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
|
Stewen J, Kruse K, Godoi-Filip AT, Zenia, Jeong HW, Adams S, Berkenfeld F, Stehling M, Red-Horse K, Adams RH, Pitulescu ME. Eph-ephrin signaling couples endothelial cell sorting and arterial specification. Nat Commun 2024; 15:2539. [PMID: 38570531 PMCID: PMC10991410 DOI: 10.1038/s41467-024-46300-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 02/21/2024] [Indexed: 04/05/2024] Open
Abstract
Cell segregation allows the compartmentalization of cells with similar fates during morphogenesis, which can be enhanced by cell fate plasticity in response to local molecular and biomechanical cues. Endothelial tip cells in the growing retina, which lead vessel sprouts, give rise to arterial endothelial cells and thereby mediate arterial growth. Here, we have combined cell type-specific and inducible mouse genetics, flow experiments in vitro, single-cell RNA sequencing and biochemistry to show that the balance between ephrin-B2 and its receptor EphB4 is critical for arterial specification, cell sorting and arteriovenous patterning. At the molecular level, elevated ephrin-B2 function after loss of EphB4 enhances signaling responses by the Notch pathway, VEGF and the transcription factor Dach1, which is influenced by endothelial shear stress. Our findings reveal how Eph-ephrin interactions integrate cell segregation and arteriovenous specification in the vasculature, which has potential relevance for human vascular malformations caused by EPHB4 mutations.
Collapse
Affiliation(s)
- Jonas Stewen
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Kai Kruse
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
- Bioinformatics Service Unit, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Anca T Godoi-Filip
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Zenia
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Hyun-Woo Jeong
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
- Sequencing Core Facility, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Susanne Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Frank Berkenfeld
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Martin Stehling
- Flow Cytometry Unit, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany
| | - Kristy Red-Horse
- Department of Biology, Stanford University, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford, CA, USA
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany.
| | - Mara E Pitulescu
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, D-48149, Münster, Germany.
| |
Collapse
|
3
|
Gomes B, Singh A, O'Sullivan JW, Schnurr TM, Goddard PC, Loong S, Amar D, Hughes JW, Kostur M, Haddad F, Salerno M, Foo R, Montgomery SB, Parikh VN, Meder B, Ashley EA. Genetic architecture of cardiac dynamic flow volumes. Nat Genet 2024; 56:245-257. [PMID: 38082205 DOI: 10.1038/s41588-023-01587-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 10/23/2023] [Indexed: 02/04/2024]
Abstract
Cardiac blood flow is a critical determinant of human health. However, the definition of its genetic architecture is limited by the technical challenge of capturing dynamic flow volumes from cardiac imaging at scale. We present DeepFlow, a deep-learning system to extract cardiac flow and volumes from phase-contrast cardiac magnetic resonance imaging. A mixed-linear model applied to 37,653 individuals from the UK Biobank reveals genome-wide significant associations across cardiac dynamic flow volumes spanning from aortic forward velocity to aortic regurgitation fraction. Mendelian randomization reveals a causal role for aortic root size in aortic valve regurgitation. Among the most significant contributing variants, localizing genes (near ELN, PRDM6 and ADAMTS7) are implicated in connective tissue and blood pressure pathways. Here we show that DeepFlow cardiac flow phenotyping at scale, combined with genotyping data, reinforces the contribution of connective tissue genes, blood pressure and root size to aortic valve function.
Collapse
Affiliation(s)
- Bruna Gomes
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Cardiology, Pneumology and Angiology, Heidelberg University Hospital, Heidelberg, Germany
- Informatics for Life, Heidelberg, Germany
| | - Aditya Singh
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Jack W O'Sullivan
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Theresia M Schnurr
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Pagé C Goddard
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Shaun Loong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Amar
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - J Weston Hughes
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Mykhailo Kostur
- Department of Cardiology, Pneumology and Angiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Francois Haddad
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Michael Salerno
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Roger Foo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Stephen B Montgomery
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Victoria N Parikh
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Benjamin Meder
- Department of Cardiology, Pneumology and Angiology, Heidelberg University Hospital, Heidelberg, Germany
- Informatics for Life, Heidelberg, Germany
| | - Euan A Ashley
- Departments of Medicine, Genetics, Computer Science and Biomedical Data Science, Stanford University, Stanford, CA, USA.
| |
Collapse
|
4
|
Abstract
Evidence implicating Eph receptor tyrosine kinases and their ephrin ligands (that together make up the 'Eph system') in cancer development and progression has been accumulating since the discovery of the first Eph receptor approximately 35 years ago. Advances in the past decade and a half have considerably increased the understanding of Eph receptor-ephrin signalling mechanisms in cancer and have uncovered intriguing new roles in cancer progression and drug resistance. This Review focuses mainly on these more recent developments. I provide an update on the different mechanisms of Eph receptor-ephrin-mediated cell-cell communication and cell autonomous signalling, as well as on the interplay of the Eph system with other signalling systems. I further discuss recent advances in elucidating how the Eph system controls tumour expansion, invasiveness and metastasis, supports cancer stem cells, and drives therapy resistance. In addition to functioning within cancer cells, the Eph system also mediates the reciprocal communication between cancer cells and cells of the tumour microenvironment. The involvement of the Eph system in tumour angiogenesis is well established, but recent findings also demonstrate roles in immune cells, cancer-associated fibroblasts and the extracellular matrix. Lastly, I discuss strategies under evaluation for therapeutic targeting of Eph receptors-ephrins in cancer and conclude with an outlook on promising future research directions.
