151
|
Mandic L, Traxler D, Gugerell A, Zlabinger K, Lukovic D, Pavo N, Goliasch G, Spannbauer A, Winkler J, Gyöngyösi M. Molecular Imaging of Angiogenesis in Cardiac Regeneration. CURRENT CARDIOVASCULAR IMAGING REPORTS 2016; 9:27. [PMID: 27683600 PMCID: PMC5018257 DOI: 10.1007/s12410-016-9389-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Myocardial infarction (MI) leading to heart failure displays an important cause of death worldwide. Adequate restoration of blood flow to prevent this transition is a crucial factor to improve long-term morbidity and mortality. Novel regenerative therapies have been thoroughly investigated within the past decades. RECENT FINDINGS Increased angiogenesis in infarcted myocardium has shown beneficial effects on the prognosis of MI; therefore, the proangiogenic capacity of currently tested treatments is of specific interest. Molecular imaging to visualize formation of new blood vessels in vivo displays a promising option to monitor proangiogenic effects of regenerative substances. SUMMARY Based on encouraging results in preclinical models, molecular angiogenesis imaging has recently been applied in a small set of patients. This article reviews recent literature on noninvasive in vivo molecular imaging of angiogenesis after MI as an integral part of cardiac regeneration.
Collapse
Affiliation(s)
- Ljubica Mandic
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Denise Traxler
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Alfred Gugerell
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Katrin Zlabinger
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Dominika Lukovic
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Noemi Pavo
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Georg Goliasch
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Andreas Spannbauer
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Johannes Winkler
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
152
|
Rebouças JDS, Santos-Magalhães NS, Formiga FR. Cardiac Regeneration using Growth Factors: Advances and Challenges. Arq Bras Cardiol 2016; 107:271-275. [PMID: 27355588 PMCID: PMC5053196 DOI: 10.5935/abc.20160097] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 03/18/2016] [Accepted: 03/23/2016] [Indexed: 12/15/2022] Open
Abstract
Myocardial infarction is the most significant manifestation of ischemic heart disease and is associated with high morbidity and mortality. Novel strategies targeting at regenerating the injured myocardium have been investigated, including gene therapy, cell therapy, and the use of growth factors. Growth factor therapy has aroused interest in cardiovascular medicine because of the regeneration mechanisms induced by these biomolecules, including angiogenesis, extracellular matrix remodeling, cardiomyocyte proliferation, stem-cell recruitment, and others. Together, these mechanisms promote myocardial repair and improvement of the cardiac function. This review aims to address the strategic role of growth factor therapy in cardiac regeneration, considering its innovative and multifactorial character in myocardial repair after ischemic injury. Different issues will be discussed, with emphasis on the regeneration mechanisms as a potential therapeutic resource mediated by growth factors, and the challenges to make these proteins therapeutically viable in the field of cardiology and regenerative medicine. Resumo O infarto do miocárdio representa a manifestação mais significativa da cardiopatia isquêmica e está associado a elevada morbimortalidade. Novas estratégias vêm sendo investigadas com o intuito de regenerar o miocárdio lesionado, incluindo a terapia gênica, a terapia celular e a utilização de fatores de crescimento. A terapia com fatores de crescimento despertou interesse em medicina cardiovascular, devido aos mecanismos de regeneração induzidos por essas biomoléculas, incluindo angiogênese, remodelamento da matriz extracelular, proliferação de cardiomiócitos e recrutamento de células-tronco, dentre outros. Em conjunto, tais mecanismos promovem a reparação do miocárdio e a melhora da função cardíaca. Esta revisão pretende abordar o papel estratégico da terapia, com fatores de crescimento, para a regeneração cardíaca, considerando seu caráter inovador e multifatorial sobre o reparo do miocárdio após dano isquêmico. Diferentes questões serão discutidas, destacando-se os mecanismos de regeneração como recurso terapêutico potencial mediado por fatores de crescimento e os desafios para tornar essas proteínas terapeuticamente viáveis no âmbito da cardiologia e da medicina regenerativa.
Collapse
Affiliation(s)
- Juliana de Souza Rebouças
- Laboratório de Imunopatologia Keizo-Asami - Universidade
Federal de Pernambuco (UFPE), Recife, PE - Brazil
| | | | - Fabio Rocha Formiga
- Programa de Pós-Graduação em Biologia Celular e
Molecular Aplicada - Universidade de Pernambuco (UPE), Recife, PE - Brazil
- Curso de Pós-Graduação em Patologia
(UFBA/FIOCRUZ) - Centro de Pesquisas Gonçalo Moniz, Fundação
Oswaldo Cruz (FIOCRUZ), Salvador, BA - Brazil
| |
Collapse
|
153
|
Henning RJ. Therapeutic angiogenesis: angiogenic growth factors for ischemic heart disease. Future Cardiol 2016; 12:585-99. [PMID: 27420190 DOI: 10.2217/fca-2016-0006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Stem cells encode vascular endothelial growth factors (VEGFs), fibroblastic growth factors (FGFs), stem cell factor, stromal cell-derived factor, platelet growth factor and angiopoietin that can contribute to myocardial vascularization. VEGFs and FGFs are the most investigated growth factors. VEGFs regulate angiogenesis and vasculogenesis. FGFs stimulate vessel cell proliferation and differentiation and are regulators of endothelial cell migration, proliferation and survival. Clinical trials of VEGF or FGF for myocardial angiogenesis have produced disparate results. The efficacy of therapeutic angiogenesis can be improved by: (1) identifying the most optimal patients; (2) increased knowledge of angiogenic factor pharmacokinetics and proper dose; (3) prolonging contact of angiogenic factors with the myocardium; (4) increasing the efficiency of VEGF or FGF gene transduction; and (5) utilizing PET or MRI to measure myocardial perfusion and perfusion reserve.
Collapse
Affiliation(s)
- Robert J Henning
- The University of South Florida and the James A. Haley Hospital, Tampa, FL 33612 USA
| |
Collapse
|
154
|
Signorelli SS, Volsi GL, Pitruzzella A, Fiore V, Mangiafico M, Vanella L, Parenti R, Rizzo M, Volti GL. Circulating miR-130a, miR-27b, and miR-210 in Patients With Peripheral Artery Disease and Their Potential Relationship With Oxidative Stress. Angiology 2016; 67:945-950. [PMID: 26980776 DOI: 10.1177/0003319716638242] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Some emerging risk factors such as oxidative stress biomarkers and microRNAs (miRs) may add additional value to the established risk factors for peripheral artery disease (PAD). We enrolled 27 patients with PAD and 27 age-matched controls. We examined the levels of a series of miRs (miR-130a, miR-27b, and miR-210) in serum samples. The level of well-established oxidative stress biomarkers, such as lipid hydroperoxides, isoprostanes, hemeoxygenase-1 (HO-1) and reduced glutathione, was also measured in plasma and their relationship with the miRs was determined. Levels of miR-130a, miR-27b, and miR-210 were significantly increased in patients with PAD when compared to the controls. The level of miR-130 was positively correlated with body mass index, whereas miR-210 was inversely associated with pain-free walking distance (PfWD). None of the evaluated miRs was associated with lowered PfWD of patients with PAD (stage IIa > 250 m, IIb < 250 m) or oxidative stress parameters. In conclusion, our findings suggest the need for more research to assess if miRs can serve as useful markers for the early diagnosis and monitoring of PAD.
Collapse
Affiliation(s)
| | - Guido Li Volsi
- 2 Biomedical and Biotechnological Science, University of Catania, Catania, Italy
| | - Alessandro Pitruzzella
- 3 Department of Experimental Biomedicine and Clinical Neuroscience, University of Palermo, Palermo, Italy
| | - Valerio Fiore
- 1 Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Marco Mangiafico
- 1 Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Luca Vanella
- 4 Department of Drug Science, University of Catania, Catania, Italy
| | - Rosalba Parenti
- 2 Biomedical and Biotechnological Science, University of Catania, Catania, Italy
| | - Manfredi Rizzo
- 5 Biomedical Department of Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Giovanni Li Volti
- 2 Biomedical and Biotechnological Science, University of Catania, Catania, Italy
| |
Collapse
|
155
|
EphrinB2/EphB4 pathway in postnatal angiogenesis: a potential therapeutic target for ischemic cardiovascular disease. Angiogenesis 2016; 19:297-309. [PMID: 27216867 DOI: 10.1007/s10456-016-9514-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 05/13/2016] [Indexed: 01/12/2023]
Abstract
Ischemic cardiovascular disease remains one of the leading causes of morbidity and mortality in the world. Proangiogenic therapy appears to be a promising and feasible strategy for the patients with ischemic cardiovascular disease, but the results of preclinical and clinical trials are limited due to the complicated mechanisms of angiogenesis. Facilitating the formation of functional vessels is important in rescuing the ischemic cardiomyocytes. EphrinB2/EphB4, a novel pathway in angiogenesis, plays a critical role in both microvascular growth and neovascular maturation. Hence, investigating the mechanisms of EphrinB2/EphB4 pathway in angiogenesis may contribute to the development of novel therapeutics for ischemic cardiovascular disease. Previous reviews mainly focused on the role of EphrinB2/EphB4 pathway in embryo vascular development, but their role in postnatal angiogenesis in ischemic heart disease has not been fully illustrated. Here, we summarized the current knowledge of EphrinB2/EphB4 in angiogenesis and their interaction with other angiogenic pathways in ischemic cardiovascular disease.
Collapse
|
156
|
Awada HK, Johnson LA, Hitchens TK, Foley LM, Wang Y. Factorial Design of Experiments to Optimize Multiple Protein Delivery for Cardiac Repair. ACS Biomater Sci Eng 2016; 2:879-886. [PMID: 33440484 DOI: 10.1021/acsbiomaterials.6b00146] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Myocardial infarction (MI) is a major cardiovascular disease responsible for millions of deaths annually. Protein therapies can potentially repair and regenerate the infarcted myocardium. However, because of the short half-lives of proteins in vivo, their low retention at the target tissue, and the lack of spatiotemporal cues upon injection, the efficacy of protein therapy can be limited. This efficacy can be improved by utilizing controlled release systems to overcome shortcomings associated with a direct bolus injection. Equally important is the determination of an optimal combination of different proteins having distinct roles in cardiac function and repairs to prevent or reverse the multiple pathologies that develop after infarction. In this work, we used a rat MI model to test a combination of potentially complementary proteins: tissue inhibitor of metalloproteinases 3 (TIMP-3), interleukin-10 (IL-10), basic fibroblast growth factor (FGF-2), and stromal cell-derived factor 1 alpha (SDF-1α). To achieve controlled and timed release of the proteins per their physiologic cues during proper tissue repair, we used a fibrin gel-coacervate composite. TIMP-3 and IL-10 were encapsulated in fibrin gel to offer early release, while FGF-2 and SDF-1α were encapsulated in heparin-based coacervates and distributed in the same fibrin gel to offer sustained release. We utilized a powerful statistical tool, factorial design of experiments (DOE), to refine this protein combination based on its improvement of ejection fraction 4 weeks after MI. We found that TIMP-3, FGF-2, and SDF-1α demonstrated significant contributions toward improving the ejection fraction, while the IL-10's effect was insignificant. The results also suggested that the higher doses tested for TIMP-3, FGF-2, and SDF-1α had greater benefit on function than lower doses and that there existed slight antagonism between TIMP-3 and FGF-2. Taken together, we conclude that factorial DOE can guide the evolution of multiple protein therapies in a small number of runs, saving time, money, and resources for finding the optimal dose and composition.
