1
|
Zhu Y, Su SA, Shen J, Ma H, Le J, Xie Y, Xiang M. Recent advances of the Ephrin and Eph family in cardiovascular development and pathologies. iScience 2024; 27:110556. [PMID: 39188984 PMCID: PMC11345580 DOI: 10.1016/j.isci.2024.110556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/28/2024] Open
Abstract
Erythropoietin-producing hepatoma (Eph) receptors, comprising the largest family of receptor tyrosine kinases (RTKs), exert profound influence on diverse biological processes and pathological conditions such as cancer. Interacting with their corresponding ligands, erythropoietin-producing hepatoma receptor interacting proteins (Ephrins), Eph receptors regulate crucial events like embryonic development, tissue boundary formation, and tumor cell survival. In addition to their well-established roles in embryonic development and cancers, emerging evidence highlights the pivotal contribution of the Ephrin/Eph family to cardiovascular physiology and pathology. Studies have elucidated their involvement in cardiovascular development, atherosclerosis, postnatal angiogenesis, and, more recently, cardiac fibrosis and calcification, suggesting a promising avenue for therapeutic interventions in cardiovascular diseases. There remains a need for a comprehensive synthesis of their collective impact in the cardiovascular context. By exploring the intricate interactions between Eph receptors, ephrins, and cardiovascular system, this review aims to provide a holistic understanding of their roles and therapeutic potential in cardiovascular health and diseases.
Collapse
Affiliation(s)
- Yuan Zhu
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Sheng-an Su
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Jian Shen
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Hong Ma
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Jixie Le
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Yao Xie
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| | - Meixiang Xiang
- Department of Cardiology, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou 310009, China
| |
Collapse
|
2
|
Zhu F, Chen J, Luo M, Yao D, Hu X, Guo Y. EphrinB2 promotes the human aortic smooth muscle cell growth and migration via mediating F-actin remodeling. Vascular 2023; 31:142-151. [PMID: 34854323 DOI: 10.1177/17085381211052196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES To evaluate the potential effect of EphrinB2 in human thoracic aortic dissection (TAD) and to illustrate the mechanisms governing the role of EphrinB2 in the growth of human aortic smooth muscle cells (HASMC). METHODS In the study, EphrinB2 expression was investigated by qRT-PCR and immunohistochemistry in 12 pairs of TAD and adjacent human tissues. HASMCs were used for in vitro experiments. Next, EphrinB2 overexpression and depletion in HASMCs were established by EphrinB2-overexpressing vectors and small interfering RNA, respectively. The transfection efficiency was evaluated by qRT-PCR and Western blot. The effects of overexpression and depletion of EphrinB2 on cell proliferation, migration, and invasion were tested in vitro. Cell Counting Kit-8, flow cytometry and transwell migration/invasion, and wound healing assay were used to explore the function of EphrinB2 on HASMC cell lines. The relationship between EphrinB2 and F-actin was assessed by Western blot, immunofluorescence, and Co-IP. RESULTS We found that EphrinB2 was a prognostic biomarker of TAD patients. Moreover, EphrinB2 expression negatively correlated to aortic dissection tissues, and disease incidence of males, suggesting that EphrinB2 might act as a TAD suppressor by promoting proliferation or decreasing apoptosis in HASMC. Next, over-expression of EphrinB2 in HASMC lines drove cell proliferation, migration, and invasion, and inhibited apoptosis while knockdown EphrinB2 showed the opposite phenomenon, respectively. Furthermore, the level of F-actin in mRNA, protein, and distribution in HASMC cell lines highly matched with the expression of EphrinB2, which indicated that EphrinB2 could mediate the HASMC cytoskeleton via inducing F-actin. CONCLUSIONS In conclusion, our results first provided the pivotal role of EphrinB2 in HASMC proliferation initiated by mediating F-actin and demonstrated a prognostic biomarker and the potential targets for therapy to prevent thoracic aortic dissection.
Collapse
Affiliation(s)
- Fan Zhu
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Disease Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Jia Chen
- Department of Laboratory Medicine, Longhua Hospital, 74754Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingyao Luo
- State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, 34736Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dongting Yao
- Department of Laboratory Medicine, Longhua Hospital, 74754Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaobo Hu
- Department of Laboratory Medicine, Longhua Hospital, 74754Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanyuan Guo
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Disease Hospital, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
3
|
Zheng S, Sun F, Tian X, Zhu Z, Wang Y, Zheng W, Liu T, Wang W. Roles of Eph/ephrin signaling pathway in repair and regeneration for ischemic cerebrovascular and cardiovascular diseases. JOURNAL OF NEURORESTORATOLOGY 2022. [DOI: 10.1016/j.jnrt.2022.100040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
4
|
Baek KI, Chang SS, Chang CC, Roustaei M, Ding Y, Wang Y, Chen J, O'Donnell R, Chen H, Ashby JW, Xu X, Mack JJ, Cavallero S, Roper M, Hsiai TK. Vascular Injury in the Zebrafish Tail Modulates Blood Flow and Peak Wall Shear Stress to Restore Embryonic Circular Network. Front Cardiovasc Med 2022; 9:841101. [PMID: 35369301 PMCID: PMC8971683 DOI: 10.3389/fcvm.2022.841101] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/21/2022] [Indexed: 12/16/2022] Open
Abstract
Mechano-responsive signaling pathways enable blood vessels within a connected network to structurally adapt to partition of blood flow between organ systems. Wall shear stress (WSS) modulates endothelial cell proliferation and arteriovenous specification. Here, we study vascular regeneration in a zebrafish model by using tail amputation to disrupt the embryonic circulatory loop (ECL) at 3 days post fertilization (dpf). We observed a local increase in blood flow and peak WSS in the Segmental Artery (SeA) immediately adjacent to the amputation site. By manipulating blood flow and WSS via changes in blood viscosity and myocardial contractility, we show that the angiogenic Notch-ephrinb2 cascade is hemodynamically activated in the SeA to guide arteriogenesis and network reconnection. Taken together, ECL amputation induces changes in microvascular topology to partition blood flow and increase WSS-mediated Notch-ephrinb2 pathway, promoting new vascular arterial loop formation and restoring microcirculation.
Collapse
Affiliation(s)
- Kyung In Baek
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shyr-Shea Chang
- Department of Mathematics, University of California, Los Angeles, Los Angeles, CA, United States
- Center for Studies in Physics and Biology, The Rockefeller University, New York, NY, United States
- Developmental Biology Program, Sloan Kettering Institute, New York, NY, United States
| | - Chih-Chiang Chang
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Mehrdad Roustaei
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yichen Ding
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Yixuan Wang
- Department of Mathematics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Justin Chen
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Ryan O'Donnell
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
| | - Hong Chen
- Vascular Biology Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Julianne W. Ashby
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Xiaolei Xu
- Zebrafish Genetics, Mayo Clinic, Rochester, MN, United States
| | - Julia J. Mack
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Susana Cavallero
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Marcus Roper
- Department of Mathematics, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tzung K. Hsiai
- Department of Medicine and Bioengineering, University of California, Los Angeles, Los Angeles, CA, United States
- Division of Cardiology, Department of Medicine, School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
5
|
Nicin L, Schroeter SM, Glaser SF, Schulze-Brüning R, Pham MD, Hille SS, Yekelchyk M, Kattih B, Abplanalp WT, Tombor L, Müller OJ, Braun T, Meder B, Reich C, Arsalan M, Holubec T, Walther T, Emrich F, Krishnan J, Zeiher AM, John D, Dimmeler S. A human cell atlas of the pressure-induced hypertrophic heart. NATURE CARDIOVASCULAR RESEARCH 2022; 1:174-185. [PMID: 39195989 PMCID: PMC11357985 DOI: 10.1038/s44161-022-00019-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 01/05/2022] [Indexed: 08/29/2024]
Abstract
Pathological cardiac hypertrophy is a leading cause of heart failure, but knowledge of the full repertoire of cardiac cells and their gene expression profiles in the human hypertrophic heart is missing. Here, by using large-scale single-nucleus transcriptomics, we present the transcriptional response of human cardiomyocytes to pressure overload caused by aortic valve stenosis and describe major alterations in cardiac cellular crosstalk. Hypertrophied cardiomyocytes had reduced input from endothelial cells and fibroblasts. Genes encoding Eph receptor tyrosine kinases, particularly EPHB1, were significantly downregulated in cardiomyocytes of the hypertrophied heart. Consequently, EPHB1 activation by its ligand ephrin (EFN)B2, which is mainly expressed by endothelial cells, was reduced. EFNB2 inhibited cardiomyocyte hypertrophy in vitro, while silencing its expression in endothelial cells induced hypertrophy in co-cultured cardiomyocytes. Our human cell atlas of the hypertrophied heart highlights the importance of intercellular crosstalk in disease pathogenesis and provides a valuable resource.
