet for 16 weeks compared with controls, and the proliferative and migratory activities of atherosclerotic EPCs were significantly decreased. Accordingly, SOCE amplitude, as well as spontaneous or VEGF-induced Ca(2+) oscillations, decreased in atherosclerotic EPCs. These results may be associated with the downregulated expression of Stim1, Orai1, and TRPC1, which are major mediators of SOCE. In addition, eNOS expression and phosphorylation at Ser(1177), which are critical regulators of EPC function, were markedly reduced in the atherosclerotic EPCs. The impairment of eNOS activity could also be induced by using an SOCE inhibitor or by Stim1 gene silencing, indicating a link between the activities of eNOS and SOCE in AS. Furthermore, decreased SOCE function inhibited EPC proliferation and migration in vitro. In conclusion, our results showed that the reduction of SOCE induced EPC dysfunction during AS, potentially through downregulation of store-operated calcium channel (SOCC) components and impaired eNOS activity. Approaches aimed at reestablishing SOCE activity may thus improve the function of EPCs during AS.
Collapse
Affiliation(s)
- Lian-You Wang
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University , Chongqing, People's Republic of China
| | - Ji-Hang Zhang
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University , Chongqing, People's Republic of China
| | - Jie Yu
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University , Chongqing, People's Republic of China
| | - Jie Yang
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University , Chongqing, People's Republic of China
| | - Meng-Yang Deng
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University , Chongqing, People's Republic of China
| | - Hua-Li Kang
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University , Chongqing, People's Republic of China
| | - Lan Huang
- Institute of Cardiovascular Diseases of PLA, Xinqiao Hospital, Third Military Medical University , Chongqing, People's Republic of China
| |
Collapse
|
78
|
Balestrieri ML, Rizzo MR, Barbieri M, Paolisso P, D'Onofrio N, Giovane A, Siniscalchi M, Minicucci F, Sardu C, D'Andrea D, Mauro C, Ferraraccio F, Servillo L, Chirico F, Caiazzo P, Paolisso G, Marfella R. Sirtuin 6 expression and inflammatory activity in diabetic atherosclerotic plaques: effects of incretin treatment. Diabetes 2015; 64:1395-406. [PMID: 25325735 DOI: 10.2337/db14-1149] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The role of sirtuin 6 (SIRT6) in atherosclerotic progression of diabetic patients is unknown. We evaluated SIRT6 expression and the effect of incretin-based therapies in carotid plaques of asymptomatic diabetic and nondiabetic patients. Plaques were obtained from 52 type 2 diabetic and 30 nondiabetic patients undergoing carotid endarterectomy. Twenty-two diabetic patients were treated with drugs that work on the incretin system, GLP-1 receptor agonists, and dipeptidyl peptidase-4 inhibitors for 26 ± 8 months before undergoing the endarterectomy. Compared with nondiabetic plaques, diabetic plaques had more inflammation and oxidative stress, along with a lesser SIRT6 expression and collagen content. Compared with non-GLP-1 therapy-treated plaques, GLP-1 therapy-treated plaques presented greater SIRT6 expression and collagen content, and less inflammation and oxidative stress, indicating a more stable plaque phenotype. These results were supported by in vitro observations on endothelial progenitor cells (EPCs) and endothelial cells (ECs). Indeed, both EPCs and ECs treated with high glucose (25 mmol/L) in the presence of GLP-1 (100 nmol/L liraglutide) presented a greater SIRT6 and lower nuclear factor-κB expression compared with cells treated only with high glucose. These findings establish the involvement of SIRT6 in the inflammatory pathways of diabetic atherosclerotic lesions and suggest its possible positive modulation by incretin, the effect of which is associated with morphological and compositional characteristics of a potential stable plaque phenotype.