Collapse
Affiliation(s)
| | - Louis A Johnson
- SnapDat Inc., 733 West Foster Avenue, State College, Pennsylvania 16801, United States
| | | | | | | |
Collapse
|
157
|
Mykhaylichenko VY, Kubyshkin AV, Samarin SA, Fomochkina II, Anisimova LV. Experimental induction of reparative morphogenesis and adaptive reserves in the ischemic myocardium using multipotent mesenchymal bone marrow-derived stem cells. ACTA ACUST UNITED AC 2016; 23:95-104. [PMID: 27102896 DOI: 10.1016/j.pathophys.2016.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 04/06/2016] [Accepted: 04/12/2016] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Current experimental research has proven the efficacy of transplantation bone marrow-derived mesenchymal stem cells (MSC) in the treatment of myocardial infarction (MI). The one of the main purposes of research was to evaluate the comparative data of the MSC transplantation with (5-azacytidine) and without commitment and to assess the post transplantation effects. METHODS The efficiency of intravenous cardiomyoplasty by infusion of MSC was evaluated in female Wistar-Kyoto rats with myocardial infarction model using echocardiography, morphological study, morphometry, immunohistostaining, data from in situ hybridization, and by measurement of blood serum levels of nitric oxide, endothelin-1, vascular endothelial growth factor (VEGF), and fibroblast growth factor 2 (FGF2). RESULTS The transplanted MSC were detected in all layers of the myocardium; MCS actively participate in the formation of blood vessels and connective tissue in the scar zone. There was no observable differentiation of male MSC into cardiomyocytes in female rats with MI. However, MSC transplantation leads to significant improvement in vascularization in the area of MI, elevation blood serum levels of nitric oxide, VEGF, and FGF2. No significant differences were identified morphologically between the two groups of animals after transplantation with unmodified MSC or commited MSC (5-azaC). CONCLUSION Intravenous transplantation of MSC without commitment in rats with MI improves the contractile function of the heart, the morphology of the myocardium, and should be recommended for further clinical investigation as an alternative approach to deal with heart diseases.
Collapse
Affiliation(s)
- V Yu Mykhaylichenko
- Department of General Surgery, Medical Academy named after S.I. Georgievskiy of the V.I. Vernadsky Crimean Federal University, Lenin Blvd, 5/7, 295006 Simferopol, Republic of Crimea, Russian Federation
| | - A V Kubyshkin
- Department of General and Clinical Pathophysiology, Medical Academy named after S.I. Georgievskiy of the V.I. Vernadsky Crimean Federal University, Republic of Crimea, Russian Federation.
| | - S A Samarin
- Department of Anesthesiology, Medical Academy named after S.I. Georgievskiy of the V.I. Vernadsky Crimean Federal University, Republic of Crimea, Russian Federation
| | - I I Fomochkina
- Department of General and Clinical Pathophysiology, Medical Academy named after S.I. Georgievskiy of the V.I. Vernadsky Crimean Federal University, Republic of Crimea, Russian Federation
| | - L V Anisimova
- Department of General and Clinical Pathophysiology, Medical Academy named after S.I. Georgievskiy of the V.I. Vernadsky Crimean Federal University, Republic of Crimea, Russian Federation
| |
Collapse
|
158
|
Awada HK, Hwang MP, Wang Y. Towards comprehensive cardiac repair and regeneration after myocardial infarction: Aspects to consider and proteins to deliver. Biomaterials 2016; 82:94-112. [PMID: 26757257 PMCID: PMC4872516 DOI: 10.1016/j.biomaterials.2015.12.025] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/15/2015] [Accepted: 12/19/2015] [Indexed: 12/13/2022]
Abstract
Ischemic heart disease is a leading cause of death worldwide. After the onset of myocardial infarction, many pathological changes take place and progress the disease towards heart failure. Pathologies such as ischemia, inflammation, cardiomyocyte death, ventricular remodeling and dilation, and interstitial fibrosis, develop and involve the signaling of many proteins. Proteins can play important roles in limiting or countering pathological changes after infarction. However, they typically have short half-lives in vivo in their free form and can benefit from the advantages offered by controlled release systems to overcome their challenges. The controlled delivery of an optimal combination of proteins per their physiologic spatiotemporal cues to the infarcted myocardium holds great potential to repair and regenerate the heart. The effectiveness of therapeutic interventions depends on the elucidation of the molecular mechanisms of the cargo proteins and the spatiotemporal control of their release. It is likely that multiple proteins will provide a more comprehensive and functional recovery of the heart in a controlled release strategy.
Collapse
Affiliation(s)
- Hassan K Awada
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Mintai P Hwang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Yadong Wang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, PA 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| |
Collapse
|
159
|
Howangyin KY, Zlatanova I, Pinto C, Ngkelo A, Cochain C, Rouanet M, Vilar J, Lemitre M, Stockmann C, Fleischmann BK, Mallat Z, Silvestre JS. Myeloid-Epithelial-Reproductive Receptor Tyrosine Kinase and Milk Fat Globule Epidermal Growth Factor 8 Coordinately Improve Remodeling After Myocardial Infarction via Local Delivery of Vascular Endothelial Growth Factor. Circulation 2016; 133:826-39. [PMID: 26819373 PMCID: PMC4767109 DOI: 10.1161/circulationaha.115.020857] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 01/22/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND In infarcted heart, improper clearance of dying cells by activated neighboring phagocytes may precipitate the transition to heart failure. We analyzed the coordinated role of 2 major mediators of efferocytosis, the myeloid-epithelial-reproductive protein tyrosine kinase (Mertk) and the milk fat globule epidermal growth factor (Mfge8), in directing cardiac remodeling by skewing the inflammatory response after myocardial infarction. METHODS AND RESULTS We generated double-deficient mice for Mertk and Mfge8 (Mertk(-/-)/Mfge8(-/-)) and challenged them with acute coronary ligature. Compared with wild-type, Mertk-deficient (Mertk(-/-)), or Mfge8-deficient (Mfge8(-/-)) animals, Mertk(-/-)/Mfge8(-/-) mice displayed greater alteration in cardiac function and remodeling. Mertk and Mfge8 were expressed mainly by cardiac Ly6C(High and Low) monocytes and macrophages. In parallel, Mertk(-/-)/Mfge8(-/-) bone marrow chimeras manifested increased accumulation of apoptotic cells, enhanced fibrotic area, and larger infarct size, as well as reduced angiogenesis. We found that the abrogation of efferocytosis affected neither the ability of circulating monocytes to infiltrate cardiac tissue nor the number of resident Ly6C(High) and Ly6C(How) monocytes/macrophages populating the infarcted milieu. In contrast, combined Mertk and Mfge8 deficiency in Ly6C(High)/Ly6C(Low) monocytes/macrophages either obtained from in vitro differentiation of bone marrow cells or isolated from infarcted hearts altered their capacity of efferocytosis and subsequently blunted vascular endothelial growth factor A (VEGFA) release. Using LysMCre(+)/VEGFA(fl/fl) mice, we further identified an important role for myeloid-derived VEGFA in improving cardiac function and angiogenesis. CONCLUSIONS After myocardial infarction, Mertk- and Mfge8-expressing monocyte/macrophages synergistically engage the clearance of injured cardiomyocytes, favoring the secretion of VEGFA to locally repair the dysfunctional heart.
Collapse
Affiliation(s)
- Kiave-Yune Howangyin
- From INSERM UMRS 970, Université Paris Descartes, Sorbonne Paris Cité, France (K.-Y.H., I.Z., C.P., A.N., M.R., J.V., M.L., C.S., Z.M., J.-S.S.); Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Germany (C.C.); Institute of Physiology I, Life & Brain Center, University of Bonn, Germany (B.K.F.); and Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, UK (Z.M.)
| | - Ivana Zlatanova
- From INSERM UMRS 970, Université Paris Descartes, Sorbonne Paris Cité, France (K.-Y.H., I.Z., C.P., A.N., M.R., J.V., M.L., C.S., Z.M., J.-S.S.); Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Germany (C.C.); Institute of Physiology I, Life & Brain Center, University of Bonn, Germany (B.K.F.); and Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, UK (Z.M.)
| | - Cristina Pinto
- From INSERM UMRS 970, Université Paris Descartes, Sorbonne Paris Cité, France (K.-Y.H., I.Z., C.P., A.N., M.R., J.V., M.L., C.S., Z.M., J.-S.S.); Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Germany (C.C.); Institute of Physiology I, Life & Brain Center, University of Bonn, Germany (B.K.F.); and Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, UK (Z.M.)
| | - Anta Ngkelo
- From INSERM UMRS 970, Université Paris Descartes, Sorbonne Paris Cité, France (K.-Y.H., I.Z., C.P., A.N., M.R., J.V., M.L., C.S., Z.M., J.-S.S.); Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Germany (C.C.); Institute of Physiology I, Life & Brain Center, University of Bonn, Germany (B.K.F.); and Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, UK (Z.M.)
| | - Clément Cochain
- From INSERM UMRS 970, Université Paris Descartes, Sorbonne Paris Cité, France (K.-Y.H., I.Z., C.P., A.N., M.R., J.V., M.L., C.S., Z.M., J.-S.S.); Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Germany (C.C.); Institute of Physiology I, Life & Brain Center, University of Bonn, Germany (B.K.F.); and Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, UK (Z.M.)
| | - Marie Rouanet
- From INSERM UMRS 970, Université Paris Descartes, Sorbonne Paris Cité, France (K.-Y.H., I.Z., C.P., A.N., M.R., J.V., M.L., C.S., Z.M., J.-S.S.); Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Germany (C.C.); Institute of Physiology I, Life & Brain Center, University of Bonn, Germany (B.K.F.); and Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, UK (Z.M.)