Collapse
Affiliation(s)
- Luka Nicin
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Sam Michael Schroeter
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Simone Franziska Glaser
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Ralf Schulze-Brüning
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
| | - Minh-Duc Pham
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
- Cardiac Metabolism Group, Department of Cardiology, Goethe University Frankfurt, Frankfurt, Germany
| | - Susanne S Hille
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, Kiel, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Berlin, Germany
| | - Michail Yekelchyk
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Badder Kattih
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Wesley Tyler Abplanalp
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Lukas Tombor
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, University Hospital Schleswig-Holstein, Kiel, Germany
- German Center for Cardiovascular Research (DZHK), partner site Hamburg/Kiel/Lübeck, Berlin, Germany
| | - Thomas Braun
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
- Department of Cardiac Development and Remodelling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Benjamin Meder
- Institute for Cardiomyopathies, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Berlin, Germany
| | - Christoph Reich
- Institute for Cardiomyopathies, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Cardiovascular Research (DZHK), partner site Heidelberg/Mannheim, Berlin, Germany
| | - Mani Arsalan
- Department of Cardiovascular Surgery, Goethe University Hospital, Frankfurt, Germany
| | - Tomas Holubec
- Department of Cardiovascular Surgery, Goethe University Hospital, Frankfurt, Germany
| | - Thomas Walther
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
- Department of Cardiovascular Surgery, Goethe University Hospital, Frankfurt, Germany
| | - Fabian Emrich
- Department of Cardiovascular Surgery, Goethe University Hospital, Frankfurt, Germany
| | - Jaya Krishnan
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
- Cardiac Metabolism Group, Department of Cardiology, Goethe University Frankfurt, Frankfurt, Germany
| | - Andreas M Zeiher
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - David John
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration, Goethe University Frankfurt, Frankfurt, Germany.
- German Center for Cardiovascular Research (DZHK), partner site Frankfurt Rhine-Main, Berlin, Germany.
- Cardiopulmonary Institute, Goethe University Frankfurt, Frankfurt, Germany.
| |
Collapse
|
6
|
Wang Y, Shao C, Qi L, Tan J, Zhao Y, Xue M, Li X, Cheng W, Li X, Yin J, Shi Y, Wang Y, Wang K, Hu H, Yan S. EphrinB2-RhoA upregulation attenuates sympathetic hyperinnervation and decreases the incidence of ventricular arrhythmia after myocardial infarction. J Cardiol 2021; 79:423-431. [PMID: 34750029 DOI: 10.1016/j.jjcc.2021.10.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 09/28/2021] [Accepted: 10/06/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Cardiac sympathetic hyperinnervation after myocardial infarction (MI) is associated with a high incidence of lethal arrhythmia. Erythropoietin-producing hepatoma interactor B2 (EphrinB2), a diffusible axonal chemorepellent that can induce growth cone collapse and axon repulsion of several neuronal populations, is crucial in neurodevelopment during disease development and progression. However, whether EphrinB2 could inhibit cardiac sympathetic hyperinnervation after MI remains unclear. METHODS AND RESULTS A rat model of MI was developed by left anterior descending coronary artery ligation. EphrinB2 expression was markedly increased in the infarcted border at 3 days after MI. Downregulation of EphrinB2 by intramyocardial injection of lentivirus carrying EphrinB2-shRNA significantly increased sympathetic hyperinnervation along with downregulated RhoA expression. In contrast, injection of EphrinB2-overexpressing lentivirus markedly upregulated EphrinB2, concomitant with inhibition of sympathetic sprouting and upregulated RhoA expression, accompanied by decreased incidence of ventricular arrhythmias (VAs). However, co-administering EphrinB2-overexpressing lentivirus and Fasudil (Rho kinase inhibitor) nearly abolished the inhibition of nerve sprouting effect. Additionally, EphrinB2 expression did not affect nerve growth factor level in the infarcted heart. CONCLUSIONS Overexpression of EphrinB2 may ameliorate MI-induced sympathetic hyperinnervation and further reduce the incidence of VAs, at least in part by activating RhoA-mediated axonal retraction.
Collapse
Affiliation(s)
- Ye Wang
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | | | - Lei Qi
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Jiayu Tan
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Yuepeng Zhao
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Mei Xue
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Xiaolu Li
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Wenjuan Cheng
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Xinran Li
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Jie Yin
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Yugen Shi
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Yu Wang
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
| | - Kang Wang
- Department of Cardiology, Shandong Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hesheng Hu
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China.
| | - Suhua Yan
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China.
| |
Collapse
|
7
|
EphA4 is highly expressed in the atria of heart and its deletion leads to atrial hypertrophy and electrocardiographic abnormalities in rats. Life Sci 2021; 278:119595. [PMID: 33974931 DOI: 10.1016/j.lfs.2021.119595] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/25/2021] [Accepted: 05/03/2021] [Indexed: 01/12/2023]
Abstract
AIMS EphA4 is a member of the Eph receptor family, and expressed mainly in central nervous system (CNS), which is involved in CNS development and multiple diseases. Due to the variability in EphA4 expression, we wondered if EphA4 is expressed in other tissues, and what role does EphA4 play? MATERIALS AND METHODS We generated an EphA4 knockout (KO) rat line with red fluorescent marker protein encoded by the mCherry cassette inserted downstream of the EphA4 promoter as a reporter. Using this system, we observed high expression of EphA4 in the heart atria and in the brain. KEY FINDINGS EphaA4 KO rats (EphA4-/-) developed obvious atrial hypertrophy with an increased atria-to-heart weight ratio and atrial cardiomyocyte cross-sectional area at six months of age. EphA4-/- rats had reduced atrial end diastolic volume (EDV), atrial ejection fraction (EF) and left ventricular EF. They also exhibited increased amplitude of QRS complexes and QT intervals, with invisible p waves. RNA sequencing revealed that EphA4 KO altered the transcription of multiple genes involved in regulation of transcription and translation, ion binding, metabolism and cell adhesion. Deletion of EphA4 reduced IGF1 mRNA and protein expression, which is involved in cardiac remodeling. SIGNIFICANCE Our data demonstrated that EphA4 was highly expressed in the atria and its deletion caused atrial dysfunction. Our findings also suggested that the EphA4 KO rat could be a potential model for studies on atrial remodeling.
Collapse
|
8
|
Kaur K, Singh N, Dhawan RK. Potential role of EphrinA2 receptors in postconditioning induced cardioprotection in rats. Eur J Pharmacol 2020; 883:173231. [PMID: 32589885 DOI: 10.1016/j.ejphar.2020.173231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 05/20/2020] [Accepted: 05/29/2020] [Indexed: 10/24/2022]
Abstract
EphA2 receptor has emerged as a novel cardioprotective target against myocardial infarction by preserving cardiac function, limiting infarct size and inflammation and enhancing cell survival via elevating phosphorylated Akt protein levels. However, the role of Eph receptors in postconditioning remains to be elucidated. Thus, the present study was designed to explore the role of EphA2 receptors in cardioprotective mechanism of postconditioning by employing Doxazosin as EphA2 receptor agonist, Lithocholic acid as antagonist and Wortmannin as specific phosphoinositide 3-kinase (PI3K) inhibitor. In Langendorff perfused isolated rat hearts, exposure of ischemia for 30 min succeeded by reperfusion for 2 h produced cardiac damage as determined by increase in size of infarct, LVDP, liberation of LDH and CK in effluent from coronary arteries. The reperfused hearts were homogenized and tissue concentrations of TBARs, reduced GSH and Catalase were determined. A marked rise in infarct size, liberation of LDH and CK in effluent and TBARs in myocardial tissue was observed in ischemic and reperfused hearts. Ischemic postconditioning comprising of 6 alternate episodes of 10 s ischemia and 10 s reperfusion and pharmacological post-conditioning by Doxazosin infusion for 5 min Before reperfusion confers significant protection against myocardial injury as manifested by remarkably decreased infarct size, levels of LDH, CK and tissue TBARs along with increase in GSH and Catalase activity. Pre-treatment of EphA2 antagonist, Lithocholic acid and PI3K inhibitor, Wortmannin attenuated the cardioprotective effect of postconditioning. Our results suggest that EphA2 receptors may be involved in postconditioning mediated cardioprotection probably through PI3K/Akt pathway.