Collapse
Affiliation(s)
- Maria Luisa Balestrieri
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Maria Rosaria Rizzo
- Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Second University of Naples, Naples, Italy
| | - Michelangela Barbieri
- Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Second University of Naples, Naples, Italy
| | - Pasquale Paolisso
- Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Second University of Naples, Naples, Italy
| | - Nunzia D'Onofrio
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Alfonso Giovane
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | | | - Fabio Minicucci
- Department of Cardiology, Cardarelli Hospital, Naples, Italy
| | - Celestino Sardu
- Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Second University of Naples, Naples, Italy
| | - Davide D'Andrea
- Department of Cardiology, Cardarelli Hospital, Naples, Italy
| | - Ciro Mauro
- Department of Cardiology, Cardarelli Hospital, Naples, Italy
| | - Franca Ferraraccio
- Department of Clinical, Public and Preventive Medicine, Second University of Naples, Naples, Italy
| | - Luigi Servillo
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Fabio Chirico
- Department of Neurosurgery, Cardarelli Hospital, Naples, Italy
| | | | - Giuseppe Paolisso
- Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Second University of Naples, Naples, Italy
| | - Raffaele Marfella
- Department of Medical, Surgical, Neurological, Aging and Metabolic Sciences, Second University of Naples, Naples, Italy
| |
Collapse
|
79
|
Steinmetz M, Lucanus E, Zimmer S, Nickenig G, Werner N. Mobilization of sca1/flk-1 positive endothelial progenitor cells declines in apolipoprotein E-deficient mice with a high-fat diet. J Cardiol 2015; 66:532-8. [PMID: 25818640 DOI: 10.1016/j.jjcc.2015.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 01/13/2015] [Accepted: 02/05/2015] [Indexed: 01/14/2023]
Abstract
BACKGROUND Atherosclerosis features a deterioration of the endothelial layer in all stages. Restoration of the endothelium is associated with circulating stem cell antigen 1 (sca1) and vascular endothelial growth factor receptor type 2 (flk-1) positive endothelial progenitor cells (EPCs). We investigated whether EPC production and/or a mobilization from bone marrow are reduced in severe atherosclerosis. METHODS AND RESULTS EPCs in peripheral blood were diminished in ApoE-/- mice with high-fat diet (HFD) whereas bone marrow levels of these cells were not significantly altered compared to controls. In situ perfusion of the hind limbs demonstrated that EPC mobilization was reduced compared to ApoE-/- mice with normal chow, although increased plasma stromal cell-derived factor (SDF) 1α and responsivity suggested a mobilizing stimulus. The proliferation of sca1/flk-1 positive cells showed no functional impairment. EPCs could not only be significantly mobilized from the bone marrow through the application of granulocyte colony stimulating factor (GCSF), but also led by trend to a depletion of the bone marrow pool. GCSF levels in plasma were equal in ApoE-/- mice with normal chow or HFD, which excluded a decline in GCSF production. CONCLUSION The capability of the bone marrow pool to adapt the proliferation and mobilization of sca1/flk-1 positive EPCs seems overstrained in ApoE-/- mice with a HFD.
Collapse
Affiliation(s)
- Martin Steinmetz
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany; Institut National de la Santé et de la Recherche Médicale (INSERM), Unit 970, Paris Cardiovascular Research Center, Paris, France.
| | - Eva Lucanus
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Sebastian Zimmer
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Georg Nickenig
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| | - Nikos Werner
- Medizinische Klinik und Poliklinik II, Kardiologie/Angiologie/Pulmologie/Internistische Intensivmedizin, Universitätsklinikum Bonn, Bonn, Germany
| |
Collapse
|
80
|
Theodoridis K, Tudorache I, Calistru A, Cebotari S, Meyer T, Sarikouch S, Bara C, Brehm R, Haverich A, Hilfiker A. Successful matrix guided tissue regeneration of decellularized pulmonary heart valve allografts in elderly sheep. Biomaterials 2015; 52:221-8. [PMID: 25818428 DOI: 10.1016/j.biomaterials.2015.02.023] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 01/25/2015] [Accepted: 02/01/2015] [Indexed: 11/25/2022]
Abstract
In vivo repopulation of decellularized allografts with recipient cells leads to a positive remodeling of the graft matrix in juvenile sheep. In light of the increasing number of heart valve replacements among older patients (>65 years), this study focused on the potential for matrix-guided tissue regeneration in elderly sheep. Pulmonary valve replacement was performed in seven-year old sheep using decellularized (DV), decellularized and CCN1-coated (RV), or decellularized and in vitro reendothelialized pulmonary allografts (REV) (n=6, each group). CCN1 coating was applied to support re-endothelialization. In vitro re-endothelialization was conducted with endothelial-like cells derived from peripheral blood. Echocardiograms of all grafts showed adequate graft function after implantation and at explantation 3 or 6 months later. All explants were macroscopically free of thrombi at explantation, and revealed repopulation of the allografts on the adventitial side of valvular walls and proximal in the cusps. Engrafted cells expressed vimentin, sm α-actin, and myosin heavy chain 2, while luminal cell lining was positive for vWF and eNOS. Cellular repopulation of valvular matrix demonstrates the capacity for matrix-guided regeneration even in elderly sheep but is not improved by in vitro endothelialization, confirming the suitability of decellularized matrix for heart valve replacement in older individuals.