| | - José Vilar
- From INSERM UMRS 970, Université Paris Descartes, Sorbonne Paris Cité, France (K.-Y.H., I.Z., C.P., A.N., M.R., J.V., M.L., C.S., Z.M., J.-S.S.); Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Germany (C.C.); Institute of Physiology I, Life & Brain Center, University of Bonn, Germany (B.K.F.); and Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, UK (Z.M.)
| | - Mathilde Lemitre
- From INSERM UMRS 970, Université Paris Descartes, Sorbonne Paris Cité, France (K.-Y.H., I.Z., C.P., A.N., M.R., J.V., M.L., C.S., Z.M., J.-S.S.); Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Germany (C.C.); Institute of Physiology I, Life & Brain Center, University of Bonn, Germany (B.K.F.); and Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, UK (Z.M.)
| | - Christian Stockmann
- From INSERM UMRS 970, Université Paris Descartes, Sorbonne Paris Cité, France (K.-Y.H., I.Z., C.P., A.N., M.R., J.V., M.L., C.S., Z.M., J.-S.S.); Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Germany (C.C.); Institute of Physiology I, Life & Brain Center, University of Bonn, Germany (B.K.F.); and Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, UK (Z.M.)
| | - Bernd K Fleischmann
- From INSERM UMRS 970, Université Paris Descartes, Sorbonne Paris Cité, France (K.-Y.H., I.Z., C.P., A.N., M.R., J.V., M.L., C.S., Z.M., J.-S.S.); Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Germany (C.C.); Institute of Physiology I, Life & Brain Center, University of Bonn, Germany (B.K.F.); and Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, UK (Z.M.)
| | - Ziad Mallat
- From INSERM UMRS 970, Université Paris Descartes, Sorbonne Paris Cité, France (K.-Y.H., I.Z., C.P., A.N., M.R., J.V., M.L., C.S., Z.M., J.-S.S.); Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Germany (C.C.); Institute of Physiology I, Life & Brain Center, University of Bonn, Germany (B.K.F.); and Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, UK (Z.M.)
| | - Jean-Sébastien Silvestre
- From INSERM UMRS 970, Université Paris Descartes, Sorbonne Paris Cité, France (K.-Y.H., I.Z., C.P., A.N., M.R., J.V., M.L., C.S., Z.M., J.-S.S.); Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Germany (C.C.); Institute of Physiology I, Life & Brain Center, University of Bonn, Germany (B.K.F.); and Division of Cardiovascular Medicine, University of Cambridge, Addenbrooke's Hospital, UK (Z.M.).
| |
Collapse
|
160
|
Yue X, Lin X, Yang T, Yang X, Yi X, Jiang X, Li X, Li T, Guo J, Dai Y, Shi J, Wei L, Youker KA, Torre-Amione G, Yu Y, Andrade KC, Chang J. Rnd3/RhoE Modulates Hypoxia-Inducible Factor 1α/Vascular Endothelial Growth Factor Signaling by Stabilizing Hypoxia-Inducible Factor 1α and Regulates Responsive Cardiac Angiogenesis. Hypertension 2016; 67:597-605. [PMID: 26781283 DOI: 10.1161/hypertensionaha.115.06412] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 12/16/2015] [Indexed: 12/25/2022]
Abstract
The insufficiency of compensatory angiogenesis in the heart of patients with hypertension contributes to heart failure transition. The hypoxia-inducible factor 1α-vascular endothelial growth factor (HIF1α-VEGF) signaling cascade controls responsive angiogenesis. One of the challenges in reprograming the insufficient angiogenesis is to achieve a sustainable tissue exposure to the proangiogenic factors, such as HIF1α stabilization. In this study, we identified Rnd3, a small Rho GTPase, as a proangiogenic factor participating in the regulation of the HIF1α-VEGF signaling cascade. Rnd3 physically interacted with and stabilized HIF1α, and consequently promoted VEGFA expression and endothelial cell tube formation. To demonstrate this proangiogenic role of Rnd3 in vivo, we generated Rnd3 knockout mice. Rnd3 haploinsufficient (Rnd3(+/-)) mice were viable, yet developed dilated cardiomyopathy with heart failure after transverse aortic constriction stress. The poststress Rnd3(+/-) hearts showed significantly impaired angiogenesis and decreased HIF1α and VEGFA expression. The angiogenesis defect and heart failure phenotype were partially rescued by cobalt chloride treatment, a HIF1α stabilizer, confirming a critical role of Rnd3 in stress-responsive angiogenesis. Furthermore, we generated Rnd3 transgenic mice and demonstrated that Rnd3 overexpression in heart had a cardioprotective effect through reserved cardiac function and preserved responsive angiogenesis after pressure overload. Finally, we assessed the expression levels of Rnd3 in the human heart and detected significant downregulation of Rnd3 in patients with end-stage heart failure. We concluded that Rnd3 acted as a novel proangiogenic factor involved in cardiac responsive angiogenesis through HIF1α-VEGFA signaling promotion. Rnd3 downregulation observed in patients with heart failure may explain the insufficient compensatory angiogenesis involved in the transition to heart failure.
Collapse
Affiliation(s)
- Xiaojing Yue
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Xi Lin
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Tingli Yang
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Xiangsheng Yang
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Xin Yi
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Xuejun Jiang
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Xiaoyan Li
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Tianfa Li
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Junli Guo
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Yuan Dai
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Jianjian Shi
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Lei Wei
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Keith A Youker
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Guillermo Torre-Amione
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Yanhong Yu
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Kelsey C Andrade
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.)
| | - Jiang Chang
- From the Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (X.Y., Y.Y.); Texas A&M University Health Science Center, Institute of Biosciences and Technology, Houston (X.Y., X.L., T.Y., X.Y., Y.D., K.C.A., J.C.); Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (X.Y., X.J., X.L.); Cardiovascular Disease and Research Institute, Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China (T.L., J.G.); Department of Pediatrics, Indiana University School of Medicine, Indianapolis (J.S., L.W.); and Methodist DeBakey Heart and Vascular Center, The Methodist Hospital, Houston, TX (K.A.Y., G.T.-A.).
| |
Collapse
|
161
|
White CI, Jansen MA, McGregor K, Mylonas KJ, Richardson RV, Thomson A, Moran CM, Seckl JR, Walker BR, Chapman KE, Gray GA. Cardiomyocyte and Vascular Smooth Muscle-Independent 11β-Hydroxysteroid Dehydrogenase 1 Amplifies Infarct Expansion, Hypertrophy, and the Development of Heart Failure After Myocardial Infarction in Male Mice. Endocrinology 2016; 157:346-57. [PMID: 26465199 PMCID: PMC4701896 DOI: 10.1210/en.2015-1630] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Global deficiency of 11β-hydroxysteroid dehydrogenase type 1 (11β-HSD1), an enzyme that regenerates glucocorticoids within cells, promotes angiogenesis, and reduces acute infarct expansion after myocardial infarction (MI), suggesting that 11β-HSD1 activity has an adverse influence on wound healing in the heart after MI. The present study investigated whether 11β-HSD1 deficiency could prevent the development of heart failure after MI and examined whether 11β-HSD1 deficiency in cardiomyocytes and vascular smooth muscle cells confers this protection. Male mice with global deficiency in 11β-HSD1, or with Hsd11b1 disruption in cardiac and vascular smooth muscle (via SM22α-Cre recombinase), underwent coronary artery ligation for induction of MI. Acute injury was equivalent in all groups. However, by 8 weeks after induction of MI, relative to C57Bl/6 wild type, globally 11β-HSD1-deficient mice had reduced infarct size (34.7 ± 2.1% left ventricle [LV] vs 44.0 ± 3.3% LV, P = .02), improved function (ejection fraction, 33.5 ± 2.5% vs 24.7 ± 2.5%, P = .03) and reduced ventricular dilation (LV end-diastolic volume, 0.17 ± 0.01 vs 0.21 ± 0.01 mL, P = .01). This was accompanied by a reduction in hypertrophy, pulmonary edema, and in the expression of genes encoding atrial natriuretic peptide and β-myosin heavy chain. None of these outcomes, nor promotion of periinfarct angiogenesis during infarct repair, were recapitulated when 11β-HSD1 deficiency was restricted to cardiac and vascular smooth muscle. 11β-HSD1 expressed in cells other than cardiomyocytes or vascular smooth muscle limits angiogenesis and promotes infarct expansion with adverse ventricular remodeling after MI. Early pharmacological inhibition of 11β-HSD1 may offer a new therapeutic approach to prevent heart failure associated with ischemic heart disease.