Collapse
Affiliation(s)
- Kamaldeep Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India; Department of Pharmacology, Khalsa College of Pharmacy, Amritsar, India
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India.
| | - Ravi K Dhawan
- Department of Pharmacology, Khalsa College of Pharmacy, Amritsar, India
| |
Collapse
|
9
|
Du E, Li X, He S, Li X, He S. The critical role of the interplays of EphrinB2/EphB4 and VEGF in the induction of angiogenesis. Mol Biol Rep 2020; 47:4681-4690. [PMID: 32488576 DOI: 10.1007/s11033-020-05470-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 04/25/2020] [Indexed: 12/12/2022]
Abstract
The significant role of VEGF (vascular endothelial growth factor) as an angiogenesis inducer is well recognized. Besides VEGF, EphrinB2/EphB4 also plays essential roles in vascular development and postnatal angiogenesis. Compared with classical proangiogenic factors, not only does EphrinB2/EphB4 promote sprouting of new vessels, it is also involved in the vessel maturation. Given their involvement in many physiologic and pathological conditions, EphB4 and EphrinB2 are increasingly recognized as attractive therapeutic targets for angiogenesis-related diseases through modulating their expression and function. Previous works mainly focused on the individual role of VEGF and EphrinB2/EphB4 in angiogenesis, respectively, but the correlation between EphrinB2/EphB4 and VEGF in angiogenesis has not been fully disclosed. Here, we summarize the structure and bidirectional signaling of EphrinB2/EphB4, provide an overview on the relationship between EphrinB2/EphB4 signaling and VEGF pathway in angiogenesis and highlight the associated potential usefulness in anti-angiogenetic therapy.
Collapse
Affiliation(s)
- Enming Du
- Henan Eye Institute, Zhengzhou, 450003, Henan, China.,Henan Eye Hospital, Zhengzhou, 450003, Henan, China.,Henan Key Laboratory of Ophthalmology and Visual Science, Zhengzhou, 450003, Henan, China.,People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China.,People's Hospital of Henan University, Zhengzhou, 450003, Henan, China.,Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Xue Li
- Henan Eye Institute, Zhengzhou, 450003, Henan, China.,Henan Eye Hospital, Zhengzhou, 450003, Henan, China.,Henan Key Laboratory of Ophthalmology and Visual Science, Zhengzhou, 450003, Henan, China.,People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China.,People's Hospital of Henan University, Zhengzhou, 450003, Henan, China.,Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Siyu He
- Henan Eye Institute, Zhengzhou, 450003, Henan, China.,Henan Eye Hospital, Zhengzhou, 450003, Henan, China.,Henan Key Laboratory of Ophthalmology and Visual Science, Zhengzhou, 450003, Henan, China.,People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China.,People's Hospital of Henan University, Zhengzhou, 450003, Henan, China.,Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China
| | - Xiaohua Li
- Henan Eye Institute, Zhengzhou, 450003, Henan, China. .,Henan Eye Hospital, Zhengzhou, 450003, Henan, China. .,Henan Key Laboratory of Ophthalmology and Visual Science, Zhengzhou, 450003, Henan, China. .,People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China. .,People's Hospital of Henan University, Zhengzhou, 450003, Henan, China. .,Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China.
| | - Shikun He
- Henan Eye Institute, Zhengzhou, 450003, Henan, China. .,Henan Eye Hospital, Zhengzhou, 450003, Henan, China. .,Henan Provincial People's Hospital, Zhengzhou, 450003, Henan, China. .,Departments of Pathology and Ophthalmology, Keck School of Medicine of the University of Southern California, USC Roski Eye Institute, Los Angeles, CA, 90033, USA.
| |
Collapse
|
10
|
Su SA, Xie Y, Zhang Y, Xi Y, Cheng J, Xiang M. Essential roles of EphrinB2 in mammalian heart: from development to diseases. Cell Commun Signal 2019; 17:29. [PMID: 30909943 PMCID: PMC6434800 DOI: 10.1186/s12964-019-0337-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/11/2019] [Indexed: 12/11/2022] Open
Abstract
EphrinB2, a membrane-tethered ligand preferentially binding to its receptor EphB4, is ubiquitously expressed in all mammals. Through the particular bidirectional signaling, EphrinB2 plays a critical role during the development of cardiovascular system, postnatal angiogenesis physiologically and pathologically, and cardiac remodeling after injuries as an emerging role. This review highlights the pivotal involvement of EphrinB2 in heart, from developmental cardiogenesis to pathological cardiac remodeling process. Further potential translational therapies will be discussed in targeting EphrinB2 signaling, to better understand the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Sheng-An Su
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yuhao Zhang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yutao Xi
- Texas Heart Institute, Houston, 77030, USA.
| | - Jie Cheng
- Texas Heart Institute, Houston, 77030, USA
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
11
|
Horton JL, Virag J. Use of Multifactorial Treatments to Address the Challenge of Translating Experimental Myocardial Infarct Reduction Strategies. Int J Mol Sci 2019; 20:E1449. [PMID: 30909376 PMCID: PMC6471438 DOI: 10.3390/ijms20061449] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 03/13/2019] [Accepted: 03/15/2019] [Indexed: 12/27/2022] Open
Abstract
Myocardial tissue damage that occurs during an ischemic event leads to a spiraling deterioration of cardiac muscle structural and functional integrity. Reperfusion is the only known efficacious strategy and is the most commonly used treatment to reduce injury and prevent remodeling. However, timing is critical, and the procedure is not always feasible for a variety of reasons. The complex molecular basis for cardioprotection has been studied for decades but formulation of a viable therapeutic that can significantly attenuate myocardial injury remains elusive. In this review, we address barriers to the development of a fruitful approach that will substantially improve the prognosis of those suffering from this widespread and largely unmitigated disease. Furthermore, we proffer that ephrinA1, a candidate molecule that satisfies many of the important criteria discussed, possesses robust potential to overcome these hurdles and thus offers protection that surpasses the limitations currently observed.
Collapse
Affiliation(s)
| | - Jitka Virag
- Department of Physiology, Brody School of Medicine, 600 Moye Blvd, East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|
12
|
Yuan C, Wang P, Zhu S, Liu Z, Wang W, Geng T, Dissanayaka WL, Jin L, Zhang C. Overexpression of ephrinB2 in stem cells from apical papilla accelerates angiogenesis. Oral Dis 2019; 25:848-859. [PMID: 30667136 DOI: 10.1111/odi.13042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/17/2018] [Accepted: 01/16/2019] [Indexed: 12/30/2022]
Abstract
OBJECTIVES We aimed to accelerate angiogenesis in pulp regeneration by modulating ephrinB2 expression in stem cells from apical papilla (SCAPs). MATERIALS AND METHODS Stem cells from apical papilla were transducted with ephrinB2-lentiviral expression vector (ephrinB2-SCAPs) in experimental group and green fluorescent protein (GFP-SCAPs) in control group. The transduction efficiency was confirmed by real-time PCR and Western blot assays. MTT assay was performed to detect the proliferative capacity of SCAPs after transduction. In vitro Matrigel assay and in vivo Matrigel plug assay were carried out to evaluate the angiogenic capacity. RESULTS Results showed that ephrinB2-SCAPs had significantly higher ephrinB2 expression than GFP-SCAPs. EphrinB2-SCAPs upregulated vascular endothelial growth factor (VEGF) secretion under hypoxia. In vitro Matrigel assay demonstrated that human umbilical vein endothelial cells (HUVECs) cocultured with ephrinB2-SCAPs under hypoxia formed vascular-like structures earlier than GFP-SCAPs. Animal experiments confirmed that SCAPs co-transplanted with HUVECs enabled to generate greater amount of blood vessels than SCAPs alone. EphrinB2-SCAPs produced increased number of blood vessels with references to GFP-SCAPs, and those co-transplanted with HUVECs generated vessels with larger and functional tubule volumes. CONCLUSIONS Regulating ephrinB2 expression in SCAPs may act as a new avenue for enhancing angiogenesis in dental pulp regeneration.