Collapse
Affiliation(s)
- Karolina Theodoridis
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany; Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Igor Tudorache
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany; Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Alexandru Calistru
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany; Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Serghei Cebotari
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany; Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Tanja Meyer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany; Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Samir Sarikouch
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Christoph Bara
- Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Ralph Brehm
- Institute of Anatomy, University of Veterinary Medicine Hannover, Germany
| | - Axel Haverich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany; Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Andres Hilfiker
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Hannover Medical School, Hannover, Germany; Department of Cardiothoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
81
|
Lu C, Zhang X, Zhang D, Pei E, Xu J, Tang T, Ye M, Uzan G, Zhi K, Li M, Zuo K. Short Time Tripterine Treatment Enhances Endothelial Progenitor Cell Function via Heat Shock Protein 32. J Cell Physiol 2015; 230:1139-47. [PMID: 25336054 DOI: 10.1002/jcp.24849] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 10/14/2014] [Indexed: 01/02/2023]
Affiliation(s)
- Chenhui Lu
- Department of Interventional Radiology; Shanghai Tenth People's Hospital; Tongji University; Shanghai China
- Shanghai Gong Li Hospital; Shanghai China
| | - Xiaoping Zhang
- Department of Interventional Radiology; Shanghai Tenth People's Hospital; Tongji University; Shanghai China
- Institute of Medical Intervention Engineering; Tongji University; Shanghai China
| | | | - Erli Pei
- Department of Interventional Radiology; Shanghai Tenth People's Hospital; Tongji University; Shanghai China
| | - Jichong Xu
- Department of Interventional Radiology; Shanghai Tenth People's Hospital; Tongji University; Shanghai China
| | - Tao Tang
- Department of Interventional Radiology; Shanghai Tenth People's Hospital; Tongji University; Shanghai China
| | - Meng Ye
- Department of Interventional Radiology; Shanghai Tenth People's Hospital; Tongji University; Shanghai China
| | - Georges Uzan
- Unite de Recherche INSERM 972; Villejuif Cedex France
| | - Kangkang Zhi
- Department of Vascular and Endovascular Surgery; Changzheng Hospital; Shanghai China
| | - Maoquan Li
- Department of Interventional Radiology; Shanghai Tenth People's Hospital; Tongji University; Shanghai China
- Institute of Medical Intervention Engineering; Tongji University; Shanghai China
| | - Keqiang Zuo
- Shanghai Tenth People's Hospital; Tongji University School of Medicine; Shanghai China
| |
Collapse
|
82
|
Cui Y, Xie X, Jia F, He J, Li Z, Fu M, Hao H, Liu Y, Liu JZ, Cowan PJ, Zhu H, Sun Q, Liu Z. Ambient fine particulate matter induces apoptosis of endothelial progenitor cells through reactive oxygen species formation. Cell Physiol Biochem 2015; 35:353-63. [PMID: 25591776 DOI: 10.1159/000369701] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2014] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND/AIMS Bone marrow (BM)-derived endothelial progenitor cells (EPCs) play a critical role in angiogenesis and vascular repair. Some environmental insults, like fine particulate matter (PM) exposure, significantly impair cardiovascular functions. However, the mechanisms for PM-induced adverse effects on cardiovascular system remain largely unknown. The present research was to study the detrimental effects of PM on EPCs and explore the potential mechanisms. METHODS PM was intranasal-distilled into male C57BL/6 mice for one month. Flow cytometry was used to measure the number of EPCs, apoptosis level of circulating EPCs and intracellular reactive oxygen species (ROS) formation. Serum TNF-α and IL-1β were measured using ELISA. To determine the role of PM-induced ROS in EPC apoptosis, PM was co-administrated with the antioxidant N-acetylcysteine (NAC) in wild type mice or used in a triple transgenic mouse line (TG) with overexpression of antioxidant enzyme network (AON) composed of superoxide dismutase (SOD)1, SOD3, and glutathione peroxidase (Gpx-1) with decreased in vivo ROS production. RESULTS PM treatment significantly decreased circulating EPC population, promoted apoptosis of EPCs in association with increased ROS production and serum TNF-α and IL-1β levels, which could be effectively reversed by either NAC treatment or overexpression of AON. CONCLUSION PM exposure significantly decreased circulating EPCs population due to increased apoptosis via ROS formation in mice.
Collapse
Affiliation(s)
- Yuqi Cui
- Dorothy M. Davis Heart and Lung Research Institute, Division of Cardiovascular Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Chen Y, Lu B, Wang J, Chen S, Lin Z, Ma X, Liu Y, Zhao B, Chen Y. Circulating CD133+ CD34+ progenitor cells and plasma stromal-derived factor-1alpha: predictive role in ischemic stroke patients. J Stroke Cerebrovasc Dis 2014; 24:319-26. [PMID: 25444027 DOI: 10.1016/j.jstrokecerebrovasdis.2014.08.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 08/20/2014] [Accepted: 08/25/2014] [Indexed: 11/28/2022] Open
Abstract
Circulating progenitor cells and stromal-derived factor-1alpha (SDF-1α) have been suggested to participate in tissue repair after ischemic injury. However, the predictive role of circulating CD133+ CD34+ progenitors and plasma SDF-1α in ischemic stroke (IS) patients remains unknown. In this study, we recruited 95 acute IS patients, 40 at-risk subjects, and 30 normal subjects. The National Institutes of Health Stroke Scale (NIHSS), infarct volume, and carotid intima-media thickness (IMT) were determined at day 1 and the modified Rankin scale (mRS) of functional outcome was assessed at day 21. The levels of circulating CD133+ CD34+ cells and plasma SDF-1α were determined by flow cytometry and enzyme-linked immunosorbent assay, respectively. Our data showed that: (1) the levels of CD133+ CD34+ cells were lower in at-risk subjects and IS patients at admission (day 1) when compared with normal controls; (2) the day 1 level of CD133+ CD34+ cells varied in IS subgroups and inversely correlated with NIHSS and carotid IMT and the level of SDF-1α inversely correlated with NIHSS and infarct volume; (3) the increment rates of circulating CD133+ CD34+ cells and plasma SDF-1α within the first week were correlated; and (4) patients with a higher level of CD133+ CD34+ cells at day 7 had a low mRS. The increased rate of CD133+ CD34+ cells in the first week was inversely associated with mRS. In conclusion, our findings demonstrate that the circulating CD133+ CD34+ progenitor cells and plasma SDF-1α can be used as predictive parameters for IS severity and outcome.