Collapse
MESH Headings
- 11-beta-Hydroxysteroid Dehydrogenase Type 1/deficiency
- 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics
- 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism
- Animals
- Cardiomegaly/etiology
- Cardiomegaly/prevention & control
- Coronary Circulation
- Crosses, Genetic
- Gene Expression Regulation
- Heart Failure/etiology
- Heart Failure/prevention & control
- Heart Ventricles/metabolism
- Heart Ventricles/pathology
- Heart Ventricles/physiopathology
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Muscle, Smooth, Vascular/enzymology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocardial Infarction/metabolism
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Neovascularization, Physiologic
- Organ Size
- Pulmonary Edema/etiology
- Pulmonary Edema/prevention & control
- Stroke Volume
Collapse
Affiliation(s)
- Christopher I White
- British Heart Foundation/University Centre for Cardiovascular Science (C.I.W., M.A.J., K.M., K.J.M., R.V.R., C.M.M., J.R.S., B.R.W., K.E.C., G.A.G.), Queens Medical Research Institute, and Edinburgh Preclinical Imaging (M.A.J., A.T., C.M.M.), College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Maurits A Jansen
- British Heart Foundation/University Centre for Cardiovascular Science (C.I.W., M.A.J., K.M., K.J.M., R.V.R., C.M.M., J.R.S., B.R.W., K.E.C., G.A.G.), Queens Medical Research Institute, and Edinburgh Preclinical Imaging (M.A.J., A.T., C.M.M.), College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Kieran McGregor
- British Heart Foundation/University Centre for Cardiovascular Science (C.I.W., M.A.J., K.M., K.J.M., R.V.R., C.M.M., J.R.S., B.R.W., K.E.C., G.A.G.), Queens Medical Research Institute, and Edinburgh Preclinical Imaging (M.A.J., A.T., C.M.M.), College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Katie J Mylonas
- British Heart Foundation/University Centre for Cardiovascular Science (C.I.W., M.A.J., K.M., K.J.M., R.V.R., C.M.M., J.R.S., B.R.W., K.E.C., G.A.G.), Queens Medical Research Institute, and Edinburgh Preclinical Imaging (M.A.J., A.T., C.M.M.), College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Rachel V Richardson
- British Heart Foundation/University Centre for Cardiovascular Science (C.I.W., M.A.J., K.M., K.J.M., R.V.R., C.M.M., J.R.S., B.R.W., K.E.C., G.A.G.), Queens Medical Research Institute, and Edinburgh Preclinical Imaging (M.A.J., A.T., C.M.M.), College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Adrian Thomson
- British Heart Foundation/University Centre for Cardiovascular Science (C.I.W., M.A.J., K.M., K.J.M., R.V.R., C.M.M., J.R.S., B.R.W., K.E.C., G.A.G.), Queens Medical Research Institute, and Edinburgh Preclinical Imaging (M.A.J., A.T., C.M.M.), College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Carmel M Moran
- British Heart Foundation/University Centre for Cardiovascular Science (C.I.W., M.A.J., K.M., K.J.M., R.V.R., C.M.M., J.R.S., B.R.W., K.E.C., G.A.G.), Queens Medical Research Institute, and Edinburgh Preclinical Imaging (M.A.J., A.T., C.M.M.), College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Jonathan R Seckl
- British Heart Foundation/University Centre for Cardiovascular Science (C.I.W., M.A.J., K.M., K.J.M., R.V.R., C.M.M., J.R.S., B.R.W., K.E.C., G.A.G.), Queens Medical Research Institute, and Edinburgh Preclinical Imaging (M.A.J., A.T., C.M.M.), College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Brian R Walker
- British Heart Foundation/University Centre for Cardiovascular Science (C.I.W., M.A.J., K.M., K.J.M., R.V.R., C.M.M., J.R.S., B.R.W., K.E.C., G.A.G.), Queens Medical Research Institute, and Edinburgh Preclinical Imaging (M.A.J., A.T., C.M.M.), College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Karen E Chapman
- British Heart Foundation/University Centre for Cardiovascular Science (C.I.W., M.A.J., K.M., K.J.M., R.V.R., C.M.M., J.R.S., B.R.W., K.E.C., G.A.G.), Queens Medical Research Institute, and Edinburgh Preclinical Imaging (M.A.J., A.T., C.M.M.), College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Gillian A Gray
- British Heart Foundation/University Centre for Cardiovascular Science (C.I.W., M.A.J., K.M., K.J.M., R.V.R., C.M.M., J.R.S., B.R.W., K.E.C., G.A.G.), Queens Medical Research Institute, and Edinburgh Preclinical Imaging (M.A.J., A.T., C.M.M.), College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh EH16 4TJ, Scotland, United Kingdom
| |
Collapse
|
162
|
Scofield SLC, Amin P, Singh M, Singh K. Extracellular Ubiquitin: Role in Myocyte Apoptosis and Myocardial Remodeling. Compr Physiol 2015; 6:527-60. [PMID: 26756642 DOI: 10.1002/cphy.c150025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ubiquitin (UB) is a highly conserved low molecular weight (8.5 kDa) protein. It consists of 76 amino acid residues and is found in all eukaryotic cells. The covalent linkage of UB to a variety of cellular proteins (ubiquitination) is one of the most common posttranslational modifications in eukaryotic cells. This modification generally regulates protein turnover and protects the cells from damaged or misfolded proteins. The polyubiquitination of proteins serves as a signal for degradation via the 26S proteasome pathway. UB is present in trace amounts in body fluids. Elevated levels of UB are described in the serum or plasma of patients under a variety of conditions. Extracellular UB is proposed to have pleiotropic roles including regulation of immune response, anti-inflammatory, and neuroprotective activities. CXCR4 is identified as receptor for extracellular UB in hematopoietic cells. Heart failure represents a major cause of morbidity and mortality in western society. Cardiac remodeling is a determinant of the clinical course of heart failure. The components involved in myocardial remodeling include-myocytes, fibroblasts, interstitium, and coronary vasculature. Increased sympathetic nerve activity in the form of norepinephrine is a common feature during heart failure. Acting via β-adrenergic receptor (β-AR), norepinephrine is shown to induce myocyte apoptosis and myocardial fibrosis. β-AR stimulation increases extracellular levels of UB in myocytes, and UB inhibits β-AR-stimulated increases in myocyte apoptosis and myocardial fibrosis. This review summarizes intracellular and extracellular functions of UB with particular emphasis on the role of extracellular UB in cardiac myocyte apoptosis and myocardial remodeling.
Collapse
Affiliation(s)
- Stephanie L C Scofield
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee, USA
| | - Parthiv Amin
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee, USA
| | - Mahipal Singh
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee, USA
| | - Krishna Singh
- Department of Biomedical Sciences, East Tennessee State University, Johnson City, Tennessee, USA; Center for Inflammation, Infectious Disease and Immunity, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee, USA; James H. Quillen VA Medical Center, East Tennessee State University, Johnson City, Tennessee, USA.,Department of Medicine, Albany Medical College, Albany, New York, USA.,Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, New York, USA
| |
Collapse
|
163
|
Margulis K, Neofytou EA, Beygui RE, Zare RN. Celecoxib Nanoparticles for Therapeutic Angiogenesis. ACS NANO 2015; 9:9416-9426. [PMID: 26244654 DOI: 10.1021/acsnano.5b04137] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Controllable induction of blood vessel formation (angiogenesis) presents an important therapeutic goal in ischemic diseases and is also beneficial in various normal physiological processes. In this study, we have shown that nanoparticles of celecoxib, a lipophilic nonsteroidal anti-inflammatory drug, effectively evoke therapeutic angiogenesis in animal models, in both normal and ischemic organs. Celecoxib is widely considered to inhibit angiogenesis, although a recent study suggests that it can instead promote blood vessel growth in cancer cell lines. The hydrophobic nature of this drug necessitates its administration in nanoparticulate form in order to elicit a perceivable pharmacological response. We developed a facile method for nanoparticle formation by solvent extraction from microemulsions in supercritical carbon dioxide. This method exploits a spontaneous formation of nanometric domains within the microemulsion system and their rapid conversion to nanoparticles by supercritical fluid. The resultant nanoparticles were administered subcutaneously to mice in a biocompatible hydrogel, and caused a 4-fold increase in blood vessel count in normally perfused skin compared with drug-free particles. They were at least as effective in inducing angiogenesis as nanoparticles of deferoxamine, a well-established neovascularization promoter. Next, we evaluated their effect on ischemic tissues in murine model of myocardial infarction. We found that celecoxib nanoparticles were able to induce a significant vascularization of ischemic myocardium and hamper the progression of heart failure, which points toward a new approach for treating ischemia.
Collapse
Affiliation(s)
- Katherine Margulis
- Department of Chemistry, Stanford University , Stanford, California 94305-5080, United States
| | - Evgenios A Neofytou
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine , Stanford, California 94305-5407, United States
| | - Ramin E Beygui
- Department of Cardiothoracic Surgery, Falk Cardiovascular Research Center, Stanford University School of Medicine , Stanford, California 94305-5407, United States
- Heart and Vascular Center, NorthBay Medical Center ,1200 B. Gale Wilson Boulevard, Fairfield, California 94533, United States
| | - Richard N Zare
- Department of Chemistry, Stanford University , Stanford, California 94305-5080, United States
| |
Collapse
|
164
|
Yu JM, Zhang XB, Jiang W, Wang HD, Zhang YN. Astragalosides promote angiogenesis via vascular endothelial growth factor and basic fibroblast growth factor in a rat model of myocardial infarction. Mol Med Rep 2015; 12:6718-26. [PMID: 26352430 PMCID: PMC4626201 DOI: 10.3892/mmr.2015.4307] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 07/28/2015] [Indexed: 12/24/2022] Open
Abstract
The aim of the present study was to evaluate the effect of astragalosides (ASTs) on angiogenesis, as well as the expression of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) following myocardial infarction (MI). MI was induced in rats by ligation of the left coronary artery. Twenty-four hours after surgery, the rats were divided into low-dose, high-dose, control and sham surgery groups (n=8 per group). The low- and high-dose groups were treated with ASTs (2.5 and 10 mg/kg/day, respectively, via intraperitoneal injection), while, the control and sham surgery group rats received saline. Serum levels, and mRNA and protein expression levels of VEGF and bFGF, as well as the microvessel density (MVD) were determined four weeks post-treatment. Twenty-four hours post-surgery, VEGF and bFGF serum levels were observed to be comparable between the groups; while at four weeks, the VEGF and bFGF levels were higher in the AST-treated rats (P<0.01). Similarly, VEGF and bFGF mRNA and protein expression levels were higher following AST treatment (P<0.05). No difference in VEGF mRNA expression between the low- and high-dose groups was noted, however, an increase in the bFGF expression levels was detected in the high-dose group. Newly generated blood vessels were observed following MI, with a significant increase in MVD observed in the AST-treated groups (P<0.05). AST promotes angiogenesis of the heart and increases VEGF and bFGF expression levels. Thus, it is hypothesized that increased VEGF and bFGF levels may contribute to the AST-induced increase in angiogenesis in rat models of MI.