Collapse
Affiliation(s)
- Elena B Pasquale
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
5
|
Crnkovic S, Rittchen S, Jandl K, Gindlhuber J, Zabini D, Mutgan AC, Valzano F, Boehm PM, Hoetzenecker K, Toller W, Veith C, Heinemann A, Schermuly RT, Olschewski A, Marsh LM, Kwapiszewska G. Divergent Roles of Ephrin-B2/EphB4 Guidance System in Pulmonary Hypertension. Hypertension 2023; 80:e17-e28. [PMID: 36519465 DOI: 10.1161/hypertensionaha.122.19479] [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: 12/23/2022]
Abstract
BACKGROUND Smooth muscle cell (SMC) expansion is one key morphological hallmark of pathologically altered vasculature and a characteristic feature of pulmonary vascular remodeling in pulmonary hypertension. Normal embryonal vessel maturation requires successful coverage of endothelial tubes with SMC, which is dependent on ephrin-B2 and EphB4 ligand-receptor guidance system. In this study, we investigated the potential role of ephrin-B2 and EphB4 on neomuscularization in adult pulmonary vascular disease. METHODS AND RESULTS Ephrin-B2 and EphB4 expression is preserved in smooth muscle and endothelial cells of remodeled pulmonary arteries. Chronic hypoxia-induced pulmonary hypertension was not ameliorated in mice with SMC-specific conditional ephrin-B2 knockout. In mice with global inducible ephrin-B2 knockout, pulmonary vascular remodeling and right ventricular hypertrophy upon chronic hypoxia exposure were significantly diminished compared to hypoxic controls, while right ventricular systolic pressure was unaffected. In contrast, EphB4 receptor kinase activity inhibition reduced right ventricular systolic pressure in hypoxia-induced pulmonary hypertension without affecting pulmonary vascular remodeling. Genetic deletion of ephrin-B2 in murine pulmonary artery SMC, and pharmacological inhibition of EphB4 in human pulmonary artery smooth muscle cells, blunted mitogen-induced cell proliferation. Loss of EphB4 signaling additionally reduced RhoA expression and weakened the interaction between human pulmonary artery smooth muscle cells and endothelial cells in a three-dimensional coculture model. CONCLUSIONS In sum, pulmonary vascular remodeling was dependent on ephrin-B2-induced Eph receptor (erythropoietin-producing hepatocellular carcinoma receptor) forward signaling in SMC, while EphB4 receptor activity was necessary for RhoA expression in SMC, interaction with endothelial cells and vasoconstrictive components of pulmonary hypertension.
Collapse
Affiliation(s)
- Slaven Crnkovic
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, Austria (S.C., D.Z., A.C.M., L.M.M., G.K.).,Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (S.C., S.R., K.J., J.G., D.Z., A.C.M., F.V., A.O., L.M.M., G.K.)
| | - Sonja Rittchen
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria (S.R., K.J., A.H.).,Otto Loewi Research Center, Division of Immunology, Medical University of Graz, Austria (S.R.).,Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (S.C., S.R., K.J., J.G., D.Z., A.C.M., F.V., A.O., L.M.M., G.K.)
| | - Katharina Jandl
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria (S.R., K.J., A.H.).,Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (S.C., S.R., K.J., J.G., D.Z., A.C.M., F.V., A.O., L.M.M., G.K.)
| | - Juergen Gindlhuber
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (S.C., S.R., K.J., J.G., D.Z., A.C.M., F.V., A.O., L.M.M., G.K.).,Department of Pathology, Medical University of Graz, Austria (J.G.)
| | - Diana Zabini
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, Austria (S.C., D.Z., A.C.M., L.M.M., G.K.).,Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (S.C., S.R., K.J., J.G., D.Z., A.C.M., F.V., A.O., L.M.M., G.K.)
| | - Ayse Ceren Mutgan
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, Austria (S.C., D.Z., A.C.M., L.M.M., G.K.).,Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (S.C., S.R., K.J., J.G., D.Z., A.C.M., F.V., A.O., L.M.M., G.K.)
| | - Francesco Valzano
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (S.C., S.R., K.J., J.G., D.Z., A.C.M., F.V., A.O., L.M.M., G.K.)
| | - Panja M Boehm
- Department of Thoracic Surgery, Medical University of Vienna, Austria (P.M.B., K.H.)
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, Austria (P.M.B., K.H.)
| | - Wolfgang Toller
- Department of Anesthesiology and Intensive Care Medicine, Medical University of Graz, Austria (W.T., A.O.)
| | - Christine Veith
- Excellence Cluster Cardio-Pulmonary Institute (CPI), University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, Germany (C.V.).,Faculty of Medicine, Justus Liebig University Giessen, Member of the German Lung Center (DZL), Germany (C.V., R.T.S.)
| | - Akos Heinemann
- Otto Loewi Research Center, Division of Pharmacology, Medical University of Graz, Austria (S.R., K.J., A.H.)
| | - Ralph T Schermuly
- Faculty of Medicine, Justus Liebig University Giessen, Member of the German Lung Center (DZL), Germany (C.V., R.T.S.)
| | - Andrea Olschewski
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (S.C., S.R., K.J., J.G., D.Z., A.C.M., F.V., A.O., L.M.M., G.K.).,Department of Anesthesiology and Intensive Care Medicine, Medical University of Graz, Austria (W.T., A.O.)
| | - Leigh M Marsh
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, Austria (S.C., D.Z., A.C.M., L.M.M., G.K.).,Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (S.C., S.R., K.J., J.G., D.Z., A.C.M., F.V., A.O., L.M.M., G.K.)
| | - Grazyna Kwapiszewska
- Otto Loewi Research Center, Division of Physiology, Medical University of Graz, Austria (S.C., D.Z., A.C.M., L.M.M., G.K.).,Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria (S.C., S.R., K.J., J.G., D.Z., A.C.M., F.V., A.O., L.M.M., G.K.).,Institute for Lung Health, Member of the German Lung Center (DZL), Giessen, Germany (G.K.)
| |
Collapse
|
6
|
Odimba U, Senthilselvan A, Farrell J, Gao Z. Identification of Sex-Specific Genetic Polymorphisms Associated with Asthma in Middle-Aged and Older Canadian Adults: An Analysis of CLSA Data. J Asthma Allergy 2023; 16:553-566. [PMID: 37197194 PMCID: PMC10184860 DOI: 10.2147/jaa.s404670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 04/29/2023] [Indexed: 05/19/2023] Open
Abstract
Purpose Asthma is a chronic heterogeneous respiratory disease resulting from a complex interplay between genetic variations and environmental exposures. There are sex disparities in the prevalence and severity of asthma in males and females. Asthma prevalence is higher in males during childhood but increases in females in adulthood. The mechanisms underlying these sex differences are not well understood; nevertheless, genetic variations, hormonal changes, and environmental influences are thought to play important roles. This study aimed to identify sex-specific genetic variants associated with asthma using CLSA genomic and questionnaire data. Methods First, we conducted a genome-wide SNP-by-sex interaction analysis on 23,323 individuals, examining 416,562 single nucleotide polymorphisms (SNPs) after quality control, followed by sex-stratified survey logistic regression of SNPs with interaction p-value less than 10¯5. Results Out of the 49 SNPs with interaction p-value less than 10-5, a sex-stratified survey logistic regression showed that five male-specific SNPs (rs6701638, rs17071077, rs254804, rs6013213, and rs2968822) in/near KIF26B, NMBR, PEPD, RTN4, and NFATC2 loci, and three female-specific SNPs (rs2968801, rs2864052, and rs9525931) in/near RTN4, and SERP2 loci were significantly associated with asthma after Bonferroni correction. An SNP (rs36213) in the EPHB1 gene was significantly associated with an increased risk of asthma in males [OR=1.35, 95% CI (1.14, 1.60)] but with a reduced risk of asthma in females [OR=0.84, 95% CI (0.76, 0.92)] after Bonferroni correction. Conclusion We discovered novel sex-specific genetic markers in/near the KIF26B, RTN4, EPHB1, NMBR, SERP2, PEPD, and NFATC2 genes that could potentially shed light on the sex differences in asthma susceptibility in males and females. Future mechanistic studies are required to understand better the underlying sex-related pathways of the identified loci in asthma development.