Collapse
Affiliation(s)
- Yusen Chen
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Bing Lu
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Jinju Wang
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio
| | - Shuzhen Chen
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio
| | - Zhijun Lin
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Xiaotang Ma
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Yajing Liu
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China
| | - Bin Zhao
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China.
| | - Yanfang Chen
- Clinical Research Center and Department of Neurology, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong, China; Department of Pharmacology & Toxicology, Boonshoft School of Medicine, Wright State University, Dayton, Ohio.
| |
Collapse
|
84
|
Han Y, Tao J, Gomer A, Ramirez-Bergeron DL. Loss of endothelial-ARNT in adult mice contributes to dampened circulating proangiogenic cells and delayed wound healing. Vasc Med 2014; 19:429-41. [PMID: 25398385 DOI: 10.1177/1358863x14559588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The recruitment and homing of circulating bone marrow-derived cells include endothelial progenitor cells (EPCs) that are critical to neovascularization and tissue regeneration of various vascular pathologies. We report here that conditional inactivation of hypoxia-inducible factor's (HIF) transcriptional activity in the endothelium of adult mice (Arnt(ΔiEC) mice) results in a disturbance of infiltrating cells, a hallmark of neoangiogenesis, during the early phases of wound healing. Cutaneous biopsy punches show distinct migration of CD31(+) cells into wounds of control mice by 36 hours. However, a significant decline in numbers of infiltrating cells with immature vascular markers, as well as decreased transcript levels of genes associated with their expression and recruitment, were identified in wounds of Arnt(ΔiEC) mice. Matrigel plug assays further confirmed neoangiogenic deficiencies alongside a reduction in numbers of proangiogenic progenitor cells from bone marrow and peripheral blood samples of recombinant vascular endothelial growth factor-treated Arnt(ΔiEC) mice. In addition to HIF's autocrine requirements in endothelial cells, our data implicate that extrinsic microenvironmental cues provided by endothelial HIF are pivotal for early migration of proangiogenic cells, including those involved in wound healing.
Collapse
Affiliation(s)
- Yu Han
- Case Cardiovascular Research Institute and University Hospitals Harrington Heart & Vascular Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA University of Rochester, School of Medicine and Dentistry, Rochester, NY, USA
| | - Jiayi Tao
- Case Cardiovascular Research Institute and University Hospitals Harrington Heart & Vascular Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Alla Gomer
- Case Cardiovascular Research Institute and University Hospitals Harrington Heart & Vascular Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Diana L Ramirez-Bergeron
- Case Cardiovascular Research Institute and University Hospitals Harrington Heart & Vascular Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
85
|
Ross MD, Wekesa AL, Phelan JP, Harrison M. Resistance exercise increases endothelial progenitor cells and angiogenic factors. Med Sci Sports Exerc 2014; 46:16-23. [PMID: 24346188 DOI: 10.1249/mss.0b013e3182a142da] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Bone marrow-derived endothelial progenitor cells (EPC) are involved in vascular growth and repair. They increase in the circulation after a single bout of aerobic exercise, potentially related to muscle ischemia. Muscular endurance resistance exercise (MERE) bouts also have the potential to induce muscle ischemia if appropriately structured. PURPOSE The objective of this study is to determine the influence of a single bout of MERE on circulating EPC and related angiogenic factors. METHODS Thirteen trained men age 22.4 ± 0.5 yr (mean ± SEM) performed a bout of MERE consisting of three sets of six exercises at participants' 15-repetition maximum without resting between repetitions or exercises. The MERE bout duration was 12.1 ± 0.6 min. Blood lactate and HR were 11.9 ± 0.9 mmol·L and 142 ± 5 bpm, respectively, at the end of MERE. Blood was sampled preexercise and at 10 min, 2 h, and 24 h postexercise. RESULTS Circulating EPC and serum concentrations of vascular endothelial growth factors (VEGF-A, VEGF-C, and VEGF-D), granulocyte colony stimulating factor, soluble Tie-2, soluble fms-like tyrosine kinase-1, and matrix metalloproteinases (MMP-1, MMP-2, MMP-3, MMP-9, and MMP-9) were higher (P < 0.05) in the postexercise period. Circulating EPC levels were unchanged at 10 min postexercise but higher at 2 h postexercise (P < 0.05). The concentration of most angiogenic factors and metalloproteinases were higher at 10 min postexercise (VEGF-A, +38%; VEGF-C, +40%; VEGF-D, +9%; soluble Tie-2, +15%; soluble fms-like tyrosine kinase-1, +24%; MMP-1, +62%; MMP-2, +3%; MMP-3, +54%; and MMP-9, +45%; all P < 0.05). Soluble E-selectin was lower (P < 0.05) at 2 and 24 h postexercise, with endothelial microparticles and thrombomodulin unchanged. CONCLUSIONS Short intense bouts of MERE can trigger increases in circulating EPC and related angiogenic factors, potentially contributing to vascular adaptation and vasculoprotection.