Collapse
Affiliation(s)
- Jun-Min Yu
- Department of Gerontology, The Fourth Clinical Medical College, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Xiao-Bo Zhang
- Department of Gerontology, The Fourth Clinical Medical College, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Wen Jiang
- Department of Gerontology, The Fourth Clinical Medical College, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hui-Dong Wang
- Department of Gerontology, The Fourth Clinical Medical College, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yi-Na Zhang
- Department of Gerontology, The Second Clinical Medical College, Harbin Medical University, Harbin, Heilongjiang 150086, P.R. China
| |
Collapse
|
165
|
Liu X, Sun X, Liao H, Dong Z, Zhao J, Zhu H, Wang P, Shen L, Xu L, Ma X, Shen C, Fan F, Wang C, Hu K, Zou Y, Ge J, Ren J, Sun A. Mitochondrial Aldehyde Dehydrogenase 2 Regulates Revascularization in Chronic Ischemia: Potential Impact on the Development of Coronary Collateral Circulation. Arterioscler Thromb Vasc Biol 2015; 35:2196-206. [PMID: 26315408 DOI: 10.1161/atvbaha.115.306012] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/30/2015] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Revascularization is an essential process to compensate for cardiac underperfusion and, therefore, preserves cardiac function in the face of chronic ischemic injury. Recent evidence suggested a vital role of aldehyde dehydrogenase 2 (ALDH2) in cardiac protection after ischemia. This study was designed to determine whether ALDH2 regulates chronic ischemia-induced angiogenesis and to explore the underlying mechanism involved. Moreover, the clinical impact of the ALDH2 mutant allele on the development of coronary collateral circulation (CCC) was evaluated. APPROACH AND RESULTS Mice limb ischemia was performed. Compared with wild-type, ALDH2 deletion significantly reduced perfusion recovery, small artery and capillary density, and increased muscle atrophy in this ischemic model. In vitro, ALDH2-knockdown reduced proliferation, migration and hypoxia triggered endothelial tube formation of endothelial cells, the effects of which were restored by ALDH2 transfection. Further examination revealed that ALDH2 regulated angiogenesis possibly through hypoxia-inducible factor-1α/vascular endothelial growth factor pathways. To further discern the role of ALDH2 deficiency in the function of bone marrow stem/progenitor cells, cross bone marrow transplantation was performed between wild-type and ALDH2-knockout mice. However, there was no significant improvement for wild-type bone marrow transplantation into knockout mice. ALDH2 genotyping was screened in 139 patients with chronic total occlusion recruited to Zhongshan Hospital (2011.10-2014.4). Patients with poor CCC (Rentrop 0-1; n=51) exhibited a higher frequency of the AA genotype than those with enriched CCC (Rentrop 2-3; n=88; 11.76% versus 1.14%; P=0 0.01). However, the AA group displayed less enriched CCC frequency in Logistic regression model when compared with the GG group (odds ratio=0.08; 95% confidence interval, 0.009-0.701; P=0 0.026). Furthermore, serum vascular endothelial growth factor level tended to be lower in patients with ALDH2 mutation. CONCLUSIONS This study demonstrated that ALDH2 possesses an intrinsic capacity to regulate angiogenesis via hypoxia-inducible factor-1α and vascular endothelial growth factor. Patients with ALDH2-deficient genotype displayed a higher risk of developing poor CCC. Therapeutic individualization based on ALDH2 allele distribution may thus improve the therapeutic benefit, especially in the East Asian decedents.
Collapse
Affiliation(s)
- Xiangwei Liu
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Xiaolei Sun
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Hua Liao
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Zhen Dong
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Jingjing Zhao
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Hong Zhu
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Peng Wang
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Li Shen
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Lei Xu
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Xin Ma
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Cheng Shen
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Fan Fan
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Cong Wang
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Kai Hu
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Yunzeng Zou
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Junbo Ge
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Jun Ren
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.)
| | - Aijun Sun
- From the Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital (X.L., H.Z., P.W., L.S., L.X., C.S., F.F., C.W., K.H., Y.Z., J.G., J.R., A.S.), Institute of Biomedical Science (X.S., L.X., X.M., Y.Z., J.G., A.S.), Department of Cardiology, Huashan Hospital (Z.D.), Fudan University, Shanghai, P.R. China; Center for Cardiovascular Research and Alternative Medicine, School of Pharmacy, University of Wyoming College of Health Sciences, Laramie (X.L., J.R.); Dongfang Hospital, Tongji University, Shanghai, P.R. China (H.L.); and Department of Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China (J.Z.).
| |
Collapse
|
166
|
Zhou BD, Guo G, Zheng LM, Zu LY, Gao W. Microparticles as novel biomarkers and therapeutic targets in coronary heart disease. Chin Med J (Engl) 2015; 128:267-72. [PMID: 25591573 PMCID: PMC4837849 DOI: 10.4103/0366-6999.149231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
| | | | | | - Ling-Yun Zu
- Department of Cardiology, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Peking University Third Hospital, Beijing 100191, China
| | | |
Collapse
|
167
|
Mendonça F, Soares R. Obesity and cancer phenotype: Is angiogenesis a missed link? Life Sci 2015; 139:16-23. [PMID: 26297445 DOI: 10.1016/j.lfs.2015.08.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 07/28/2015] [Accepted: 08/14/2015] [Indexed: 12/13/2022]
Abstract
Obesity remains nowadays one of the main threats to human health, being a problem of worldwide proportions. It is characterized by augmented storage of fatty acids in an enlarged adipose tissue. This process is possible thanks to a rich capillary network, supported by a mechanism that has also a crucial role on cancer: angiogenesis. Given that several studies point obesity as a risk factor for cancer development, angiogenesis may be approached as the missed link between these two pathologies. Understanding the different pathways behind angiogenesis becomes essential to break this link by developing new anti-angiogenic therapies or improving the actual ones. In the first phase, this paper will focus the structural and cellular changes that adipose tissue suffers in obesity. Then, the main pro-angiogenic players will be reviewed, taking into account the pathways that explain their putative role in obesity-cancer link. Finally, the clinical implications of the presented mechanisms will also be regarded, being the main focus on the anti-angiogenic therapies.
Collapse
Affiliation(s)
- Fernando Mendonça
- Department of Biochemistry, Faculty of Medicine, University of Porto, Portugal
| | - Raquel Soares
- Department of Biochemistry, Faculty of Medicine, University of Porto, Portugal; Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Portugal.
| |
Collapse
|
168
|
Awada HK, Johnson NR, Wang Y. Sequential delivery of angiogenic growth factors improves revascularization and heart function after myocardial infarction. J Control Release 2015; 207:7-17. [PMID: 25836592 PMCID: PMC4430430 DOI: 10.1016/j.jconrel.2015.03.034] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Revised: 02/17/2015] [Accepted: 03/30/2015] [Indexed: 12/28/2022]
Abstract
Treatment of ischemia through therapeutic angiogenesis faces significant challenges. Growth factor (GF)-based therapies can be more effective when concerns such as GF spatiotemporal presentation, bioactivity, bioavailability, and localization are addressed. During angiogenesis, vascular endothelial GF (VEGF) is required early to initiate neovessel formation while platelet-derived GF (PDGF-BB) is needed later to stabilize the neovessels. The spatiotemporal delivery of multiple bioactive GFs involved in angiogenesis, in a close mimic to physiological cues, holds great potential to treat ischemic diseases. To achieve sequential release of VEGF and PDGF, we embed VEGF in fibrin gel and PDGF in a heparin-based coacervate that is distributed in the same fibrin gel. In vitro, we show the benefits of this controlled delivery approach on cell proliferation, chemotaxis, and capillary formation. A rat myocardial infarction (MI) model demonstrated the effectiveness of this delivery system in improving cardiac function, ventricular wall thickness, angiogenesis, cardiac muscle survival, and reducing fibrosis and inflammation in the infarct zone compared to saline, empty vehicle, and free GFs. Collectively, our results show that this delivery approach mitigated the injury caused by MI and may serve as a new therapy to treat ischemic hearts pending further examination.
Collapse
Affiliation(s)
- Hassan K Awada
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Noah R Johnson
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Yadong Wang
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, PA 15261, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA.
| |
Collapse
|
169
|
OSM Enhances Angiogenesis and Improves Cardiac Function after Myocardial Infarction. BIOMED RESEARCH INTERNATIONAL 2015; 2015:317905. [PMID: 26146616 PMCID: PMC4471304 DOI: 10.1155/2015/317905] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 11/12/2014] [Accepted: 11/15/2014] [Indexed: 12/20/2022]
Abstract
Oncostatin M (OSM) has been reported to stimulate angiogenesis by upregulating VEGF and bFGF, implying that it could be a therapeutic strategy in treating ischemic diseases. The present study was aimed at investigating whether OSM could improve cardiac function via prompting angiogenesis following myocardial infarction (MI). Wild type (WT) and Oβ knock-out (Oβ−/−) mice were, respectively, randomized into sham group, MI + vehicle group, and MI + OSM group. WT mice displayed significantly impaired cardiac function after MI. OSM treatment attenuated cardiac dysfunction in WT MI mice, while Oβ deletion abrogated the protective effects. Besides, OSM attenuated heart hypertrophy and pulmonary congestion evidenced by decreased heart weight/body weight and lung weight/body weight ratio. Further, reduction of apoptosis and fibrosis in infarct border zone was observed in OSM treated WT MI mice compared with vehicle. Moreover, in WT mice subjected to MI, OSM treatment significantly increased capillary density along with upregulation of p-Akt and angiogenic factors VEGF and bFGF in comparison with vehicle, and this phenomenon was not found in Oβ−/− mice. In conclusion, OSM treatment preserved cardiac function, inhibited apoptosis and fibrosis, and stimulated angiogenesis via upregulating VEGF and bFGF in infarct border zone of ischemic myocardium, indicating that OSM could be a novel therapeutic target for MI.
Collapse
|
170
|
Hydrogen Sulfide as a Potential Therapeutic Target in Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:593407. [PMID: 26078809 PMCID: PMC4442300 DOI: 10.1155/2015/593407] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/29/2014] [Indexed: 12/24/2022]
Abstract
Hydrogen sulfide (H2S), produced endogenously by the activation of two major H2S-generating enzymes (cystathionine β-synthase and cystathionine γ-lyase), plays important regulatory roles in different physiologic and pathologic conditions. The abnormal metabolism of H2S is associated with fibrosis pathogenesis, causing damage in structure and function of different organs. A number of in vivo and in vitro studies have shown that both endogenous H2S level and the expressions of H2S-generating enzymes in plasma and tissues are significantly downregulated during fibrosis. Supplement with exogenous H2S mitigates the severity of fibrosis in various experimental animal models. The protective role of H2S in the development of fibrosis is primarily attributed to its antioxidation, antiapoptosis, anti-inflammation, proangiogenesis, and inhibition of fibroblasts activities. Future studies might focus on the potential to intervene fibrosis by targeting the pathway of endogenous H2S-producing enzymes and H2S itself.