Collapse
Affiliation(s)
- Changyong Yuan
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong.,HKU Shenzhen Institute of Research and Innovation, Shenzhen, China.,Dental Implant Center, Affiliated Xuzhou Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Penglai Wang
- Dental Implant Center, Affiliated Xuzhou Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Shaoyue Zhu
- Dental Implant Center, Affiliated Xuzhou Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zongxiang Liu
- Dental Implant Center, Affiliated Xuzhou Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wen Wang
- Dental Implant Center, Affiliated Xuzhou Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Tengyu Geng
- Dental Implant Center, Affiliated Xuzhou Stomatological Hospital of Xuzhou Medical University, Xuzhou, China
| | - Waruna Lakmal Dissanayaka
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong.,HKU Shenzhen Institute of Research and Innovation, Shenzhen, China
| | - Lijian Jin
- Periodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong
| | - Chengfei Zhang
- Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong.,HKU Shenzhen Institute of Research and Innovation, Shenzhen, China
| |
Collapse
|
13
|
Zheng LC, Wang XQ, Lu K, Deng XL, Zhang CW, Luo H, Xu XD, Chen XM, Yan L, Wang YQ, Shi SL. Ephrin-B2/Fc promotes proliferation and migration, and suppresses apoptosis in human umbilical vein endothelial cells. Oncotarget 2018; 8:41348-41363. [PMID: 28489586 PMCID: PMC5522204 DOI: 10.18632/oncotarget.17298] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 04/03/2017] [Indexed: 01/12/2023] Open
Abstract
Tumor growth and metastasis are angiogenesis dependent. Angiogenic growth involves endothelial cell proliferation, migration, and invasion. Ephrin-B2 is a ligand for Eph receptor tyrosine kinases and is an important mediator in vascular endothelial growth factor-mediated angiogenesis. However, research offer controversial information regarding effects of ephrin-B2 on vascular endothelial cells. In this paper, proteome analyses showed that ephrin-B2/Fc significantly activates multiple signaling pathways related to cell proliferation, survival, and migration and suppresses apoptosis and cell death. Cytological experiments further confirm that ephrin-B2/Fc stimulates endothelial cell proliferation, triggers dose-dependent migration, and suppresses cell apoptosis. Results demonstrate that soluble dose-dependent ephrinB2 can promote proliferation and migration and inhibit apoptosis of human umbilical vein endothelial cells. These results also suggest that ephrinB2 prevents ischemic disease and can potentially be a new therapeutic target for treating angiogenesis-related diseases and tumors.
Collapse
Affiliation(s)
- Li-Chun Zheng
- Medical College of Xiamen University, Jinshan Community Health Service Center, Xiamen Traditional Chinese Medical Hospital, Xiamen 361000, P.R. China.,Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Xiao-Qing Wang
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Kun Lu
- Department of Basic Medicine, Medical College of Xiamen University, Cancer Research Center of Xiamen University, Xiamen 361102, P.R. China
| | - Xiao-Ling Deng
- Department of Basic Medicine, Medical College of Xiamen University, Cancer Research Center of Xiamen University, Xiamen 361102, P.R. China
| | - Cheng-Wei Zhang
- Department of Cardiology, Affiliated Dongnan Hospital of Xiamen University, Zhangzhou 363000, P.R. China
| | - Hong Luo
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Xu-Dong Xu
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Xiao-Man Chen
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Lu Yan
- Department of Basic Medicine, Medical College of Xiamen University, Xiamen 361102, P.R. China
| | - Yi-Qing Wang
- Xiamen Heart Center, Medical College of Xiamen University, Xiamen 361000, P.R. China
| | - Song-Lin Shi
- Department of Basic Medicine, Medical College of Xiamen University, Cancer Research Center of Xiamen University, Xiamen 361102, P.R. China
| |
Collapse
|
14
|
Yuan C, Wang P, Zhu S, Zou T, Wang S, Xu J, Heng BC, Diogenes A, Zhang C. EphrinB2 Stabilizes Vascularlike Structures Generated by Endothelial Cells and Stem Cells from Apical Papilla. J Endod 2016; 42:1362-70. [PMID: 27451120 DOI: 10.1016/j.joen.2016.05.012] [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: 12/15/2015] [Revised: 04/26/2016] [Accepted: 05/14/2016] [Indexed: 12/26/2022]
Abstract
INTRODUCTION This study aimed to investigate the roles of ephrinB2 in stabilizing vascularlike structures generated by stem cells from apical papilla (SCAPs) and human umbilical vein endothelial cells (HUVECs). METHODS HUVECs were seeded alone or with SCAPs concurrently or 12 hours later. Angiogenesis and ephrinB2 phosphorylation were assayed at different time points. Additionally, ephrinB2 expression in SCAPs and HUVECs was silenced with small interfering RNA, and vascularlike structure formation within coculture was assessed; 1 × 10(5) HUVECs were seeded in transwell inserts, and 6 × 10(5) SCAPs were plated in lower wells with or without ephrinB2-Fc. Migratory cells were stained and counted. Delayed addition of ephrinB2-Fc to the coculture of HUVECs and SCAPs was performed to evaluate the role of ephrinB2 on the stabilization of vascularlike structures. RESULTS Concurrent coculture of SCAPs and HUVECs yielded significantly longer tubule lengths at 4, 8, and 12 hours (P < .05). Delayed addition of SCAPs to coculture with HUVECs resulted in vascularlike structures persisting longer than the HUVEC monoculture. Western blot confirmed that ephrinB2 phosphorylation was initiated at 0.5 hours of coculture and peaked at 1 hour. Silencing ephrinB2 expression in SCAPs and HUVECs resulted in the absence of vascularlike structures. Enhanced migration of HUVECs by SCAPs could be inhibited by ephrinB2-Fc. When ephrinB2-Fc was added at 3 hours of coculture, the vascularlike structures were stabilized for more than 12 hours as compared with 9 hours in the control group. CONCLUSIONS EphrinB2 plays an important role in the stabilization of vascularlike structures generated by HUVECs and SCAPs.
Collapse
Affiliation(s)
- Changyong Yuan
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Penglai Wang
- Dental Implant Center, Xuzhou Stomatological Hospital, Xuzhou, China
| | - Shaoyue Zhu
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China; Dental Implant Center, Xuzhou Stomatological Hospital, Xuzhou, China
| | - Ting Zou
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Shuai Wang
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Jianguang Xu
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Boon Chin Heng
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Anibal Diogenes
- Department of Endodontics, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | - Chengfei Zhang
- Comprehensive Dental Care, Endodontics, Faculty of Dentistry, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
15
|
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
|
16
|
Xiao H, Huang Q, Wang JQ, Deng QQ, Gu WP. Effect of ephrin-B2 on the expressions of angiopoietin-1 and -2 after focal cerebral ischemia/reperfusion. Neural Regen Res 2016; 11:1784-1789. [PMID: 28123421 PMCID: PMC5204233 DOI: 10.4103/1673-5374.194723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ephrin-B2 has been shown to participate in angiogenesis, but the underlying mechanisms involved remain unclear. In this study, a rat model of focal cerebral ischemia was prepared by focal middle cerebral artery occlusion, followed by 24-hour reperfusion. Then, ephrin-B2 protein was administered intracerebroventricularly for 3 consecutive days via a micro-osmotic pump. Western blot assay and quantitative real-time reverse transcription PCR demonstrated the expression levels of angiopoietin-1 (Ang-1) mRNA and protein in the penumbra cortex of the ephrin-B2 treated group were decreased at day 4 after reperfusion, and increased at day 28, while the expression levels of angiopoietin-2 (Ang-2) were highly up-regulated at all time points tested. Double immunofluorescent staining indicated that Ang-1 and Ang-2 were both expressed in vascular endothelial cells positive for CD31. These findings indicate that ephrin-B2 influences the expressions of Ang-1 and Ang-2 during angiogenesis following transient focal cerebral ischemia.