Collapse
Affiliation(s)
- Mark D Ross
- 1Department of Health, Sport and Exercise Science, Waterford Institute of Technology, Waterford, IRELAND; and 2School of Life, Sport and Social Sciences, Edinburgh Napier University, Edinburgh, Scotland, UNITED KINGDOM
| | | | | | | |
Collapse
|
86
|
The effects of endothelial progenitor cells on rat atherosclerosis. Biotechnol Appl Biochem 2014; 62:186-92. [DOI: 10.1002/bab.1254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 05/23/2014] [Indexed: 12/13/2022]
|
87
|
Adly AAM, El-Sherif NH, Ismail EAR, El-Zaher YA, Farouk A, El-Refaey AM, Wahba MS. Vascular Dysfunction in Patients With Young β-Thalassemia. Clin Appl Thromb Hemost 2014; 21:733-44. [DOI: 10.1177/1076029614541515] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
We aimed to study the endothelial dysfunction among children and adolescents with transfusion-dependent β-thalassemia using von Willebrand factor antigen (VWF:Ag) and flow cytometric analysis of circulating CD144+ endothelial microparticles (EMPs) and endothelial progenitor cells (CD34+VEGFR2+) and assess their relation to iron overload, erythropoietin and chelation therapy as well as echocardiographic parameters and carotid intima–media thickness. The VWF:Ag, EMPs, and CD34+VEGFR2+ cells were significantly higher among patients with β-thalassemia than controls ( P < .001). The type of chelation and patients’ compliance did not influence the results. No significant correlations were found between the studied vascular markers. Patients with evident heart disease had higher VWF: Ag, EMPs, and CD34+VEGFR2+ cells than those without. Carotid intima–media thickness was increased among patients but not correlated with vascular markers. We suggest that procoagulant EMPs and VWF: Ag are involved in cardiovascular complications in patients with young β-thalassemia. CD34+VEGFR2+ cells were further increased in response to tissue injury contributing to reendothelialization and neovascularization.
Collapse
Affiliation(s)
| | | | | | - Yosra Abd El-Zaher
- Radiology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Amal Farouk
- Clinical Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | - Mohammed Samy Wahba
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
88
|
Döring Y, Pawig L, Weber C, Noels H. The CXCL12/CXCR4 chemokine ligand/receptor axis in cardiovascular disease. Front Physiol 2014; 5:212. [PMID: 24966838 PMCID: PMC4052746 DOI: 10.3389/fphys.2014.00212] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/15/2014] [Indexed: 12/18/2022] Open
Abstract
The chemokine receptor CXCR4 and its ligand CXCL12 play an important homeostatic function by mediating the homing of progenitor cells in the bone marrow and regulating their mobilization into peripheral tissues upon injury or stress. Although the CXCL12/CXCR4 interaction has long been regarded as a monogamous relation, the identification of the pro-inflammatory chemokine macrophage migration inhibitory factor (MIF) as an important second ligand for CXCR4, and of CXCR7 as an alternative receptor for CXCL12, has undermined this interpretation and has considerably complicated the understanding of CXCL12/CXCR4 signaling and associated biological functions. This review aims to provide insight into the current concept of the CXCL12/CXCR4 axis in myocardial infarction (MI) and its underlying pathologies such as atherosclerosis and injury-induced vascular restenosis. It will discuss main findings from in vitro studies, animal experiments and large-scale genome-wide association studies. The importance of the CXCL12/CXCR4 axis in progenitor cell homing and mobilization will be addressed, as will be the function of CXCR4 in different cell types involved in atherosclerosis. Finally, a potential translation of current knowledge on CXCR4 into future therapeutical application will be discussed.