Collapse
Affiliation(s)
- Ugochukwu Odimba
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John’s, Newfoundland and Labrador, Canada
| | | | - Jamie Farrell
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John’s, Newfoundland and Labrador, Canada
- Faculty of Medicine, Health Science Centre (Respirology Department), Memorial University, St John’s, Newfoundland and Labrador, Canada
| | - Zhiwei Gao
- Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University, St. John’s, Newfoundland and Labrador, Canada
- Correspondence: Zhiwei Gao, Clinical Epidemiology Unit, Division of Community Health and Humanities, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland and Labrador, A1B 3V6, Canada, Tel +17098646523, Email
| |
Collapse
|
7
|
Chen D, Hughes ED, Saunders TL, Wu J, Hernández Vásquez MN, Makinen T, King PD. Angiogenesis depends upon EPHB4-mediated export of collagen IV from vascular endothelial cells. JCI Insight 2022; 7:156928. [PMID: 35015735 PMCID: PMC8876457 DOI: 10.1172/jci.insight.156928] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022] Open
Abstract
Capillary malformation-arteriovenous malformation (CM-AVM) is a blood vascular anomaly caused by inherited loss of function mutations in RASA1 or EPHB4 genes that encode p120 Ras GTPase-activating protein (p120 RasGAP/RASA1) and Ephrin receptor B4 (EPHB4) respectively. However, whether RASA1 and EPHB4 function in the same molecular signaling pathway to regulate the blood vasculature is uncertain. Here, we show that induced endothelial cell (EC)-specific disruption of Ephb4 in mice results in accumulation of collagen IV in the EC endoplasmic reticulum leading to EC apoptotic death and defective developmental, neonatal and pathological angiogenesis, as reported previously in induced EC-specific RASA1-deficient mice. Moreover, defects in angiogenic responses in EPHB4-deficient mice can be rescued by drugs that inhibit signaling through the Ras pathway and drugs that promote collagen IV export from the ER. However, EPHB4 mutant mice that express a form of EPHB4 that is unable to physically engage RASA1 but retains protein tyrosine kinase activity show normal angiogenic responses. These findings provide strong evidence that RASA1 and EPHB4 function in the same signaling pathway to protect against the development of CM-AVM independent of physical interaction and have important implications with regards possible means of treatment of this disease.
Collapse
Affiliation(s)
- Di Chen
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, United States of America
| | - Elizabeth D Hughes
- Transgenic Animal Model Core, University of Michigan Medical School, Ann Arbor, United States of America
| | - Thomas L Saunders
- Transgenic Animal Model Core, University of Michigan Medical School, Ann Arbor, United States of America
| | - Jiangping Wu
- Research Centre, Centre hospitalier de l'Université de Montréal, Montreal, Canada
| | | | - Taija Makinen
- Department of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Philip D King
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, United States of America
| |
Collapse
|
8
|
Wang T, Liu J, Liu H, Lee SR, Gonzalez L, Gorecka J, Shu C, Dardik A. Activation of EphrinB2 Signaling Promotes Adaptive Venous Remodeling in Murine Arteriovenous Fistulae. J Surg Res 2021; 262:224-239. [PMID: 33039109 PMCID: PMC8024410 DOI: 10.1016/j.jss.2020.08.071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 08/26/2020] [Accepted: 08/30/2020] [Indexed: 12/25/2022]
Abstract
BACKGROUND Arteriovenous fistulae (AVF) are the preferred mode of vascular access for hemodialysis. Before use, AVF remodel by thickening and dilating to achieve a functional conduit via an adaptive process characterized by expression of molecular markers characteristic of both venous and arterial identity. Although signaling via EphB4, a determinant of venous identity, mediates AVF maturation, the role of its counterpart EphrinB2, a determinant of arterial identity, remains unclear. We hypothesize that EphrinB2 signaling is active during AVF maturation and may be a mechanism of venous remodeling. METHODS Aortocaval fistulae were created or sham laparotomy was performed in C57Bl/6 mice, and specimens were examined on Days 7 or 21. EphrinB2 reverse signaling was activated with EphB4-Fc applied periadventitially in vivo and in endothelial cell culture medium in vitro. Downstream signaling was assessed using immunoblotting and immunofluorescence. RESULTS Venous remodeling during AVF maturation was characterized by increased expression of EphrinB2 as well as Akt1, extracellular signal-regulated kinases 1/2 (ERK1/2), and p38. Activation of EphrinB2 with EphB4-Fc increased phosphorylation of EphrinB2, endothelial nitric oxide synthase, Akt1, ERK1/2, and p38 and was associated with increased diameter and wall thickness in the AVF. Both mouse and human endothelial cells treated with EphB4-Fc increased phosphorylation of EphrinB2, endothelial nitric oxide synthase, Akt1, ERK1/2, and p38 and increased endothelial cell tube formation and migration. CONCLUSIONS Activation of EphrinB2 signaling by EphB4-Fc was associated with adaptive venous remodeling in vivo while activating endothelial cell function in vitro. Regulation of EphrinB2 signaling may be a new strategy to improve AVF maturation and patency.