Collapse
Affiliation(s)
- Hui Xiao
- Department of Neurology, Changsha Municipal Central Hospital, Changsha, Hunan Province, China
| | - Qing Huang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Jia-Qi Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Qing-Qing Deng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Wen-Ping Gu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| |
Collapse
|
17
|
Cardiac Sympathetic Nerve Sprouting and Susceptibility to Ventricular Arrhythmias after Myocardial Infarction. Cardiol Res Pract 2015; 2015:698368. [PMID: 26793403 PMCID: PMC4697091 DOI: 10.1155/2015/698368] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 12/02/2015] [Indexed: 12/04/2022] Open
Abstract
Ventricular arrhythmogenesis is thought to be a common cause of sudden cardiac death following myocardial infarction (MI). Nerve remodeling as a result of MI is known to be an important genesis of life-threatening arrhythmias. It is hypothesized that neural modulation might serve as a therapeutic option of malignant arrhythmias. In fact, left stellectomy or β-blocker therapy is shown to be effective in the prevention of ventricular tachyarrhythmias (VT), ventricular fibrillation (VF), and sudden cardiac death (SCD) after MI both in patients and in animal models. Results from decades of research already evidenced a positive relationship between abnormal nerve density and ventricular arrhythmias after MI. In this review, we summarized the molecular mechanisms involved in cardiac sympathetic rejuvenation and mechanisms related to sympathetic hyperinnervation and arrhythmogenesis after MI and analyzed the potential therapeutic implications of nerve sprouting modification for ventricular arrhythmias and SCD control.
Collapse
|
18
|
Dalton JE, Glover AC, Hoodless L, Lim EK, Beattie L, Kirby A, Kaye PM. The neurotrophic receptor Ntrk2 directs lymphoid tissue neovascularization during Leishmania donovani infection. PLoS Pathog 2015; 11:e1004681. [PMID: 25710496 PMCID: PMC4339582 DOI: 10.1371/journal.ppat.1004681] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 01/12/2015] [Indexed: 01/31/2023] Open
Abstract
The neurotrophic tyrosine kinase receptor type 2 (Ntrk2, also known as TrkB) and its ligands brain derived neurotrophic factor (Bdnf), neurotrophin-4 (NT-4/5), and neurotrophin-3 (NT-3) are known primarily for their multiple effects on neuronal differentiation and survival. Here, we provide evidence that Ntrk2 plays a role in the pathologic remodeling of the spleen that accompanies chronic infection. We show that in Leishmania donovani-infected mice, Ntrk2 is aberrantly expressed on splenic endothelial cells and that new maturing blood vessels within the white pulp are intimately associated with F4/80(hi)CD11b(lo)CD11c(+) macrophages that express Bdnf and NT-4/5 and have pro-angiogenic potential in vitro. Furthermore, administration of the small molecule Ntrk2 antagonist ANA-12 to infected mice significantly inhibited white pulp neovascularization but had no effect on red pulp vascular remodeling. We believe this to be the first evidence of the Ntrk2/neurotrophin pathway driving pathogen-induced vascular remodeling in lymphoid tissue. These studies highlight the therapeutic potential of modulating this pathway to inhibit pathological angiogenesis.
Collapse
Affiliation(s)
- Jane E. Dalton
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| | - Amy C. Glover
- Jack Birch Unit, Department of Biology, University of York, York, United Kingdom
| | - Laura Hoodless
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| | - Eng-Kiat Lim
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| | - Lynette Beattie
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| | - Alun Kirby
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
| | - Paul M. Kaye
- Centre for Immunology and Infection, Department of Biology and Hull York Medical School, University of York, York, United Kingdom
- * E-mail:
| |
Collapse
|
19
|
Abstract
The erythropoietin-producing hepatocellular carcinoma (Eph) receptor tyrosine kinase family plays important roles in developmental processes, adult tissue homeostasis, and various diseases. Interaction with Eph receptor-interacting protein (ephrin) ligands on the surface of neighboring cells triggers Eph receptor kinase-dependent signaling. The ephrins can also transmit signals, leading to bidirectional cell contact-dependent communication. Moreover, Eph receptors and ephrins can function independently of each other through interplay with other signaling systems. Given their involvement in many pathological conditions ranging from neurological disorders to cancer and viral infections, Eph receptors and ephrins are increasingly recognized as attractive therapeutic targets, and various strategies are being explored to modulate their expression and function. Eph receptor/ephrin upregulation in cancer cells, the angiogenic vasculature, and injured or diseased tissues also offer opportunities for Eph/ephrin-based targeted drug delivery and imaging. Thus, despite the challenges presented by the complex biology of the Eph receptor/ephrin system, exciting possibilities exist for therapies exploiting these molecules.
Collapse
Affiliation(s)
- Antonio Barquilla
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037; ,
| | | |
Collapse
|
20
|
Increased EphB2 expression predicts cholangiocarcinoma metastasis. Tumour Biol 2014; 35:10031-41. [DOI: 10.1007/s13277-014-2295-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 06/30/2014] [Indexed: 11/26/2022] Open
|
21
|
|
22
|
Lorentz CU, Parrish DC, Alston EN, Pellegrino MJ, Woodward WR, Hempstead BL, Habecker BA. Sympathetic denervation of peri-infarct myocardium requires the p75 neurotrophin receptor. Exp Neurol 2013; 249:111-9. [PMID: 24013014 PMCID: PMC3826885 DOI: 10.1016/j.expneurol.2013.08.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 08/23/2013] [Accepted: 08/27/2013] [Indexed: 12/22/2022]
Abstract
Development of cardiac sympathetic heterogeneity after myocardial infarction contributes to ventricular arrhythmias and sudden cardiac death. Regions of sympathetic hyperinnervation and denervation appear in the viable myocardium beyond the infarcted area. While elevated nerve growth factor (NGF) is implicated in sympathetic hyperinnervation, the mechanisms underlying denervation are unknown. Recent studies show that selective activation of the p75 neurotrophin receptor (p75(NTR)) in sympathetic neurons causes axon degeneration. We used mice that lack p75(NTR) to test the hypothesis that activation of p75(NTR) causes peri-infarct sympathetic denervation after cardiac ischemia-reperfusion. Wild type hearts exhibited sympathetic denervation adjacent to the infarct 24h and 3 days after ischemia-reperfusion, but no peri-infarct sympathetic denervation occurred in p75(NTR)-/- mice. Sympathetic hyperinnervation was found in the distal peri-infarct myocardium in both genotypes 3 days after MI, and hyperinnervation was increased in the p75(NTR)-/- mice. By 7 days after ischemia-reperfusion, cardiac sympathetic innervation density returned back to sham-operated levels in both genotypes, indicating that axonal pruning did not require p75(NTR). Prior studies revealed that proNGF is elevated in the damaged left ventricle after ischemia-reperfusion, as is mRNA encoding brain-derived neurotrophic factor (BDNF). ProNGF and BDNF preferentially bind p75(NTR) rather than TrkA on sympathetic neurons. Immunohistochemistry using Bdnf-HA mice confirmed the presence of BDNF or proBDNF in the infarct after ischemia-reperfusion. Thus, at least two p75(NTR) ligands are elevated in the left ventricle after ischemia-reperfusion where they may stimulate p75(NTR)-dependent denervation of peri-infarct myocardium. In contrast, NGF-induced sympathetic hyperinnervation in the distal peri-infarct ventricle is attenuated by p75(NTR).