Collapse
Affiliation(s)
- Yvonne Döring
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Germany
| | - Lukas Pawig
- Institute for Molecular Cardiovascular Research, RWTH Aachen University Aachen, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Germany ; German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance Munich, Germany ; Cardiovascular Research Institute Maastricht, University of Maastricht Maastricht, Netherlands
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research, RWTH Aachen University Aachen, Germany
| |
Collapse
|
89
|
Liu Z, Ding X, Fang F, Wang R, Chen Y, Ma Y, Zhang G, Kang X. Higher numbers of circulating endothelial progenitor cells in stroke patients with intracranial arterial stenosis. BMC Neurol 2013; 13:161. [PMID: 24188156 PMCID: PMC3829095 DOI: 10.1186/1471-2377-13-161] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 10/28/2013] [Indexed: 12/15/2022] Open
Abstract
Background Bone marrow-derived endothelial stem cells participate in vascular repairs. Numbers of circulating endothelial progenitor cells (cEPCs) are associated with atherosclerosis. Fibrinogen plays a key role in atherosclerosis. Objective was to assess if cEPC counts were associated with atherosclerotic intracranial artery stenosis (IAS). Methods Three hundred subjects (108 patients with stroke and IAS (IAS), 120 control patients with stroke without IAS (CP), and 72 healthy controls (HC)) were retrospectively analyzed. cEPCs were identified and counted by flow cytometry using CD34, CD133 and KDR. Plasma fibrinogen was measured by immunoturbidimetry. cEPC counts were compared between the three groups. Results cEPC numbers were significantly higher in IAS (0.059 ± 0.031%) than in CP (0.026 ± 0.012%) (P < 0.001) and HC (0.021 ± 0.011%) (P < 0.001), but without difference between CP and HC (P = 0.401). Multiple logistic regression analysis showed that cEPC levels (OR 3.31, 95%CI 1.26-8.87, P = 0.025; IAS vs. CP) were independent markers of IAS after adjustment for hypertension, diabetes and smoking. No significant correlation between cEPC counts and plasma fibrinogen levels was observed (P > 0.05). Conclusion cEPC numbers were associated with degrees of IAS. This measurement may be useful for non-invasive evaluation of atherosclerotic IAS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xixiong Kang
- The Centre for Laboratory Diagnosis, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
90
|
Effect of therapeutic inhibition of TNF on circulating endothelial progenitor cells in patients with rheumatoid arthritis. Mediators Inflamm 2013; 2013:537539. [PMID: 24222719 PMCID: PMC3810060 DOI: 10.1155/2013/537539] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 07/19/2013] [Accepted: 08/05/2013] [Indexed: 01/22/2023] Open
Abstract
Endothelial dysfunction has been detected in RA patients and seems to be reversed by control of inflammation. Low circulating endothelial progenitor cells (EPCs) have been described in many conditions associated with increased cardiovascular risk, including RA. The aim of this study was to investigate the effect of inhibition of TNF on EPCs in RA patients.
Seventeen patients with moderate-severe RA and 12 sex and age-matched controls were evaluated. Endothelial biomarkers were tested at baseline and after 3 months. EPCs were identified from peripheral blood mononuclear cells by cytofluorimetry using anti-CD34 and anti-vascular endothelial growth factor-receptor 2. Asymmetric dimethylarginine (ADMA) was tested by ELISA and flow-mediated dilatation (FMD) by ultrasonography. Circulating EPCs were significantly lower in RA patients than in controls (P = 0.001). After 3 months EPCs increased significantly (P = 0.0006) while ADMA levels significantly decreased (P = 0.001). An inverse correlation between mean increase in EPCs number and mean decrease of DAS28 after treatment was observed (r = −0.56, P = 0.04). EPCs inversely correlated with ADMA (r = −0.41, P = 0.022). No improvement of FMD was detected. Short-term treatment with anti-TNF was able to increase circulating EPCs concurrently with a proportional decrease of disease activity suggesting that therapeutic intervention aimed at suppressing the inflammatory process might positively affect the endothelial function.
Collapse
|
91
|
LOX-1, OxLDL, and atherosclerosis. Mediators Inflamm 2013; 2013:152786. [PMID: 23935243 PMCID: PMC3723318 DOI: 10.1155/2013/152786] [Citation(s) in RCA: 526] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 06/16/2013] [Indexed: 01/07/2023] Open
Abstract
Oxidized low-density lipoprotein (OxLDL) contributes to the atherosclerotic plaque formation and progression by several mechanisms, including the induction of endothelial cell activation and dysfunction, macrophage foam cell formation, and smooth muscle cell migration and proliferation. Vascular wall cells express on their surface several scavenger receptors that mediate the cellular effects of OxLDL. The lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is the main OxLDL receptor of endothelial cells, and it is expressed also in macrophages and smooth muscle cells. LOX-1 is almost undetectable under physiological conditions, but it is upregulated following the exposure to several proinflammatory and proatherogenic stimuli and can be detected in animal and human atherosclerotic lesions. The key contribution of LOX-1 to the atherogenic process has been confirmed in animal models; LOX-1 knockout mice exhibit reduced intima thickness and inflammation and increased expression of protective factors; on the contrary, LOX-1 overexpressing mice present an accelerated atherosclerotic lesion formation which is associated with increased inflammation. In humans, LOX-1 gene polymorphisms were associated with increased susceptibility to myocardial infarction. Inhibition of the LOX-1 receptor with chemicals or antisense nucleotides is currently being investigated and represents an emerging approach for controlling OxLDL-LOX-1 mediated proatherogenic effects.