Collapse
Affiliation(s)
- Tun Wang
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Jia Liu
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Haiyang Liu
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Shin-Rong Lee
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Luis Gonzalez
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Jolanta Gorecka
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut
| | - Chang Shu
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China; State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Alan Dardik
- The Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, Yale School of Medicine, New Haven, Connecticut; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, Connecticut; Department of Surgery, VA Connecticut Healthcare System, West Haven, Connecticut.
| |
Collapse
|
9
|
Liu L, Zeng P, Xue F, Yuan Z, Zhou X. Multi-trait transcriptome-wide association studies with probabilistic Mendelian randomization. Am J Hum Genet 2021; 108:240-256. [PMID: 33434493 PMCID: PMC7895847 DOI: 10.1016/j.ajhg.2020.12.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022] Open
Abstract
A transcriptome-wide association study (TWAS) integrates data from genome-wide association studies and gene expression mapping studies for investigating the gene regulatory mechanisms underlying diseases. Existing TWAS methods are primarily univariate in nature, focusing on analyzing one outcome trait at a time. However, many complex traits are correlated with each other and share a common genetic basis. Consequently, analyzing multiple traits jointly through multivariate analysis can potentially improve the power of TWASs. Here, we develop a method, moPMR-Egger (multiple outcome probabilistic Mendelian randomization with Egger assumption), for analyzing multiple outcome traits in TWAS applications. moPMR-Egger examines one gene at a time, relies on its cis-SNPs that are in potential linkage disequilibrium with each other to serve as instrumental variables, and tests its causal effects on multiple traits jointly. A key feature of moPMR-Egger is its ability to test and control for potential horizontal pleiotropic effects from instruments, thus maximizing power while minimizing false associations for TWASs. In simulations, moPMR-Egger provides calibrated type I error control for both causal effects testing and horizontal pleiotropic effects testing and is more powerful than existing univariate TWAS approaches in detecting causal associations. We apply moPMR-Egger to analyze 11 traits from 5 trait categories in the UK Biobank. In the analysis, moPMR-Egger identified 13.15% more gene associations than univariate approaches across trait categories and revealed distinct regulatory mechanisms underlying systolic and diastolic blood pressures.
Collapse
Affiliation(s)
- Lu Liu
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Ping Zeng
- Department of Epidemiology and Biostatistics, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Fuzhong Xue
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, MI 48109, USA; Center for Statistical Genetics, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
10
|
Wu T, Wang Y, Shi W, Zhang BQ, Raelson J, Yao YM, Wu HD, Xu ZX, Marois-Blanchet FC, Ledoux J, Blunck R, Sheng JZ, Hu SJ, Luo H, Wu J. A Variant in the Nicotinic Acetylcholine Receptor Alpha 3 Subunit Gene Is Associated With Hypertension Risks in Hypogonadic Patients. Front Genet 2020; 11:539862. [PMID: 33329690 PMCID: PMC7728919 DOI: 10.3389/fgene.2020.539862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 11/10/2020] [Indexed: 12/29/2022] Open
Abstract
Ephb6 gene knockout causes hypertension in castrated mice. EPHB6 controls catecholamine secretion by adrenal gland chromaffin cells (AGCCs) in a testosterone-dependent way. Nicotinic acetylcholine receptor (nAChR) is a ligand-gated Ca2+/Na+ channel, and its opening is the first signaling event leading to catecholamine secretion by AGCCs. There is a possibility that nAChR might be involved in EPHB6 signaling, and thus sequence variants of its subunit genes are associated with hypertension risks. CHRNA3 is the major subunit of nAChR used in human and mouse AGCCs. We conducted a human genetic study to assess the association of CHRNA3 variants with hypertension risks in hypogonadic males. The study cohort included 1,500 hypogonadic Chinese males with (750 patients) or without (750 patients) hypertension. The result revealed that SNV rs3743076 in the fourth intron of CHRNA3 was significantly associated with hypertension risks in the hypogonadic males. We further showed that EPHB6 physically interacted with CHRNA3 in AGCCs, providing a molecular basis for nAChR being in the EPHB6 signaling pathway.
Collapse
Affiliation(s)
- Tao Wu
- Institute of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yujia Wang
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.,Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Shi
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Bi-Qi Zhang
- Institute of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - John Raelson
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Yu-Mei Yao
- Department of Cardiology, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Huan-Dong Wu
- Institute of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zao-Xian Xu
- Institute of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | | | - Jonathan Ledoux
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Rikard Blunck
- Department of Physics, University of Montreal, Montreal, QC, Canada
| | - Jian-Zhong Sheng
- Department of Pathology and Physiopathology, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shen-Jiang Hu
- Institute of Cardiology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Hongyu Luo
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Jiangping Wu
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.,Nephrology Service, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| |
Collapse
|
11
|
NOX5-induced uncoupling of endothelial NO synthase is a causal mechanism and theragnostic target of an age-related hypertension endotype. PLoS Biol 2020; 18:e3000885. [PMID: 33170835 PMCID: PMC7654809 DOI: 10.1371/journal.pbio.3000885] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 09/18/2020] [Indexed: 02/07/2023] Open
Abstract
Hypertension is the most important cause of death and disability in the elderly. In 9 out of 10 cases, the molecular cause, however, is unknown. One mechanistic hypothesis involves impaired endothelium-dependent vasodilation through reactive oxygen species (ROS) formation. Indeed, ROS forming NADPH oxidase (Nox) genes associate with hypertension, yet target validation has been negative. We re-investigate this association by molecular network analysis and identify NOX5, not present in rodents, as a sole neighbor to human vasodilatory endothelial nitric oxide (NO) signaling. In hypertensive patients, endothelial microparticles indeed contained higher levels of NOX5—but not NOX1, NOX2, or NOX4—with a bimodal distribution correlating with disease severity. Mechanistically, mice expressing human Nox5 in endothelial cells developed—upon aging—severe systolic hypertension and impaired endothelium-dependent vasodilation due to uncoupled NO synthase (NOS). We conclude that NOX5-induced uncoupling of endothelial NOS is a causal mechanism and theragnostic target of an age-related hypertension endotype. Nox5 knock-in (KI) mice represent the first mechanism-based animal model of hypertension. The causes of hypertension are not understood; treatments are symptomatic and prevent only few of the associated risks. This study applies network medicine to identify a subgroup of patients with NADPH oxidase 5-induced uncoupling of nitric oxide synthase as the cause of age-related hypertension, enabling a first-in-class mechanism-based treatment of hypertension.