Collapse
Affiliation(s)
- Christina U. Lorentz
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, Oregon 97239, USA
| | - Diana C. Parrish
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, Oregon 97239, USA
| | - Eric N. Alston
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, Oregon 97239, USA
| | - Michael J. Pellegrino
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, Oregon 97239, USA
| | - William R. Woodward
- Department of Neurology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, Oregon 97239, USA
| | - Barbara L. Hempstead
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medical College, 1300 York Avenue, New York, NY 10065, USA
| | - Beth A. Habecker
- Department of Physiology and Pharmacology, Oregon Health and Science University, 3181 SW Sam Jackson Park Rd., Portland, Oregon 97239, USA
| |
Collapse
|
23
|
O'Neal WT, Griffin WF, Dries-Devlin JL, Kent SD, Chen J, Willis MS, Virag JAI. Ephrin-Eph signaling as a potential therapeutic target for the treatment of myocardial infarction. Med Hypotheses 2013; 80:738-44. [PMID: 23562676 DOI: 10.1016/j.mehy.2013.02.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 01/27/2013] [Accepted: 02/28/2013] [Indexed: 01/22/2023]
Abstract
Although numerous strategies have been developed to reduce the initial ischemic insult and cellular injury that occurs during myocardial infarction (MI), few have progressed into the clinical arena. The epidemiologic and economic impact of MI necessitates the development of innovative therapies to rapidly and effectively reduce the initial injury and subsequent cardiac dysfunction. The Eph receptors and their cognate ligands, the ephrins, are the largest family of receptor tyrosine kinases, and their signaling has been shown to play a diverse role in various cellular processes. The recent advances in the study of ephrin-Eph signaling have shown promising progress in many fields of medicine. They have been implicated in the pathophysiology of various cancers and in the regulation of inflammation and apoptosis. Recent studies have shown that manipulation of ephrin-Eph cell signaling can favorably influence cardiomyocyte viability and ultimately preserve cardiac function post-MI. In this article, we explore the hypothesis that manipulation of ephrin-Eph signaling may potentially be a novel therapeutic target in the treatment of MI through alteration of the cellular processes that govern injury and wound healing.
Collapse
Affiliation(s)
- Wesley T O'Neal
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Roura S, Bagó JR, Soler-Botija C, Pujal JM, Gálvez-Montón C, Prat-Vidal C, Llucià-Valldeperas A, Blanco J, Bayes-Genis A. Human umbilical cord blood-derived mesenchymal stem cells promote vascular growth in vivo. PLoS One 2012; 7:e49447. [PMID: 23166670 PMCID: PMC3500294 DOI: 10.1371/journal.pone.0049447] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 10/09/2012] [Indexed: 01/01/2023] Open
Abstract
Stem cell therapies are promising strategies to regenerate human injured tissues, including ischemic myocardium. Here, we examined the acquisition of properties associated with vascular growth by human umbilical cord blood-derived mesenchymal stem cells (UCBMSCs), and whether they promoted vascular growth in vivo. UCBMSCs were induced in endothelial cell-specific growth medium (EGM-2) acquiring new cell markers, increased Ac-LDL uptake, and migratory capacity as assessed by qRT-PCR, Western blotting, indirect immunofluorescence, and invasion assays. Angiogenic and vasculogenic potentials could be anticipated by in vitro experiments showing self organization into Matrigel-mediated cell networks, and activation of circulating angiogenic-supportive myeloid cells. In mice, following subcutaneous co-injection with Matrigel, UCBMSCs modified to co-express bioluminescent (luciferases) and fluorescent proteins were demonstrated to participate in the formation of new microvasculature connected with the host circulatory system. Response of UCBMSCs to ischemia was explored in a mouse model of acute myocardial infarction (MI). UCBMSCs transplanted using a fibrin patch survived 4 weeks post-implantation and organized into CD31+network structures above the infarcted myocardium. MI-treated animals showed a reduced infarct scar and a larger vessel-occupied area in comparison with MI-control animals. Taken together, the presented results show that UCBMSCs can be induced in vitro to acquire angiogenic and vasculogenic properties and contribute to vascular growth in vivo.
Collapse
Affiliation(s)
- Santiago Roura
- ICREC Research Program, Fundació Institut dInvestigació en Ciències de la Salut Germans Trias i Pujol (IGTP), Badalona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Zozulya SA, Udovichenko IP. [Eph family receptors as therapeutic targets]. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2012; 38:267-79. [PMID: 22997698 DOI: 10.1134/s106816201203017x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Anti-angiogenic therapy is currently a commonly accepted and rapidly developing approach in oncology and other pathologies linked to aberrant neovascularization. Discovery and validation of additional molecular targets in angiogenesis is needed due to the limitations of the existing clinical therapeutics inhibiting activity of vascular endothelial growth factor (VEGF) and its receptors. A brief review of normal and pathological biological functions of the Eph family of receptor tyrosine kinases and their ephrin ligands is presented, and the approaches to developing therapeutics with anti- and pro-angiogenic and anti-tumor activity based on selective molecular modulation of Eph-ephrin signaling pairs are discussed. Functional roles of Eph-kinases and ephrins in such mechanisms of cancerogenesis as cell proliferation and invasion are also addressed.
Collapse
|
26
|
Nievergall E, Lackmann M, Janes PW. Eph-dependent cell-cell adhesion and segregation in development and cancer. Cell Mol Life Sci 2012; 69:1813-42. [PMID: 22204021 PMCID: PMC11114713 DOI: 10.1007/s00018-011-0900-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2011] [Revised: 11/06/2011] [Accepted: 11/28/2011] [Indexed: 01/23/2023]
Abstract
Numerous studies attest to essential roles for Eph receptors and their ephrin ligands in controlling cell positioning and tissue patterning during normal and oncogenic development. These studies suggest multiple, sometimes contradictory, functions of Eph-ephrin signalling, which under different conditions can promote either spreading and cell-cell adhesion or cytoskeletal collapse, cell rounding, de-adhesion and cell-cell segregation. A principle determinant of the balance between these two opposing responses is the degree of receptor/ligand clustering and activation. This equilibrium is likely altered in cancers and modulated by somatic mutations of key Eph family members that have emerged as candidate cancer markers in recent profiling studies. In addition, cross-talk amongst Ephs and with other signalling pathways significantly modulates cell-cell adhesion, both between and within Eph- and ephrin-expressing cell populations. This review summarises our current understanding of how Eph receptors control cell adhesion and morphology, and presents examples demonstrating the importance of these events in normal development and cancer.
Collapse
Affiliation(s)
- Eva Nievergall
- Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, VIC 3800 Australia
- Present Address: Haematology Department, SA Pathology, Frome Road, Adelaide, SA 5000 Australia
| | - Martin Lackmann
- Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, VIC 3800 Australia
| | - Peter W. Janes
- Department of Biochemistry and Molecular Biology, Monash University, Wellington Road, Clayton, VIC 3800 Australia
| |
Collapse
|
27
|
Mierke CT. Endothelial cell's biomechanical properties are regulated by invasive cancer cells. MOLECULAR BIOSYSTEMS 2012; 8:1639-49. [PMID: 22498801 DOI: 10.1039/c2mb25024a] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Most cancer-related deaths are caused by the ability of cancer cells to metastasize. This process includes the dissemination of cancer cells from the primary tumor side and their migration to targeted organ sites. During the migration of cancer cells through the connective tissue microenvironment, which consists of endothelial cells and extracellular matrix components, biomechanical properties are crucial for the efficiency and speed of cancer cell invasion and subsequently, metastases formation. Biomechanics can enable cancer cells to migrate through tissue, transmigrate through basement membranes as well as endothelial monolayers and form metastases in targeted organs. The current focus of cancer research still lies on the investigation of cancer cell's biochemical and molecular capabilities such as molecular genetics and gene signaling, but these approaches ignore the mechanical nature of the invasion process of cancer cells. Moreover, even the role of the endothelium during the transmigration and invasion of cells is not clear, it has been seen as a passive barrier, but this could not explain all novel findings. This review discusses how cancer cells alter the structural, biochemical and mechanical properties of the endothelium to regulate their own invasiveness through extracellular matrices and hence, through the tissue microenvironment. Finally, this review sheds light on the mechanical properties of cancer cells and the interacting endothelium and points out the importance of the mechanical properties as a critical determinant for the efficiency of cancer cell invasion and the overall progression of cancer. In conclusion, the regulation of the endothelial cell's biomechanical properties by cancer cells is a critical determinant of cancer cell invasiveness and may affect the future development of new cancer treatments.