Collapse
|
92
|
Chimowitz MI. The Feinberg Award Lecture 2013: treatment of intracranial atherosclerosis: learning from the past and planning for the future. Stroke 2013; 44:2664-9. [PMID: 23821232 DOI: 10.1161/strokeaha.113.001290] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Marc I Chimowitz
- Department of Neurology, Medical University of South Carolina, Charleston, SC, USA.
| |
Collapse
|
93
|
Menghini R, Casagrande V, Federici M. MicroRNAs in Endothelial Senescence and Atherosclerosis. J Cardiovasc Transl Res 2013; 6:924-30. [DOI: 10.1007/s12265-013-9487-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 06/10/2013] [Indexed: 10/26/2022]
|
94
|
Condorelli RA, Calogero AE, Vicari E, Duca Y, Favilla V, Morgia G, Cimino S, La Vignera S. Endothelial progenitor cells and erectile dysfunction: a brief review on diagnostic significance and summary of our experience. Aging Male 2013; 16:29-32. [PMID: 23597264 DOI: 10.3109/13685538.2013.789159] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The article provides a brief review of the literature concerning the diagnostic use of endothelial progenitor cells in patients with erectile dysfunction. In particular, patients with arterial erectile dysfunction could benefit from the use of this diagnostic marker, which in clinical practice can be used together with more conventional methods such as the penile Doppler. It is very important to acquire diagnostic tools for the diagnosis of sub clinical form of endothelial dysfunction in these patients, in particular when the erectile dysfunction is associated with cardiovascular risk factors.
Collapse
Affiliation(s)
- Rosita A Condorelli
- Department of Medical and Pediatric Sciences, University of Catania, Catania, Italy
| | | | | | | | | | | | | | | |
Collapse
|
95
|
Condorelli RA, Calogero AE, Vicari E, Favilla V, Morgia G, Cimino S, La Vignera S. Vascular regenerative therapies for the treatment of erectile dysfunction: current approaches. Andrology 2013; 1:533-40. [DOI: 10.1111/j.2047-2927.2013.00087.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Revised: 03/01/2013] [Accepted: 03/09/2013] [Indexed: 12/20/2022]
Affiliation(s)
- R. A. Condorelli
- Section of Endocrinology; Andrology and Internal Medicine; Department of Medical and Pediatric Sciences; University of Catania; Catania; Italy
| | - A. E. Calogero
- Section of Endocrinology; Andrology and Internal Medicine; Department of Medical and Pediatric Sciences; University of Catania; Catania; Italy
| | - E. Vicari
- Section of Endocrinology; Andrology and Internal Medicine; Department of Medical and Pediatric Sciences; University of Catania; Catania; Italy
| | - V. Favilla
- Department of Urology; University of Catania; Catania; Italy
| | - G. Morgia
- Department of Urology; University of Catania; Catania; Italy
| | - S. Cimino
- Department of Urology; University of Catania; Catania; Italy
| | - S. La Vignera
- Section of Endocrinology; Andrology and Internal Medicine; Department of Medical and Pediatric Sciences; University of Catania; Catania; Italy
| |
Collapse
|
96
|
CONSTANTINESCU MIHAELAIOANA, CONSTANTINESCU DANPETRU, ANDERCOU AUREL, MIRONIUC IONAUREL. Influence of sildenafil and donepezil administration on the serum redox balance in experimentally induced lower limb critical ischemia. CLUJUL MEDICAL (1957) 2013; 86:250-8. [PMID: 26527957 PMCID: PMC4462506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/10/2013] [Accepted: 09/11/2013] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Chronic lower limb ischemia (CLLI) leads to endothelial cell dysfunctions and endothelial lesions. The use of substances that release nitric oxide and activate endothelial nitric oxide synthase has proved to be useful in increasing angiogenesis and arteriogenesis under critical ischemia conditions. OBJECTIVES To investigate the therapeutic effect of Sildenafil and Donepezil with a vasodilating action in experimentally induced CLLI and on serum redox homeostasis. MATERIAL AND METHOD The research was performed in 3 groups of rats (n=10 animals/group) with experimentally induced CLLI: group I - control group; group II - animals treated postoperatively with a therapeutic dose of sildenafil, and group III - animals treated postoperatively with a therapeutic dose of donepezil. Oxidative stress (OS) indicators (malondialdehyde - MDA, protein carbonyls - PC), antioxidant (AO) defense indicators (reduced glutathione - GSH and oxidized glutathione - GSSH), and ceruloplasmin (CP) were determined on days 7, 14, 21 and 30. Statistical processing was performed using the Excel application (Microsoft Office 2007), with the StatsDirect v.2.7.2 software. RESULTS Changes in OS were evidenced in all groups on account of a decrease in MDA and PC. The greatest OS decrease in all groups was on day 30. AO defence changes were represented by decreased levels of GSH and GSSG in all groups, at the studied moments. Intracellular AO defense in the cytosol, nucleus and mitochondria was similar in all groups, (decreased GSH, GSSG and GSH/GSSG ratio). We found increased extracellular levels of GSH, GSSG, and CP and increased extracellular GSH/GSSG ratio at level compared to values on day 7. CONCLUSIONS 1) The administration of sildenafil (group II) and donepezil (group III) has favorable effects on reducing OS in experimentally induced CLLI. 2) Sildenafil and Donepezil administration stimulates extracellular AO defense on account of CP. 3) Sildenafil and Donepezil administration influences intracellular redox homeostasis on account of the GSH/GSSG couple, the major redox buffer in the body.