Collapse
|
12
|
Neuber C, Tröster A, Löser R, Belter B, Schwalbe H, Pietzsch J. The Pyrazolo[3,4- d]pyrimidine-Based Kinase Inhibitor NVP-BHG712: Effects of Regioisomers on Tumor Growth, Perfusion, and Hypoxia in EphB4-Positive A375 Melanoma Xenografts. Molecules 2020; 25:molecules25215115. [PMID: 33153234 PMCID: PMC7662635 DOI: 10.3390/molecules25215115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/02/2022] Open
Abstract
In a previous study, EphB4 was demonstrated to be a positive regulator of A375-melanoma growth but a negative regulator of tumor vascularization and perfusion. To distinguish between EphB4 forward and ephrinB2 reverse signaling, we used the commercially available EphB4 kinase inhibitor NVP-BHG712 (NVP), which was later identified as its regioisomer NVPiso. Since there have been reported significant differences between the inhibition profiles of NVP and NVPiso, we compared the influence of NVP and NVPiso on tumor characteristics under the same experimental conditions. Despite the different inhibitory profiles of NVP and NVPiso, the comparative study conducted here showed the same EphB4-induced effects in vivo as in the previous investigation. This confirmed the conclusion that EphB4-ephrinB2 reverse signaling is responsible for increased tumor growth as well as decreased tumor vascularization and perfusion. These results are further substantiated by microarrays showing differences between mock-transfected and EphB4-transfected (A375-EphB4) cells with respect to at least 9 angiogenesis-related proteins. Decreased expression of vascular endothelial growth factor (VEGF), angiotensin 1 (Ang-1), and protein kinase B (Akt/PKB), together with the increased expression of tissue inhibitor of metalloproteinase-1 (TIMP-1) and transforming growth factor beta-2 (TGF-β2), is consistent with the impaired vascularization of A375-EphB4 xenografts. Functional overexpression of EphB4 in A375-EphB4 cells was confirmed by activation of a variety of signaling pathways, including the Janus kinase/signal transducers and activators of transcription (JAK/STAT), rat sarcoma virus/rapidly accelerated fibrosarcoma/mitogen activated protein kinase kinase (Ras/Raf/MEK), and nuclear factor kappa-B (NFkB) pathways.
Collapse
Affiliation(s)
- Christin Neuber
- Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany; (C.N.); (R.L.); (B.B.)
| | - Alix Tröster
- Centre for Biomolecular Magnetic Resonance (BMRZ), Institute of Organic Chemistry and Chemical Biology, Johann Wolfgang Goethe-University Frankfurt a. M., Max-von-Laue-Strasse 7, 60438 Frankfurt, Germany; (A.T.); (H.S.)
| | - Reik Löser
- Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany; (C.N.); (R.L.); (B.B.)
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, 01069 Dresden, Germany
| | - Birgit Belter
- Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany; (C.N.); (R.L.); (B.B.)
| | - Harald Schwalbe
- Centre for Biomolecular Magnetic Resonance (BMRZ), Institute of Organic Chemistry and Chemical Biology, Johann Wolfgang Goethe-University Frankfurt a. M., Max-von-Laue-Strasse 7, 60438 Frankfurt, Germany; (A.T.); (H.S.)
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany; (C.N.); (R.L.); (B.B.)
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, 01069 Dresden, Germany
- Correspondence: ; Tel.: +49-351-260-2622
| |
Collapse
|
13
|
Yang C, Shu C, Wang L, Li X, He H, Li J, Zhu J, Yang Y, Dardik A. EphB4 signaling maintains the contractile phenotype of adult venous smooth muscle cells. Am J Transl Res 2020; 12:4522-4531. [PMID: 32913525 PMCID: PMC7476139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 07/18/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Autologous vein grafting remains the gold standard for surgical bypass grafts. However, vein bypasses still have significant incidence of failure. Ephrin type-B receptor 4 (EphB4), the embryonic venous determinant, may modulate vein graft adaptation. Although EphB4 is expressed in venous endothelial and smooth muscle cells (SMCs), it is not known whether EphB4 is functional in human SMCs. MATERIALS AND METHODS Human adult venous SMCs were obtained from the inferior vena cava of an adult human liver donor. Primary SMCs were stimulated with EphrinB2/Fc or transfected with an EphB4-expression vector (GV219-EphB4). Expression of SMC phenotype markers, migration, and proliferation were evaluated. RESULTS Activation of EphB4 with EphrinB2/Fc increased the mRNA and protein expression of the venous SMC contractile markers alpha actin, calponin-1, SM22, and MYH11, while decreasing the expression of the synthetic marker osteopontin. EphrinB2/Fc treatment inhibited SMC migration, but not proliferation. In addition, overexpression of EphB4 increased mRNA expression of SMC contractile markers, while decreasing expression of the apoptosis marker caspase-9. CONCLUSIONS EphB4 was present and functional in adult human venous SMCs. Stimulation of EphB4 increased expression of contractile SMC phenotypic markers and decreased SMC migration in vitro, functioning to retain the contractile phenotype of SMCs. EphB4 activation, therefore, recapitulates changes observed during vein graft adaptation to the arterial environment in vivo. EphB4 represents a new strategy to inhibit neointimal hyperplasia during vein graft adaption.
Collapse
Affiliation(s)
- Chenzi Yang
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South UniversityChangsha, Hunan, China
- Vascular Disease Institute, Central South UniversityChangsha, Hunan, China
| | - Chang Shu
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South UniversityChangsha, Hunan, China
- Vascular Disease Institute, Central South UniversityChangsha, Hunan, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, Peking Union Medical CollegeBeijing, China
| | - Lunchang Wang
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South UniversityChangsha, Hunan, China
- Vascular Disease Institute, Central South UniversityChangsha, Hunan, China
| | - Xin Li
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South UniversityChangsha, Hunan, China
- Vascular Disease Institute, Central South UniversityChangsha, Hunan, China
| | - Hao He
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South UniversityChangsha, Hunan, China
- Vascular Disease Institute, Central South UniversityChangsha, Hunan, China
| | - Jiehua Li
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South UniversityChangsha, Hunan, China
- Vascular Disease Institute, Central South UniversityChangsha, Hunan, China
| | - Jieting Zhu
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South UniversityChangsha, Hunan, China
- Vascular Disease Institute, Central South UniversityChangsha, Hunan, China
| | - Yibo Yang
- Department of Sport Surgery and Sport Medicine, Hunan Provincial People’s HospitalChangsha, Hunan, China
| | - Alan Dardik
- Department of Surgery, Yale School of MedicineNew Haven, CT, USA
| |
Collapse
|
14
|
EPHA4 regulates vascular smooth muscle cell contractility and is a sex-specific hypertension risk gene in individuals with type 2 diabetes. J Hypertens 2020; 37:775-789. [PMID: 30817459 DOI: 10.1097/hjh.0000000000001948] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE We investigated the association of genetic variants of EPHA4, a receptor tyrosine kinase, with hypertension, and its role in vascular smooth muscle cell (VSMC) contractility. METHODS Data from two human genetic studies, ADVANCE and HCHS/SOL, were analyzed for association of EPHA4 single nucleotide variants (SNVs) with hypertension risks. The effect of EPHA4 signalling on mouse VSMC contractility was assessed. RESULTS We identified a SNV (rs75843691 hg19 chr2:g.222395371 C>G), located in the third intron of EPHA4 gene, being significantly associated with hypertension in human female patients (P value = 8.3 × 10, below the Bonferroni-corrected critical P value) but not male patients with type 2 diabetes from the ADVANCE clinical trial. We found that EPHA4 was expressed in VSMCs and its stimulation by anti-EPHA4 antibody led to reduced VSMC contractility. Estrogen enhanced the contractility-lowering effect of EPHA4 stimulation. Conversely, siRNA knockdown of Epha4 expression in VSMCs resulted in increased contractility of VSMCs from female mice but not from male mice. CONCLUSION EPHA4 appears to be a sex-specific hypertension risk gene in type 2 diabetic patients. Forward EPHA4 signalling reduces VSMC contractility, and estrogen is a modifier of this effect. The effect of EPHA4 on VSMCs contractility explains the association of EPHA4 gene with hypertension risks in female patients.