Collapse
Affiliation(s)
- Claudia T Mierke
- Faculty of Physics and Earth Science, Institute of Experimental Physics I, Soft Matter Physics Division, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
28
|
Vadivel A, van Haaften T, Alphonse RS, Rey-Parra GJ, Ionescu L, Haromy A, Eaton F, Michelakis E, Thébaud B. Critical role of the axonal guidance cue EphrinB2 in lung growth, angiogenesis, and repair. Am J Respir Crit Care Med 2011; 185:564-74. [PMID: 22161159 DOI: 10.1164/rccm.201103-0545oc] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Lung diseases characterized by alveolar damage currently lack efficient treatments. The mechanisms contributing to normal and impaired alveolar growth and repair are incompletely understood. Axonal guidance cues (AGC) are molecules that guide the outgrowth of axons to their targets. Among these AGCs, members of the Ephrin family also promote angiogenesis, cell migration, and organogenesis outside the nervous system. The role of Ephrins during alveolar growth and repair is unknown. OBJECTIVES We hypothesized that EphrinB2 promotes alveolar development and repair. METHODS We used in vitro and in vivo manipulation of EphrinB2 signaling to assess the role of this AGC during normal and impaired lung development. MEASUREMENTS AND MAIN RESULTS In vivo EphrinB2 knockdown using intranasal siRNA during the postnatal stage of alveolar development in rats arrested alveolar and vascular growth. In a model of O(2)-induced arrested alveolar growth in newborn rats, air space enlargement, loss of lung capillaries, and pulmonary hypertension were associated with decreased lung EphrinB2 and receptor EphB4 expression. In vitro, EphrinB2 preserved alveolar epithelial cell viability in O(2), decreased O(2)-induced alveolar epithelial cell apoptosis, and accelerated alveolar epithelial cell wound healing, maintained lung microvascular endothelial cell viability, and proliferation and vascular network formation. In vivo, treatment with intranasal EphrinB2 decreased alveolar epithelial and endothelial cell apoptosis, preserved alveolar and vascular growth in hyperoxic rats, and attenuated pulmonary hypertension. CONCLUSION The AGC EphrinB2 may be a new therapeutic target for lung repair and pulmonary hypertension.
Collapse
Affiliation(s)
- Arul Vadivel
- Department of Pediatrics, School of Human Development, Women and Children’s Health Research Institute, Edmonton, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Pitavastatin-induced angiogenesis and arteriogenesis is mediated by Notch1 in a murine hindlimb ischemia model without induction of VEGF. J Transl Med 2011; 91:691-703. [PMID: 21301413 PMCID: PMC3807100 DOI: 10.1038/labinvest.2011.5] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Notch signaling is reported to regulate angiogenesis, interacting with vascular endothelial growth factor (VEGF) signaling. HMG CoA reductase inhibitors (statins) also alter Notch signaling in vascular cells, but the mechanism and involvement of Notch and VEGF signaling in statin-mediated angiogenesis remain unclear. Here, we examined how statins activate the endothelial Notch1, and promote angiogenesis and arteriogenesis. We examined blood flow recovery after hindlimb ischemia in wild-type (WT) and Notch1 mutant mice treated with or without pitavastatin (3 mg/kg/day, p.o.). Although VEGF induction was not altered in ischemic limbs, pitavastatin promoted blood flow recovery in ischemic limbs in control mice but not in Notch1 mutant mice. Furthermore, pitavastatin induced endothelial ephrinB2 downstream of Notch1 and increased the density of both capillaries and arterioles in the ischemic limbs of WT but not of Notch1 mutant mice. Pitavastatin (100 nmol/l) rapidly activated γ-secretase and Notch1 in human umbilical vein endothelial cells without VEGF induction, which was suppressed by pharmacological inhibition and knockdown of Akt. Pitavastatin also augmented endothelial proliferation and tube formation on Matrigel, which were suppressed by either γ-secretase inhibition or knockdown of Notch1. Pitavastatin-induced microvascular sprouting was also impaired in Notch1 mutant aortic explants. Taken together, pitavastatin activates Notch1 through Akt-dependent stimulation of γ-secretase in endothelial cells, and thereby increases vasculogenesis without VEGF induction.
Collapse
|
30
|
Dries JL, Kent SD, Virag JAI. Intramyocardial administration of chimeric ephrinA1-Fc promotes tissue salvage following myocardial infarction in mice. J Physiol 2011; 589:1725-40. [PMID: 21282286 DOI: 10.1113/jphysiol.2010.202366] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The purpose of this study was to investigate the role of intramyocardial administration of chimeric ephrinA1-Fc in modulating the extent of injury and inflammation in non reperfused myocardial infarction (MI). Our results show that intramyocardial injection of 6 μg ephrinA1-Fc into the border zone immediately after permanent coronary artery ligation in B6129s mice resulted in 50% reduction of infarct size, 64% less necrosis, 35% less chamber dilatation and 32% less left ventricular free wall thinning at 4 days post-MI. In the infarct zone, Ly6G+ neutrophil density was 57% reduced and CD45+ leukocyte density was 21% reduced. Myocyte damage was also reduced in ephrinA1-Fc-treated hearts, as evidenced by 54% reduced serum cardiac troponin I. Further, we observed decreased cleaved PARP, increased BAG-1 protein expression, increased phosphorylated AKT/total AKT protein, and reduced NF-κB protein with ephrinA1-Fc administration, indicating improved cellular survival. Of the eight EphA receptors known to be expressed in mice (A1–A8), RT-PCR revealed that A1–A4, A6 and A7 were expressed in the uninjured adult myocardium. Expression of EphA1–A3 and EphA7 were significantly increased following MI while EphA6 expression decreased. Treatment with ephrinA1-Fc further increased EphA1 and EphA2 gene expression and resulted in a 2-fold increase in EphA4. Upregulation and combinatorial activation of these receptors may promote tissue survival. We have identified a novel, beneficial role for ephrinA1-Fc administration at the time of MI, and propose this as a promising new target for infarct salvage in non reperfused MI. More experiments are in progress to identify receptor-expressing cell types as well as the functional implications of receptor activation.
Collapse
Affiliation(s)
- Jessica L Dries
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Blvd, Greenville, NC 27834, USA
| | | | | |
Collapse
|
31
|
Chan J, Mably JD. Dissection of cardiovascular development and disease pathways in zebrafish. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 100:111-53. [PMID: 21377626 DOI: 10.1016/b978-0-12-384878-9.00004-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
The use of animal models in medicine has contributed significantly to the development of drug treatments and surgical procedures for the last century, in particular for cardiovascular disease. In order to model human disease in an animal, an appreciation of the strengths and limitations of the system are required to interpret results and design the logical sequence of steps toward clinical translation. As the world's population ages, cardiovascular disease will become even more prominent and further progress will be essential to stave off what seems destined to become a massive public health issue. Future treatments will require the imaginative application of current models as well as the generation of new ones. In this review, we discuss the resources available for modeling cardiovascular disease in zebrafish and the varied attributes of this system. We then discuss current zebrafish disease models and their potential that has yet to be exploited.
Collapse
Affiliation(s)
- Joanne Chan
- Vascular Biology Program, Department of Surgery, Children's Hospital Boston, and Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
32
|
Duffy GP, D'Arcy S, Ahsan T, Nerem RM, O'Brien T, Barry F. Mesenchymal stem cells overexpressing ephrin-b2 rapidly adopt an early endothelial phenotype with simultaneous reduction of osteogenic potential. Tissue Eng Part A 2010; 16:2755-68. [PMID: 20491587 DOI: 10.1089/ten.tea.2009.0623] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Restoration of the vascular supply to ischemic tissues is of high clinical relevance, and proangiogenic therapies aim to reduce morbidity and mortality rates associated with the onset of cardiovascular disease. Stem cell therapy has been proposed as a potentially useful proangiogenic therapy. Mesenchymal stem cells (MSCs) have been shown to be proangiogenic and produce a number of cytokines involved in vessel development and maturation. Preclinical studies have reported increased angiogenesis after MSC delivery to the heart, and similar outcomes have been reported in recent clinical trials. Stem-cell-mediated neovascularization has been augmented by genetic modification with overexpression of angiogenic cytokines, including vascular endothelial growth factor (VEGF) and platelet-derived growth factor, showing promising results. In this study we aimed to enhance the proangiogenic capability of MSCs. MSCs were genetically modified to overexpress a versatile molecule, Ephrin-B2, involved in tissue morphogenesis and vascular development to enhance inherent neovascularization potential. Using nucleofection, Ephrin-B2 was transiently overexpressed on the cell surface of MSCs to recapitulate embryonic signaling and promote neovascularization. Ephrin-B2-expressing MSCs adopted an early endothelial phenotype under endothelial cell culture conditions increasing expression of von Willebrand factor and VEGF-Receptor 2. The cells had an increased ability to form vessel-like structures, produce VEGF, and incorporate into newly formed endothelial cell structures. These data indicate that MSCs expressing Ephrin-B2 represent a novel proangiogenic cell source to promote neovascularization in ischemic tissues.