Collapse
Affiliation(s)
| | | | - AUREL ANDERCOU
- 2nd Surgical Clinic, County Clinic Hospital, Cluj-Napoca, Romania
| | | |
Collapse
|
97
|
Lin CP, Lin FY, Huang PH, Chen YL, Chen WC, Chen HY, Huang YC, Liao WL, Huang HC, Liu PL, Chen YH. Endothelial progenitor cell dysfunction in cardiovascular diseases: role of reactive oxygen species and inflammation. BIOMED RESEARCH INTERNATIONAL 2012; 2013:845037. [PMID: 23484163 PMCID: PMC3591199 DOI: 10.1155/2013/845037] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Accepted: 11/13/2012] [Indexed: 12/31/2022]
Abstract
Endothelial progenitor cells (EPCs) move towards injured endothelium or inflamed tissues and incorporate into foci of neovascularisation, thereby improving blood flow and tissue repair. Patients with cardiovascular diseases have been shown to exhibit reduced EPC number and function. It has become increasingly apparent that these changes may be effected in response to enhanced oxidative stress, possibly as a result of systemic and localised inflammatory responses. The interplay between inflammation and oxidative stress affects the initiation, progression, and complications of cardiovascular diseases. Recent studies suggest that inflammation and oxidative stress modulate EPC bioactivity. Clinical medications with anti-inflammatory and antioxidant properties, such as statins, thiazolidinediones, angiotensin II receptor 1 blockers, and angiotensin-converting enzyme inhibitors, are currently administered to patients with cardiovascular diseases. These medications appear to exert beneficial effects on EPC biology. This review focuses on EPC biology and explores the links between oxidative stress, inflammation, and development of cardiovascular diseases.
Collapse
Affiliation(s)
- Chih-Pei Lin
- Department of Biotechnology and Laboratory Science in Medicine and Institute of Biotechnology in Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Department of Pathology and Laboratory Medicine, Department of Internal Medicine and Divisions of Biochemistry and Cardiology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei 112, Taiwan
| | - Feng-Yen Lin
- Department of Internal Medicine, School of Medicine, Taipei Medical University and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
| | - Po-Hsun Huang
- Department of Pathology and Laboratory Medicine, Department of Internal Medicine and Divisions of Biochemistry and Cardiology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine and Cardiovascular Research Center, National Yang-Ming University, Taipei 112, Taiwan
- Faculty of Medicine and Institute of Clinical Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Wen-Chi Chen
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Huey-Yi Chen
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Yu-Chuen Huang
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Wen-Ling Liao
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Huey-Chun Huang
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yung-Hsiang Chen
- Graduate Institute of Integrated Medicine, School of Chinese Medicine, College of Chinese Medicine and Department of Medical Laboratory Science and Biotechnology, College of Health Care, China Medical University, Taichung 404, Taiwan
- Departments of Urology, Obstetrics and Gynecology and Medical Research, Genetics Centre and Center for Personalized Medicine, China Medical University Hospital, Taichung 404, Taiwan
| |
Collapse
|
98
|
Endothelial progenitor cells: the promise of cell-based therapies for acute lung injury. Inflamm Res 2012; 62:3-8. [PMID: 23138575 DOI: 10.1007/s00011-012-0570-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 10/03/2012] [Accepted: 10/22/2012] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) are defined as a special type of stem cell that have been found to directly incorporate into injured vessels and that participate in angiogenesis and reconstruction by differentiation into endothelial cells. EPCs are widely used to therapeutically treat cardiovascular disease, limb ischemia and vascular repair. However, the role of EPCs in inflammatory diseases, especially in lung injury, is less studied. OBJECTIVE To investigate the application of EPCs to vascular repair, and the role of EPCs in acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). METHODS A computer-based online search was performed in the PubMed database and Web of Science database for articles published, concerning EPCs, angiogenesis, ALI/ARDS and stem cell transplantation CONCLUSION EPCs have a therapeutic potential for vascular regeneration and may emerge as novel strategy for the diseases that are associated with ALI/ARDS.
Collapse
|
99
|
MicroRNAs in Vascular Biology. Int J Vasc Med 2012; 2012:794898. [PMID: 23056947 PMCID: PMC3463915 DOI: 10.1155/2012/794898] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Revised: 08/17/2012] [Accepted: 08/21/2012] [Indexed: 02/08/2023] Open
Abstract
Vascular inflammation is an important component of the pathophysiology of cardiovascular diseases, such as hypertension, atherosclerosis, and aneurysms. All vascular cells, including endothelial cells (ECs) and vascular smooth muscle cells (VSMCs), and infiltrating cells, such as macrophages, orchestrate a series of pathological events. Despite dramatic improvements in the treatment of atherosclerosis, the molecular basis of vascular inflammation is not well understood. In the last decade, microRNAs (miRNAs) have been revealed as novel regulators of vascular inflammation. Each miRNAs suppresses a set of genes, forming complex regulatory network. This paper provides an overview of current advances that have been made in revealing the roles of miRNAs during vascular inflammation. Recent studies show that miRNAs not only exist inside cells but also circulate in blood. These circulating miRNAs are useful biomarkers for diagnosis of cardiovascular diseases. Furthermore, recent studies demonstrate that circulating miRNAs are delivered into certain recipient cells and act as messengers. These studies suggest that miRNAs provide new therapeutic opportunities.
Collapse
|