Collapse
|
15
|
Shi W, Ye B, Rame M, Wang Y, Cioca D, Reibel S, Peng J, Qi S, Vitale N, Luo H, Wu J. The receptor tyrosine kinase EPHB6 regulates catecholamine exocytosis in adrenal gland chromaffin cells. J Biol Chem 2020; 295:7653-7668. [PMID: 32321761 DOI: 10.1074/jbc.ra120.013251] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/20/2020] [Indexed: 11/06/2022] Open
Abstract
The erythropoietin-producing human hepatocellular receptor EPH receptor B6 (EPHB6) is a receptor tyrosine kinase that has been shown previously to control catecholamine synthesis in the adrenal gland chromaffin cells (AGCCs) in a testosterone-dependent fashion. EPHB6 also has a role in regulating blood pressure, but several facets of this regulation remain unclear. Using amperometry recordings, we now found that catecholamine secretion by AGCCs is compromised in the absence of EPHB6. AGCCs from male knockout (KO) mice displayed reduced cortical F-actin disassembly, accompanied by decreased catecholamine secretion through exocytosis. This phenotype was not observed in AGCCs from female KO mice, suggesting that testosterone, but not estrogen, contributes to this phenotype. Of note, reverse signaling from EPHB6 to ephrin B1 (EFNB1) and a 7-amino acid-long segment in the EFNB1 intracellular tail were essential for the regulation of catecholamine secretion. Further downstream, the Ras homolog family member A (RHOA) and FYN proto-oncogene Src family tyrosine kinase (FYN)-proto-oncogene c-ABL-microtubule-associated monooxygenase calponin and LIM domain containing 1 (MICAL-1) pathways mediated the signaling from EFNB1 to the defective F-actin disassembly. We discuss the implications of EPHB6's effect on catecholamine exocytosis and secretion for blood pressure regulation.
Collapse
Affiliation(s)
- Wei Shi
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Bei Ye
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada.,Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Marion Rame
- Institut des Neurosciences Cellulaires et Intégratives, UPR-3212 Centre National de la Recherche Scientifique and Université de Strasbourg, Strasbourg, France
| | - Yujia Wang
- Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | | | | | - Junzheng Peng
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Shijie Qi
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Intégratives, UPR-3212 Centre National de la Recherche Scientifique and Université de Strasbourg, Strasbourg, France
| | - Hongyu Luo
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Jiangping Wu
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada .,Nephrology Department, CHUM, Montreal, Quebec, Canada
| |
Collapse
|
16
|
Liu S, Jiang X, Lu H, Xing M, Qiao Y, Zhang C, Zhang W. HuR (Human Antigen R) Regulates the Contraction of Vascular Smooth Muscle and Maintains Blood Pressure. Arterioscler Thromb Vasc Biol 2020; 40:943-957. [PMID: 32075416 DOI: 10.1161/atvbaha.119.313897] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE HuR (human antigen R)-an RNA-binding protein-is involved in regulating mRNA stability by binding adenylate-uridylate-rich elements. This study explores the role of HuR in the regulation of smooth muscle contraction and blood pressure. Approach and Results: Vascular HuRSMKO (smooth muscle-specific HuR knockout) mice were generated by crossbreeding HuRflox/flox mice with α-SMA (α-smooth muscle actin)-Cre mice. As compared with CTR (control) mice, HuRSMKO mice showed hypertension and cardiac hypertrophy. HuR levels were decreased in aortas from hypertensive patients and SHRs (spontaneously hypertensive rats), and overexpression of HuR could lower the blood pressure of SHRs. Contractile response to vasoconstrictors was increased in mesenteric artery segments isolated from HuRSMKO mice. The functional abnormalities in HuRSMKO mice were attributed to decreased mRNA and protein levels of RGS (regulator of G-protein signaling) protein(s) RGS2, RGS4, and RGS5, which resulted in increased intracellular calcium increase. Consistently, the degree of intracellular calcium ion increase in HuR-deficient smooth muscle cells was reduced by overexpression of RGS2, RGS4, or RGS5. Finally, administration of RGS2 and RGS5 reversed the elevated blood pressure in HuRSMKO mice. CONCLUSIONS Our findings indicate that HuR regulates vascular smooth muscle contraction and maintains blood pressure by modulating RGS expression.