Collapse
|
171
|
Wan N, Liu X, Zhang XJ, Zhao Y, Hu G, Wan F, Zhang R, Zhu X, Xia H, Li H. Toll-interacting protein contributes to mortality following myocardial infarction through promoting inflammation and apoptosis. Br J Pharmacol 2015; 172:3383-96. [PMID: 25765712 DOI: 10.1111/bph.13130] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 02/27/2015] [Accepted: 03/03/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Toll-interacting protein (Tollip) is an endogenous inhibitor of toll-like receptors, a superfamily that plays a pivotal role in various pathological conditions, including myocardial infarction (MI). However, the exact role of Tollip in MI remains unknown. EXPERIMENTAL APPROACH MI models were established in Tollip knockout (KO) mice, mice with cardiac-specific overexpression of human Tollip gene and in their Tollip(+/+) and non-transgenic controls respectively. The effects of Tollip on MI were evaluated by mortality, infarct size and cardiac function. Hypoxia-induced cardiomyocyte damage was investigated in vitro to confirm the role of Tollip in heart damage. KEY RESULTS Tollip expression was dramatically up-regulated in human ischaemic hearts and infarcted mice hearts. MI-induced mortality, infarct size and cardiac dysfunction were decreased in Tollip-KO mice compared with Tollip(+/+) controls. Ischaemic hearts from Tollip-KO mice exhibited decreased inflammatory cell infiltration and reduced NF-κB activation. Tollip depletion also alleviated myocardial apoptosis by down-regulating pro-apoptotic protein levels and up-regulating anti-apoptotic protein expressions in infarct border zone. Conversely, MI effects were exacerbated in mice with cardiac-specific Tollip overexpression. This aggravated MI injury by Tollip in vivo was confirmed with in vitro assays. Inhibition of Akt signalling was associated with the detrimental effects of Tollip on MI injury; activation of Akt largely reversed the deleterious effects of Tollip on MI-induced cardiomyocyte death. CONCLUSIONS AND IMPLICATIONS Tollip promotes inflammatory and apoptotic responses after MI, leading to increased mortality and aggravated cardiac dysfunction. These findings suggest that Tollip may serve as a novel therapeutic target for the treatment of MI.
Collapse
Affiliation(s)
- Nian Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Xiaoxiong Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Yichao Zhao
- Department of Cardiology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Gangying Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Fengwei Wan
- Department of Emergency, The Second Artillery General Hospital of Chinese People's Liberation Army Qinghe Clinic, Beijing, China
| | - Rui Zhang
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Xueyong Zhu
- Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Hao Xia
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Cardiovascular Research Institute of Wuhan University, Wuhan, China
| |
Collapse
|
172
|
Pascual-Gil S, Garbayo E, Díaz-Herráez P, Prosper F, Blanco-Prieto M. Heart regeneration after myocardial infarction using synthetic biomaterials. J Control Release 2015; 203:23-38. [DOI: 10.1016/j.jconrel.2015.02.009] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 02/03/2015] [Accepted: 02/04/2015] [Indexed: 12/24/2022]
|
173
|
Qu Z, Xu H, Tian Y. Effects of angiotensin-converting enzyme inhibition and bradykinin peptides in rats with myocardial infarction. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:3410-3417. [PMID: 26045879 PMCID: PMC4440188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/11/2015] [Accepted: 02/28/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND AND OBJECTIVE Angiotensin-converting enzyme (ACE) inhibitors have been reported to decrease myocardial remodeling and faciliate cardiac function improvement in the setting myocardial infarction by affecting bradykinin. The purpose of this study was to evaluate the combination effects of perindopril and bradykinin (BK) in rats with myocardial infarction. METHODS Wistar Rats underwent to left anterior descending (LAD) coronary artery ligation were allocated into MI group (n=6); Perindopril group (n=7); Perindopril+BK group (n=7). An additional sham operation group (Sham group, n=6) were also established. After 4 weeks, the left ventricle function, myocardial tissue morphology, myocardial collagen volume faction, infracted ventricular wall thickness, myocardial infarction area and neovascular formation were evaluated. RESULTS Combination treatment with perindopril and BK were showed significant improvement on LVEDV, LVEF and LVFS than MI group. Moreover, a significant improvement on LVEF was found in Perindopril+BK group than Perindopril group but not on LVEDV and LVFS between these two groups. Furthermore, neo-vessel density was significantly increased in Perindopril+BK group than other groups while no significant improvement on vessel density was found after the treatment of perindopril. In addition, myocardial infarction thickness improvement was found in Perindopril and group than MI group while combination treatment with perindopril and BK can significant improve the myocardial infarction thickness than perindopril only. CONCLUSIONS Combination treatment with ACE inhibitor perindopril and BK can significantly improve the ventricle function in the rat model of myocardial infarction. Our data suggest BK can serve as adjuvant treatment in myocardial infarction treatment.
Collapse
Affiliation(s)
- Zhe Qu
- Department of Cardiology, Renmin Hospital of Wuhan UniversityWuhan 430060, China
| | - Hongxin Xu
- Department of Cardiology, Renmin Hospital of Wuhan UniversityWuhan 430060, China
| | - Yihao Tian
- Department of Cardiology, Renmin Hospital of Wuhan UniversityWuhan 430060, China
- Department of Anatomy, Basic Medical College of Wuhan UniversityWuhan 430071, China
| |
Collapse
|
174
|
Oxidative stress-induced apoptotic insults to rat osteoblasts are attenuated by nitric oxide pretreatment via GATA-5-involved regulation of Bcl-X L gene expression and protein translocation. Arch Toxicol 2015; 90:905-16. [DOI: 10.1007/s00204-015-1491-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 02/23/2015] [Indexed: 12/15/2022]
|
175
|
Daskalopoulos EP, Vilaeti AD, Barka E, Mantzouratou P, Kouroupis D, Kontonika M, Tourmousoglou C, Papalois A, Pantos C, Blankesteijn WM, Agathopoulos S, Kolettis TM. Attenuation of post-infarction remodeling in rats by sustained myocardial growth hormone administration. Growth Factors 2015; 33:250-8. [PMID: 26290214 DOI: 10.3109/08977194.2015.1072527] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Prevention of left ventricular remodeling is an important therapeutic target post-myocardial infarction. Experimentally, treatment with growth hormone (GH) is beneficial, but sustained local administration has not been thoroughly investigated. We studied 58 rats (322 ± 4 g). GH was administered via a biomaterial-scaffold, following in vitro and in vivo evaluation of degradation and drug-release curves. Treatment consisted of intra-myocardial injection of saline or alginate-hydrogel, with or without GH, 10 min after permanent coronary artery ligation. Echocardiographic and histologic remodeling-indices were examined 3 weeks post-ligation, followed by immunohistochemical evaluation of angiogenesis, collagen, macrophages and myofibroblasts. GH-release completed at 3 days and alginate-degradation at ∼7 days. Alginate + GH consistently improved left ventricular end-diastolic and end-systolic diameters, ventricular sphericity, wall tension index and infarct-thickness. Microvascular-density and myofibroblast-count in the infarct and peri-infarct areas were higher after alginate + GH. Macrophage-count and collagen-content did not differ between groups. Early, sustained GH-administration enhances angiogenesis and myofibroblast-activation and ameliorates post-infarction remodeling.
Collapse
Affiliation(s)
- Evangelos P Daskalopoulos
- a Department of Pharmacology , Cardiovascular Research Institute Maastricht, Maastricht University , Maastricht , The Netherlands
- b Cardiovascular Research Institute , Ioannina , Athens , Greece
| | - Agapi D Vilaeti
- b Cardiovascular Research Institute , Ioannina , Athens , Greece
| | - Eleonora Barka
- b Cardiovascular Research Institute , Ioannina , Athens , Greece
- c Ceramics and Composites Laboratory, Department of Materials Science and Engineering, University of Ioannina , Ioannina , Greece
| | - Polixeni Mantzouratou
- d Department of Pharmacology , Medical School, University of Athens , Athens , Greece
| | - Dimitrios Kouroupis
- e Department of Biomedical Research, Foundation for Research and Technology-Hellas , Institute of Molecular Biology and Biotechnology , Ioannina , Greece , and
| | | | | | - Apostolos Papalois
- b Cardiovascular Research Institute , Ioannina , Athens , Greece
- f Experimental Research Center ELPEN , Pikermi , Athens , Greece
| | - Constantinos Pantos
- d Department of Pharmacology , Medical School, University of Athens , Athens , Greece
| | - W Matthijs Blankesteijn
- a Department of Pharmacology , Cardiovascular Research Institute Maastricht, Maastricht University , Maastricht , The Netherlands
| | - Simeon Agathopoulos
- c Ceramics and Composites Laboratory, Department of Materials Science and Engineering, University of Ioannina , Ioannina , Greece
| | | |
Collapse
|
176
|
Cao J, Tsenovoy PL, Thompson EA, Falck JR, Touchon R, Sodhi K, Rezzani R, Shapiro JI, Abraham NG. Agonists of epoxyeicosatrienoic acids reduce infarct size and ameliorate cardiac dysfunction via activation of HO-1 and Wnt1 canonical pathway. Prostaglandins Other Lipid Mediat 2015; 116-117:76-86. [PMID: 25677507 PMCID: PMC5553685 DOI: 10.1016/j.prostaglandins.2015.01.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 01/15/2015] [Accepted: 01/16/2015] [Indexed: 12/27/2022]
Abstract
Myocardial infarction (MI) is complicated by ventricular fibrosis and associated diastolic and systolic failure. Emerging studies implicate Wnt1 signaling in the formation of new blood vessels. Epoxyeicosatrienoic acids (EETs)-mediated up-regulation of heme oxygenase-1 (HO-1) protects against the detrimental consequences of MI in several animal models, however, the mechanism(s) by which this occurs remains unclear. The aim of this study was to examine these mechanisms in the LAD ligation animal model of post infarcted heart failure. Specifically, we sought to clarify the mechanistic basis of the interactions of the Wnt1 canonical pathway, HO-1 and associated angiogenesis. Human microvascular endothelial cells (HMECs) were exposed to anoxia and treated with the EET agonist, NUDSA, in the presence and absence of tin mesoporphyrin (SnMP). Increased capillary density, and Wnt1 and HO-1 expression occurred in cells treated with NUDSA. Anoxic HMECs treated with NUDSA and Wnt1 siRNA, exhibited decreased in the expression of β-catenin and the Wnt1 target gene, PPARδ (p<0.05 vs. NUDSA). Furthermore, blocking the Wnt 1 antagonist, Dickkopf 1, by siRNA increased β-catenin and PPARδ expression, and this effect was further enhanced by the concurrent administration of NUDSA. In in vivo experiments, C57B16 mice were divided into 4 groups: sham, mice with MI via LAD ligation and mice with MI treated with NUDSA, with and without SnMP. Increased fractional area change (FAC) and myocardial angiogenesis were observed in mice treated with NUDSA (p<0.05 vs. MI). Increased expression of HO-1, Wnt1, β-catenin, adiponectin, and phospho-endothelial nitric oxide synthetase (p-eNOS), and a decrease in the glycosylated subunit of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase, gp91(phox) expression occurred in cardiac tissue of mice treated with NUDSA (p<0.05 vs. MI). SnMP reversed these effects. This novel study demonstrates that increasing the canonical Wnt1 signaling cascade with the subsequent increase in HO-1, adiponectin and angiogenesis ameliorates fibrosis and cardiac dysfunction in a mouse model of MI and supports the hypothesis that HO-1 is an integral component of the EETs-adiponectin axis and is central for the control of resistance to fibrosis and vascular dysfunction and in part determine how they influence the cellular/vascular homeostasis and provides insight into the mechanisms involved in vascular dysfunction as well as potential targets for the treatment of CVD.