Collapse
Affiliation(s)
- Garry P Duffy
- Regenerative Medicine Institute (REMEDI), National University of Ireland Galway, Ireland
| | | | | | | | | | | |
Collapse
|
33
|
Rohrbeck A, Borlak J. Cancer genomics identifies regulatory gene networks associated with the transition from dysplasia to advanced lung adenocarcinomas induced by c-Raf-1. PLoS One 2009; 4:e7315. [PMID: 19812696 PMCID: PMC2754338 DOI: 10.1371/journal.pone.0007315] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 09/13/2009] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Lung cancer is a leading cause of cancer morbidity. To improve an understanding of molecular causes of disease a transgenic mouse model was investigated where targeted expression of the serine threonine kinase c-Raf to respiratory epithelium induced initially dysplasia and subsequently adenocarcinomas. This enables dissection of genetic events associated with precancerous and cancerous lesions. METHODOLOGY/PRINCIPAL FINDINGS By laser microdissection cancer cell populations were harvested and subjected to whole genome expression analyses. Overall 473 and 541 genes were significantly regulated, when cancer versus transgenic and non-transgenic cells were compared, giving rise to three distinct and one common regulatory gene network. At advanced stages of tumor growth predominately repression of gene expression was observed, but genes previously shown to be up-regulated in dysplasia were also up-regulated in solid tumors. Regulation of developmental programs as well as epithelial mesenchymal and mesenchymal endothelial transition was a hall mark of adenocarcinomas. Additionally, genes coding for cell adhesion, i.e. the integrins and the tight and gap junction proteins were repressed, whereas ligands for receptor tyrosine kinase such as epi- and amphiregulin were up-regulated. Notably, Vegfr- 2 and its ligand Vegfd, as well as Notch and Wnt signalling cascades were regulated as were glycosylases that influence cellular recognition. Other regulated signalling molecules included guanine exchange factors that play a role in an activation of the MAP kinases while several tumor suppressors i.e. Mcc, Hey1, Fat3, Armcx1 and Reck were significantly repressed. Finally, probable molecular switches forcing dysplastic cells into malignantly transformed cells could be identified. CONCLUSIONS/SIGNIFICANCE This study provides insight into molecular pertubations allowing dysplasia to progress further to adenocarcinoma induced by exaggerted c-Raf kinase activity.
Collapse
Affiliation(s)
- Astrid Rohrbeck
- Department of Molecular Medicine and Medical Biotechnology, Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
| | - Jürgen Borlak
- Department of Molecular Medicine and Medical Biotechnology, Fraunhofer Institute of Toxicology and Experimental Medicine, Hannover, Germany
- Center for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
34
|
Nicosia RF. The aortic ring model of angiogenesis: a quarter century of search and discovery. J Cell Mol Med 2009; 13:4113-36. [PMID: 19725916 PMCID: PMC4496118 DOI: 10.1111/j.1582-4934.2009.00891.x] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2009] [Accepted: 08/11/2009] [Indexed: 12/14/2022] Open
Abstract
The aortic ring model has become one of the most widely used methods to study angiogenesis and its mechanisms. Many factors have contributed to its popularity including reproducibility, cost effectiveness, ease of use and good correlation with in vivo studies. In this system aortic rings embedded in biomatrix gels and cultured under chemically defined conditions generate arborizing vascular outgrowths which can be stimulated or inhibited with angiogenic regulators. Originally based on the rat aorta, the aortic ring model was later adapted to the mouse for the evaluation of specific molecular alterations in genetically modified animals. Viral transduction of the aortic rings has enabled investigators to overexpress genes of interest in the aortic cultures. Experiments on angiogenic mechanisms have demonstrated that formation of neovessels in aortic cultures is regulated by macrophages, pericytes and fibroblasts through a complex molecular cascade involving growth factors, inflammatory cytokines, axonal guidance cues, extracellular matrix (ECM) molecules and matrix-degrading proteolytic enzymes. These studies have shown that endothelial sprouting can be effectively blocked by depleting the aortic explants of macrophages or by interfering with the angiogenic cascade at multiple levels including growth factor signalling, cell adhesion and proteolytic degradation of the ECM. In this paper, we review the literature in this field and retrace the journey from our first morphological descriptions of the aortic outgrowths to the latest breakthroughs in the cellular and molecular regulation of aortic vessel growth and regression.
Collapse
Affiliation(s)
- R F Nicosia
- Pathology and Laboratory Medicine Services, Veterans Administration Puget Sound Health Care System, Seattle, WA 98108, USA.
| |
Collapse
|
35
|
The ephrinB2/EphB4 axis is dysregulated in osteoprogenitors from myeloma patients and its activation affects myeloma bone disease and tumor growth. Blood 2009; 114:1803-12. [PMID: 19597185 DOI: 10.1182/blood-2009-01-201954] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Myeloma bone disease is caused by uncoupling of osteoclastic bone resorption and osteoblastic bone formation. Bidirectional signaling between the cell-surface ligand ephrinB2 and its receptor, EphB4, is involved in the coupling of osteoblastogenesis and osteoclastogenesis and in angiogenesis. EphrinB2 and EphB4 expression in mesenchymal stem cells (MSCs) from myeloma patients and in bone cells in myelomatous bones was lower than in healthy counterparts. Wnt3a induced up-regulation of EphB4 in patient MSCs. Myeloma cells reduced expression of these genes in MSCs, whereas in vivo myeloma cell-conditioned media reduced EphB4 expression in bone. In osteoclast precursors, EphB4-Fc induced ephrinB2 phosphorylation with subsequent inhibition of NFATc1 and differentiation. In MSCs, EphB4-Fc did not induce ephrinB2 phosphorylation, whereas ephrinB2-Fc induced EphB4 phosphorylation and osteogenic differentiation. EphB4-Fc treatment of myelomatous SCID-hu mice inhibited myeloma growth, osteoclastosis, and angiogenesis and stimulated osteoblastogenesis and bone formation, whereas ephrinB2-Fc stimulated angiogenesis, osteoblastogenesis, and bone formation but had no effect on osteoclastogenesis and myeloma growth. These chimeric proteins had similar effects on normal bone. Myeloma cells expressed low to undetectable ephrinB2 and EphB4 and did not respond to the chimeric proteins. The ephrinB2/EphB4 axis is dysregulated in MM, and its activation by EphB4-Fc inhibits myeloma growth and bone disease.
Collapse
|
36
|
Cristofaro B, Emanueli C. Possible novel targets for therapeutic angiogenesis. Curr Opin Pharmacol 2008; 9:102-8. [PMID: 19071062 PMCID: PMC2698077 DOI: 10.1016/j.coph.2008.11.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2008] [Revised: 11/04/2008] [Accepted: 11/05/2008] [Indexed: 01/13/2023]
Abstract
An increasing number of studies about the molecular basis of angiogenesis are rapidly disclosing novel signal pathways involved in the blood vessel formation process. This review will focus on bone morphogenic proteins, Hedgehog, Notch, ephrins, neuropilins, neurotrophins and netrins. These recently discovered angiogenesis mediators are involved in vascular development during embryogenesis and, interestingly, they are shared between the nervous and vascular systems. They represent new potential targets in the vasculature and suggest novel therapeutic opportunities.
Collapse
Affiliation(s)
- Brunella Cristofaro
- Division of Experimental Cardiovascular Medicine, Bristol Heart Institute, University of Bristol, Bristol, UK
| | | |
Collapse
|