Collapse
Affiliation(s)
- Shanshan Liu
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine (S.L., H.L., C.Z., W.Z.), Qilu Hospital of Shandong University, Jinan, China
| | - Xiuxin Jiang
- Department of General Surgery (X.J.), Qilu Hospital of Shandong University, Jinan, China
| | - Hanlin Lu
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine (S.L., H.L., C.Z., W.Z.), Qilu Hospital of Shandong University, Jinan, China
| | - Mengdan Xing
- Department of Cognitive Neuroscience, The Key Laboratory of MOE for Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, China (M.X., Y.Q.)
| | - Yanning Qiao
- Department of Cognitive Neuroscience, The Key Laboratory of MOE for Modern Teaching Technology, Center for Teacher Professional Ability Development, Shaanxi Normal University, Xi'an, China (M.X., Y.Q.)
| | - Cheng Zhang
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine (S.L., H.L., C.Z., W.Z.), Qilu Hospital of Shandong University, Jinan, China
| | - Wencheng Zhang
- From the Department of Cardiology, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine (S.L., H.L., C.Z., W.Z.), Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
17
|
Luxán G, Stewen J, Díaz N, Kato K, Maney SK, Aravamudhan A, Berkenfeld F, Nagelmann N, Drexler HC, Zeuschner D, Faber C, Schillers H, Hermann S, Wiseman J, Vaquerizas JM, Pitulescu ME, Adams RH. Endothelial EphB4 maintains vascular integrity and transport function in adult heart. eLife 2019; 8:45863. [PMID: 31782728 PMCID: PMC6884395 DOI: 10.7554/elife.45863] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 11/15/2019] [Indexed: 12/17/2022] Open
Abstract
The homeostasis of heart and other organs relies on the appropriate provision of nutrients and functional specialization of the local vasculature. Here, we have used mouse genetics, imaging and cell biology approaches to investigate how homeostasis in the adult heart is controlled by endothelial EphB4 and its ligand ephrin-B2, which are known regulators of vascular morphogenesis and arteriovenous differentiation during development. We show that inducible and endothelial cell-specific inactivation of Ephb4 in adult mice is compatible with survival, but leads to rupturing of cardiac capillaries, cardiomyocyte hypertrophy, and pathological cardiac remodeling. In contrast, EphB4 is not required for integrity and homeostasis of capillaries in skeletal muscle. Our analysis of mutant mice and cultured endothelial cells shows that EphB4 controls the function of caveolae, cell-cell adhesion under mechanical stress and lipid transport. We propose that EphB4 maintains critical functional properties of the adult cardiac vasculature and thereby prevents dilated cardiomyopathy-like defects.
Collapse
Affiliation(s)
- Guillermo Luxán
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Jonas Stewen
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Noelia Díaz
- Regulatory Genomics Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Katsuhiro Kato
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Sathish K Maney
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Anusha Aravamudhan
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Frank Berkenfeld
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Nina Nagelmann
- Department of Clinical Radiology, University Hospital Münster, Münster, Germany
| | - Hannes Ca Drexler
- Bioanalytical Mass Spectrometry Unit, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Dagmar Zeuschner
- Electron Microscopy Unit, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Cornelius Faber
- Department of Clinical Radiology, University Hospital Münster, Münster, Germany
| | - Hermann Schillers
- Institute for Physiology II, University of Münster, Münster, Germany
| | - Sven Hermann
- European Institute for Molecular Imaging, University of Münster, Münster, Germany
| | - John Wiseman
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Juan M Vaquerizas
- Regulatory Genomics Laboratory, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Mara E Pitulescu
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Faculty of Medicine, University of Münster, Münster, Germany
| |
Collapse
|
18
|
Brown IAM, Diederich L, Good ME, DeLalio LJ, Murphy SA, Cortese-Krott MM, Hall JL, Le TH, Isakson BE. Vascular Smooth Muscle Remodeling in Conductive and Resistance Arteries in Hypertension. Arterioscler Thromb Vasc Biol 2019; 38:1969-1985. [PMID: 30354262 DOI: 10.1161/atvbaha.118.311229] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is a leading cause of death worldwide and accounts for >17.3 million deaths per year, with an estimated increase in incidence to 23.6 million by 2030. 1 Cardiovascular death represents 31% of all global deaths 2 -with stroke, heart attack, and ruptured aneurysms predominantly contributing to these high mortality rates. A key risk factor for cardiovascular disease is hypertension. Although treatment or reduction in hypertension can prevent the onset of cardiovascular events, existing therapies are only partially effective. A key pathological hallmark of hypertension is increased peripheral vascular resistance because of structural and functional changes in large (conductive) and small (resistance) arteries. In this review, we discuss the clinical implications of vascular remodeling, compare the differences between vascular smooth muscle cell remodeling in conductive and resistance arteries, discuss the genetic factors associated with vascular smooth muscle cell function in hypertensive patients, and provide a prospective assessment of current and future research and pharmacological targets for the treatment of hypertension.
Collapse
Affiliation(s)
- Isola A M Brown
- From the Robert M. Berne Cardiovascular Research Center (I.A.M.B., M.E.G., L.J.D., S.A.M., B.E.I.)
| | - Lukas Diederich
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University, Dusseldorf, Germany (L.D., M.M.C.-K.)
| | - Miranda E Good
- From the Robert M. Berne Cardiovascular Research Center (I.A.M.B., M.E.G., L.J.D., S.A.M., B.E.I.)
| | - Leon J DeLalio
- From the Robert M. Berne Cardiovascular Research Center (I.A.M.B., M.E.G., L.J.D., S.A.M., B.E.I.).,Department of Pharmacology (L.J.D.)
| | - Sara A Murphy
- From the Robert M. Berne Cardiovascular Research Center (I.A.M.B., M.E.G., L.J.D., S.A.M., B.E.I.)
| | - Miriam M Cortese-Krott
- Cardiovascular Research Laboratory, Division of Cardiology, Pneumology and Angiology, Medical Faculty, Heinrich Heine University, Dusseldorf, Germany (L.D., M.M.C.-K.)
| | - Jennifer L Hall
- Lillehei Heart Institute (J.L.H.).,Division of Cardiology, Department of Medicine (J.L.H.), University of Minnesota, Minneapolis.,American Heart Association, Dallas, TX (J.L.H.)
| | - Thu H Le
- Division of Nephrology, Department of Medicine (T.H.L.)