Collapse
Affiliation(s)
- Jian Cao
- Chinese PLA General Hospital, Beijing 100853, China
| | | | - Ellen A Thompson
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, United States
| | - John R Falck
- University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, United States
| | - Robert Touchon
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, United States
| | - Komal Sodhi
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, United States
| | - Rita Rezzani
- New York Medical College, Valhalla, NY, United States
| | - Joseph I Shapiro
- Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, United States
| | - Nader G Abraham
- New York Medical College, Valhalla, NY, United States; Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25701, United States.
| |
Collapse
|
177
|
Michelis KC, Boehm M, Kovacic JC. New vessel formation in the context of cardiomyocyte regeneration--the role and importance of an adequate perfusing vasculature. Stem Cell Res 2014; 13:666-82. [PMID: 24841067 PMCID: PMC4213356 DOI: 10.1016/j.scr.2014.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 03/16/2014] [Accepted: 04/18/2014] [Indexed: 02/08/2023] Open
Abstract
The history of revascularization for cardiac ischemia dates back to the early 1960's when the first coronary artery bypass graft procedures were performed in humans. With this 50 year history of providing a new vasculature to ischemic and hibernating myocardium, a profound depth of experience has been amassed in clinical cardiovascular medicine as to what does, and does not work in the context of cardiac revascularization, alleviating ischemia and adequacy of myocardial perfusion. These issues are of central relevance to contemporary cell-based cardiac regenerative approaches. While the cardiovascular cell therapy field is surging forward on many exciting fronts, several well accepted clinical axioms related to the cardiac arterial supply appear to be almost overlooked by some of our current basic conceptual and experimental cell therapy paradigms. We present here information drawn from five decades of the clinical revascularization experience, review relevant new data on vascular formation via cell therapy, and put forward the case that for optimal cell-based cardiac regeneration due attention must be paid to providing an adequate vascular supply.
Collapse
Affiliation(s)
- Katherine C Michelis
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manfred Boehm
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jason C Kovacic
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
178
|
Wen Z, Huang W, Feng Y, Cai W, Wang Y, Wang X, Liang J, Wani M, Chen J, Zhu P, Chen JM, Millard RW, Fan GC, Wang Y. MicroRNA-377 regulates mesenchymal stem cell-induced angiogenesis in ischemic hearts by targeting VEGF. PLoS One 2014; 9:e104666. [PMID: 25251394 PMCID: PMC4174502 DOI: 10.1371/journal.pone.0104666] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 07/10/2014] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs have been appreciated in various cellular functions, including the regulation of angiogenesis. Mesenchymal-stem-cells (MSCs) transplanted to the MI heart improve cardiac function through paracrine-mediated angiogenesis. However, whether microRNAs regulate MSC induced angiogenesis remains to be clarified. Using microRNA microarray analysis, we identified a microRNA expression profile in hypoxia-treated MSCs and observed that among all dysregulated microRNAs, microRNA-377 was decreased the most significantly. We also validated that vascular endothelial growth factor (VEGF) is a target of microRNA-377 using dual-luciferase reporter assay and Western-blotting. Knockdown of endogenous microRNA-377 promoted tube formation in human umbilical vein endothelial cells. We then engineered rat MSCs with lentiviral vectors to either overexpress microRNA-377 (MSC miR-377) or knockdown microRNA-377 (MSC Anti-377) to investigate whether microRNA-377 regulated MSC-induced myocardial angiogenesis, using MSCs infected with lentiviral empty vector to serve as controls (MSC Null). Four weeks after implantation of the microRNA-engineered MSCs into the infarcted rat hearts, the vessel density was significantly increased in MSC Anti-377-hearts, and this was accompanied by reduced fibrosis and improved myocardial function as compared to controls. Adverse effects were observed in MSC miR-377-treated hearts, including reduced vessel density, impaired myocardial function, and increased fibrosis in comparison with MSC Null-group. These findings indicate that hypoxia-responsive microRNA-377 directly targets VEGF in MSCs, and knockdown of endogenous microRNA-377 promotes MSC-induced angiogenesis in the infarcted myocardium. Thus, microRNA-377 may serve as a novel therapeutic target for stem cell-based treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Zhili Wen
- Department of Infectious Disease, Nanchang University Medical School, Nanchang, Jiangxi, China
- Department of Pathology and Lab Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Wei Huang
- Department of Pathology and Lab Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Yuliang Feng
- Department of Pathology and Lab Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Wenfeng Cai
- Department of Pathology and Lab Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Yuhua Wang
- Department of Pathology and Lab Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Xiaohong Wang
- Department of Pharmacology and Cell Biophysics, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Jialiang Liang
- Department of Pathology and Lab Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Mashhood Wani
- Department of Pathology and Lab Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Jing Chen
- Department of Environmental Health, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Pin Zhu
- Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, Guandong, People’s Republic of China
| | - Ji-Mei Chen
- Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Guangzhou, Guandong, People’s Republic of China
| | - Ronald W. Millard
- Department of Pharmacology and Cell Biophysics, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
| | - Yigang Wang
- Department of Pathology and Lab Medicine, University of Cincinnati Medical Center, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
179
|
Keszler A, Brandal G, Baumgardt S, Ge ZD, Pratt PF, Riess ML, Bienengraeber M. Far red/near infrared light-induced protection against cardiac ischemia and reperfusion injury remains intact under diabetic conditions and is independent of nitric oxide synthase. Front Physiol 2014; 5:305. [PMID: 25202275 PMCID: PMC4141548 DOI: 10.3389/fphys.2014.00305] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Accepted: 07/26/2014] [Indexed: 12/27/2022] Open
Abstract
Far red/near-infrared light (NIR) promotes a wide range of biological effects including tissue protection but whether and how NIR is capable of acutely protecting myocardium against ischemia and reperfusion injury in vivo is not fully elucidated. Our previous work indicates that NIR exposure immediately before and during early reperfusion protects the myocardium against infarction through mechanisms that are nitric oxide (NO)-dependent. Here we tested the hypothesis that NIR elicits protection in a diabetic mouse model where other cardioprotective interventions such as pre- and postconditioning fail, and that the protection is independent of nitric oxide synthase (NOS). NIR reduced infarct size dose dependently. Importantly, NIR-induced protection was preserved in a diabetic mouse model (db/db) and during acute hyperglycemia, as well as in endothelial NOS(-/-) mice and in wild type mice treated with NOS inhibitor L-NAME. In in vitro experiments NIR light liberates NO from nitrosyl hemoglobin (HbNO) and nitrosyl myoglobin (MbNO) in a wavelength-(660-830 nm) and dose-dependent manner. Irradiation at 660 nm yields the highest release of NO, while at longer wavelengths a dramatic decrease of NO release can be observed. Similar wavelength dependence was observed for the protection of mice against cardiac ischemia and reperfusion injury in vivo. NIR-induced NO release from deoxymyoglobin in the presence of nitrite mildly inhibits respiration of isolated mitochondria after hypoxia. In summary, NIR applied during reperfusion protects the myocardium against infarction in an NO-dependent, but NOS-independent mechanisms, whereby mitochondria may be a target of NO released by NIR, leading to reduced reactive oxygen species generation during reperfusion. This unique mechanism preserves protection even during diabetes where other protective strategies fail.
Collapse
Affiliation(s)
- Agnes Keszler
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Garth Brandal
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Shelley Baumgardt
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Zhi-Dong Ge
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Phillip F. Pratt
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Matthias L. Riess
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
- Department of Anesthesiology, Clement J. Zablocki VA Medical CenterMilwaukee, WI, USA
- Department of Physiology, Medical College of WisconsinMilwaukee, WI, USA
| | - Martin Bienengraeber
- Department of Anesthesiology, Medical College of WisconsinMilwaukee, WI, USA
- Department of Pharmacology and Toxicology, Medical College of WisconsinMilwaukee, WI, USA
| |
Collapse
|
180
|
Lee TH, Jung H, Park KH, Bang MH, Baek NI, Kim J. Jaceosidin, a natural flavone, promotes angiogenesis via activation of VEGFR2/FAK/PI3K/AKT/NF-κB signaling pathways in endothelial cells. Exp Biol Med (Maywood) 2014; 239:1325-34. [DOI: 10.1177/1535370214533883] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Angiogenesis, the growth of new blood vessels from pre-existing vasculature, plays an important role in physiological and pathological processes such as embryonic development wound healing and revascularization of tissues after exposure to ischemia. We investigated the effects of jaceosidin, a main constituent of medicinal herbs of the genus Artemisia, on angiogenesis and signaling pathways in endothelial cells. Jaceosidin stimulated proliferation, migration and tubulogenesis of ECs as well as ex vivo sprouting from aorta rings, which are phenomena typical of angiogenesis. Jaceosidin activated vascular endothelial growth factor receptor 2 (VEGFR2, FLk-1/KDR) and angiogenic signaling molecules such as focal adhesion kinase, phosphatidylinositol 3-kinase, and its downstream target, the serine-threonine kinase AKTWe also demonstrated that jaceosidin activated the NF-κB-driven expression of a luciferase reporter gene and NF-κB binding to DNA. Jaceosidin-induced proliferation and migration of human umbilical vascular endothelial cells were strongly inhibited by the phosphatidylinositol 3-kinase inhibitor LY294002 and NF-κB inhibitor BAY11-7082, indicating that the PI3K/AKT/NF-κB signaling pathway is involved in jaceosidin-induced angiogenesis. Our results suggest that jaceosidin stimulates angiogenesis by activating the VEGFR2/FAK/PI3K/AKT/NF-κB signaling pathway and that it may be useful in developing angiogenic agents to promote the growth of collateral blood vessels in ischemic tissues.
Collapse
Affiliation(s)
- Tae Hoon Lee
- Graduate School of Biotechnology and College of Life Sciences, Kyung Hee University, Yongin 446-701, Korea
| | - Hana Jung
- Graduate School of Biotechnology and College of Life Sciences, Kyung Hee University, Yongin 446-701, Korea
| | - Keun Hyung Park
- Graduate School of Biotechnology and College of Life Sciences, Kyung Hee University, Yongin 446-701, Korea
| | - Myun Ho Bang
- Graduate School of Biotechnology and College of Life Sciences, Kyung Hee University, Yongin 446-701, Korea
| | - Nam-In Baek
- Graduate School of Biotechnology and College of Life Sciences, Kyung Hee University, Yongin 446-701, Korea
| | - Jiyoung Kim
- Graduate School of Biotechnology and College of Life Sciences, Kyung Hee University, Yongin 446-701, Korea
| |
Collapse
|
181
|
Zhang H, van Olden C, Sweeney D, Martin-Rendon E. Blood vessel repair and regeneration in the ischaemic heart. Open Heart 2014; 1:e000016. [PMID: 25332783 PMCID: PMC4189230 DOI: 10.1136/openhrt-2013-000016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 12/11/2013] [Accepted: 12/15/2013] [Indexed: 12/19/2022] Open
Abstract
The term 'therapeutic angiogenesis' originated almost two decades ago, following evidence that factors that promote blood vessel formation could be delivered to ischaemic tissues and restore blood flow. Following this proof-of-principle, safety and efficacy of the best-studied angiogenic factors (eg, vascular endothelial growth factor) were demonstrated in early clinical studies. Promising results led to the development of larger controlled trials that, unfortunately, have failed to satisfy the initial expectations of therapeutic angiogenesis for ischaemic heart disease. As the quest to delay the progression to heart failure secondary to ischaemic heart disease continues, alternative therapies have emerged as potential novel treatments to improve myocardial reperfusion and long-term heart function. The disappointing results of the clinical studies using angiogenic factors were followed by mixed results from the cell therapy trials. This review reflects the current angiogenic strategies for the ischaemic heart, their limitations and discusses future perspectives in the light of recent scientific and clinical evidence. It is proposed that combination therapies may be a new direction to advance therapeutic repair and regeneration of blood vessels in the ischaemic heart.
Collapse
Affiliation(s)
- Huajun Zhang
- Nuffield Department of Surgical Sciences , University of Oxford , Oxford , UK ; Stem Cell Research Laboratory , NHS Blood and Transplant, John Radcliife Hospital , Oxford , UK
| | - Casper van Olden
- Nuffield Department of Surgical Sciences , University of Oxford , Oxford , UK ; Stem Cell Research Laboratory , NHS Blood and Transplant, John Radcliife Hospital , Oxford , UK
| | - Dominic Sweeney
- Stem Cell Research Laboratory , NHS Blood and Transplant, John Radcliife Hospital , Oxford , UK ; Radcliffe Department of Medicine , University of Oxford , Oxford , UK
| | - Enca Martin-Rendon
- Stem Cell Research Laboratory , NHS Blood and Transplant, John Radcliife Hospital , Oxford , UK ; Radcliffe Department of Medicine , University of Oxford , Oxford , UK
| |
Collapse
|
182
|
Mahmoodzadeh S, Leber J, Zhang X, Jaisser F, Messaoudi S, Morano I, Furth PA, Dworatzek E, Regitz-Zagrosek V. Cardiomyocyte-specific Estrogen Receptor Alpha Increases Angiogenesis, Lymphangiogenesis and Reduces Fibrosis in the Female Mouse Heart Post-Myocardial Infarction. ACTA ACUST UNITED AC 2014; 5:153. [PMID: 24977106 DOI: 10.4172/2157-7013.1000153] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Experimental studies showed that 17β-estradiol (E2) and activated Estrogen Receptors (ER) protect the heart from ischemic injury. However, the underlying molecular mechanisms are not well understood. To investigate the role of ER-alpha (ERα) in cardiomyocytes in the setting of myocardial ischemia, we generated transgenic mice with cardiomyocyte-specific overexpression of ERα (ERα-OE) and subjected them to Myocardial Infarction (MI). At the basal level, female and male ERα-OE mice showed increased Left Ventricular (LV) mass, LV volume and cardiomyocyte length. Two weeks after MI, LV volume was significantly increased and LV wall thickness decreased in female and male WT-mice and male ERα-OE, but not in female ERα-OE mice. ERα-OE enhanced expression of angiogenesis and lymphangiogenesis markers (Vegf, Lyve-1), and neovascularization in the peri-infarct area in both sexes. However, attenuated level of fibrosis and higher phosphorylation of JNK signaling pathway could be detected only in female ERα-OE after MI. In conclusion, our study indicates that ERα protects female mouse cardiomyocytes from the sequelae of ischemia through induction of neovascularization in a paracrine fashion and impaired fibrosis, which together may contribute to the attenuation of cardiac remodelling.
Collapse
Affiliation(s)
- Shokoufeh Mahmoodzadeh
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin, Berlin, Germany
| | - Joachim Leber
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin, Berlin, Germany
| | - Xiang Zhang
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin, Berlin, Germany.,Department of Cardiology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | | | | | - Ingo Morano
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany
| | - Priscilla A Furth
- Departments of Oncology and Medicine and the Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Elke Dworatzek
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin, Berlin, Germany
| | - Vera Regitz-Zagrosek
- Institute of Gender in Medicine and Center for Cardiovascular Research, Charité Universitaetsmedizin, Berlin, Germany
| |
Collapse
|
183
|
Icli B, Wara AKM, Moslehi J, Sun X, Plovie E, Cahill M, Marchini JF, Schissler A, Padera RF, Shi J, Cheng HW, Raghuram S, Arany Z, Liao R, Croce K, MacRae C, Feinberg MW. MicroRNA-26a regulates pathological and physiological angiogenesis by targeting BMP/SMAD1 signaling. Circ Res 2013; 113:1231-41. [PMID: 24047927 DOI: 10.1161/circresaha.113.301780] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
RATIONALE The rapid induction and orchestration of new blood vessels are critical for tissue repair in response to injury, such as myocardial infarction, and for physiological angiogenic responses, such as embryonic development and exercise. OBJECTIVE We aimed to identify and characterize microRNAs (miR) that regulate pathological and physiological angiogenesis. METHODS AND RESULTS We show that miR-26a regulates pathological and physiological angiogenesis by targeting endothelial cell (EC) bone morphogenic protein/SMAD1 signaling in vitro and in vivo. MiR-26a expression is increased in a model of acute myocardial infarction in mice and in human subjects with acute coronary syndromes. Ectopic expression of miR-26a markedly induced EC cycle arrest and inhibited EC migration, sprouting angiogenesis, and network tube formation in matrigel, whereas blockade of miR-26a had the opposite effects. Mechanistic studies demonstrate that miR-26a inhibits the bone morphogenic protein/SMAD1 signaling pathway in ECs by binding to the SMAD1 3'-untranslated region, an effect that decreased expression of Id1 and increased p21(WAF/CIP) and p27. In zebrafish, miR-26a overexpression inhibited formation of the caudal vein plexus, a bone morphogenic protein-responsive process, an effect rescued by ectopic SMAD1 expression. In mice, miR-26a overexpression inhibited EC SMAD1 expression and exercise-induced angiogenesis. Furthermore, systemic intravenous administration of an miR-26a inhibitor, locked nucleic acid-anti-miR-26a, increased SMAD1 expression and rapidly induced robust angiogenesis within 2 days, an effect associated with reduced myocardial infarct size and improved heart function. CONCLUSIONS These findings establish miR-26a as a regulator of bone morphogenic protein/SMAD1-mediated EC angiogenic responses, and that manipulating miR-26a expression could provide a new target for rapid angiogenic therapy in ischemic disease states.
Collapse
Affiliation(s)
- Basak Icli
- From the Cardiovascular Division, Departments of Medicine (B.I., A.K.M.W., J.M., X.S., E.P., M.C., J.F.M., A.S., J.S., H.-W.C., R.L., K.C., C.M., M.W.F.) and Pathology (R.F.P.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; and Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA (S.R., Z.A.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Renault MA, Vandierdonck S, Chapouly C, Yu Y, Qin G, Metras A, Couffinhal T, Losordo DW, Yao Q, Reynaud A, Jaspard-Vinassa B, Belloc I, Desgranges C, Gadeau AP. Gli3 regulation of myogenesis is necessary for ischemia-induced angiogenesis. Circ Res 2013; 113:1148-58. [PMID: 24044950 DOI: 10.1161/circresaha.113.301546] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
RATIONALE A better understanding of the mechanism underlying skeletal muscle repair is required to develop therapies that promote tissue regeneration in adults. Hedgehog signaling has been shown previously to be involved in myogenesis and angiogenesis: 2 crucial processes for muscle development and regeneration. OBJECTIVE The objective of this study was to identify the role of the hedgehog transcription factor Gli3 in the cross-talk between angiogenesis and myogenesis in adults. METHODS AND RESULTS Using conditional knockout mice, we found that Gli3 deficiency in endothelial cells did not affect ischemic muscle repair, whereas in myocytes, Gli3 deficiency resulted in severely delayed ischemia-induced myogenesis. Moreover, angiogenesis was also significantly impaired in HSA-Cre(ERT2); Gli3(Flox/Flox) mice, demonstrating that impaired myogenesis indirectly affects ischemia-induced angiogenesis. The role of Gli3 in myocytes was then further investigated. We found that Gli3 promotes myoblast differentiation through myogenic factor 5 regulation. In addition, we found that Gli3 regulates several proangiogenic factors, including thymidine phosphorylase and angiopoietin-1 both in vitro and in vivo, which indirectly promote endothelial cell proliferation and arteriole formation. In addition, we found that Gli3 is upregulated in proliferating myoblasts by the cell cycle-associated transcription factor E2F1. CONCLUSIONS This study shows for the first time that Gli3-regulated postnatal myogenesis is necessary for muscle repair-associated angiogenesis. Most importantly, it implies that myogenesis drives angiogenesis in the setting of skeletal muscle repair and identifies Gli3 as a potential target for regenerative medicine.
Collapse
Affiliation(s)
- Marie-Ange Renault
- From the Université de Bordeaux, Adaptation cardiovasculaire à l'ischémie, U1034, Pessac, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
185
|
Abstract
Heart disease is a major cause of death and morbidity worldwide, particularly so in industrialized nations. A major contributor to its varied pathophysiology is abnormal oxidative/reductive and nitrosative stress. While oxidant-scavenging therapies for cardiovascular disease have been disappointing to date, new insights into the precise sources of oxidative/nitrosative stress, its compartmentalized targets, and the modulators designed to control it are paving the way for a very different approach. This issue of the journal presents a Forum highlighting recent work from an international network of investigators that focuses on the role of oxidative/nitrosative stress in the remodeling heart.
Collapse
|