| | - Brant E Isakson
- From the Robert M. Berne Cardiovascular Research Center (I.A.M.B., M.E.G., L.J.D., S.A.M., B.E.I.).,Department of Molecular Physiology and Biophysics (B.E.I.), University of Virginia School of Medicine, Charlottesville
| |
Collapse
|
19
|
Zhu M, Hua Y, Tang J, Zhao X, Zhang L, Zhang Y. Lentiviral-mediated ephrin B2 gene modification of rat bone marrow mesenchymal stem cells. J Int Med Res 2019; 47:3282-3298. [PMID: 31122164 PMCID: PMC6683898 DOI: 10.1177/0300060519843023] [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: 11/23/2022] Open
Abstract
Objective To determine the effect of the upregulation or knockdown of the ephrinB2 (Efnb2) gene and the effect of EphB4/EphrinB2 signalling in rat bone marrow mesenchymal stem cells (BMSCs). Methods Rat BMSCs were infected with lentivirus vectors carrying EphrinB2 and shRNA-EphrinB2. EphrinB2 mRNA and protein levels were quantified. At 28 days of culture with neuronal cell-conditioned differentiation medium, levels of microtubule-associated protein 2 (MAP2), CD133 and nestin were detected in EphrinB2/BMSCs and shEphrinB2/BMSCs using quantitative polymerase chain reaction and immunofluorescence. The ability of these cells to migrate was evaluated using a transwell assay. Results BMSCs were successfully isolated as indicated by their CD90+ CD29+ CD34– CD45– phenotype. Three days after ephrinB2 transduction, BMSC cell bodies began to shrink and differentiate into neuron-like cells. At 28 days, levels of MAP2, CD133 and nestin, as well as the number of migratory cells, were higher in lenti-EphrinB2-BMSCs than in the two control groups. The shEphrinB2/BMSCs had reduced levels of MAP2, CD133 and nestin; and a lower rate of cell migration. Similarly, increased levels of Grb4 andp21-activated kinase in the EphB4/EphrinB2 reverse signalling pathway were observed by Western blot. Conclusions LV-EphrinB2 can be efficiently transduced into BMSCs, which then differentiate into neuron-like cells.
Collapse
Affiliation(s)
- Min Zhu
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yu Hua
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Jian Tang
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiaoke Zhao
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ling Zhang
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Yue Zhang
- Department of Rehabilitation, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
20
|
Shi W, Wang Y, Peng J, Qi S, Vitale N, Kaneda N, Murata T, Luo H, Wu J. EPHB6 controls catecholamine biosynthesis by up-regulating tyrosine hydroxylase transcription in adrenal gland chromaffin cells. J Biol Chem 2019; 294:6871-6887. [PMID: 30824540 DOI: 10.1074/jbc.ra118.005767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/25/2019] [Indexed: 11/06/2022] Open
Abstract
EPHB6 is a member of the erythropoietin-producing hepatocellular kinase (EPH) family and a receptor tyrosine kinase with a dead kinase domain. It is involved in blood pressure regulation and adrenal gland catecholamine (CAT) secretion, but several facets of EPHB6-mediated CAT regulation are unclear. In this study, using biochemical, quantitative RT-PCR, immunoblotting, and gene microarray assays, we found that EPHB6 up-regulates CAT biosynthesis in adrenal gland chromaffin cells (AGCCs). We observed that epinephrine content is reduced in the AGCCs from male Ephb6-KO mice, caused by decreased expression of tyrosine hydroxylase, the rate-limiting enzyme in CAT biosynthesis. We demonstrate that the signaling pathway from EPHB6 to tyrosine hydroxylase expression in AGCCs involves Rac family small GTPase 1 (RAC1), MAP kinase kinase 7 (MKK7), c-Jun N-terminal kinase (JNK), proto-oncogene c-Jun, activator protein 1 (AP1), and early growth response 1 (EGR1). On the other hand, signaling via extracellular signal-regulated kinase (ERK1/2), p38 mitogen-activated protein kinase, and ELK1, ETS transcription factor (ELK1) was not affected by EPHB6 deletion. We further report that EPHB6's effect on AGCCs was via reverse signaling through ephrin B1 and that EPHB6 acted in concert with the nongenomic effect of testosterone to control CAT biosynthesis. Our findings elucidate the mechanisms by which EPHB6 modulates CAT biosynthesis and identify potential therapeutic targets for diseases, such as hypertension, caused by dysfunctional CAT biosynthesis.
Collapse
Affiliation(s)
- Wei Shi
- From the Research Centre and
| | - Yujia Wang
- From the Research Centre and.,the Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | | | | | - Nicolas Vitale
- the Institut des Neurosciences Cellulaires et Intégratives, UPR-3212, CNRS-Université de Strasbourg, 5 rue Blaise Pascal, 67000 Strasbourg, France, and
| | - Norio Kaneda
- the Department of Analytical Neurobiology, Faculty of Pharmacy, Meijo University, Tempaku, Nagoya 4688503, Japan
| | - Tomiyasu Murata
- the Department of Analytical Neurobiology, Faculty of Pharmacy, Meijo University, Tempaku, Nagoya 4688503, Japan
| | | | - Jiangping Wu
- From the Research Centre and .,Nephrology Department, Centre Hospitalier de l'Université de Montréal Montreal, Quebec, H2X 0A9, Canada
| |
Collapse
|
21
|
Zeng X, Hunt A, Jin SC, Duran D, Gaillard J, Kahle KT. EphrinB2-EphB4-RASA1 Signaling in Human Cerebrovascular Development and Disease. Trends Mol Med 2019; 25:265-286. [PMID: 30819650 DOI: 10.1016/j.molmed.2019.01.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/17/2019] [Accepted: 01/29/2019] [Indexed: 12/13/2022]
Abstract
Recent whole exome sequencing studies in humans have provided novel insight into the importance of the ephrinB2-EphB4-RASA1 signaling axis in cerebrovascular development, corroborating and extending previous work in model systems. Here, we aim to review the human cerebrovascular phenotypes associated with ephrinB2-EphB4-RASA1 mutations, including those recently discovered in Vein of Galen malformation: the most common and severe brain arteriovenous malformation in neonates. We will also discuss emerging paradigms of the molecular and cellular pathophysiology of disease-causing ephrinB2-EphB4-RASA1 mutations, including the potential role of somatic mosaicism. These observations have potential diagnostic and therapeutic implications for patients with rare congenital cerebrovascular diseases and their families.
Collapse
Affiliation(s)
- Xue Zeng
- Department of Genetics, Yale School of Medicine, New Haven CT, USA; Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Ava Hunt
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
| | - Sheng Chih Jin
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Daniel Duran
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
| | - Jonathan Gaillard
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale School of Medicine, New Haven CT, USA; Department of Pediatrics, Yale School of Medicine, New Haven CT, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven CT, USA.
| |
Collapse
|
22
|
Wu T, Zhang BQ, Raelson J, Yao YM, Wu HD, Xu ZX, Marois-Blanchet FC, Tahir MR, Wang Y, Bradley WE, Luo H, Wu J, Sheng JZ, Hu SJ. Analysis of the association of EPHB6, EFNB1 and EFNB3 variants with hypertension risks in males with hypogonadism. Sci Rep 2018; 8:14497. [PMID: 30262919 PMCID: PMC6160468 DOI: 10.1038/s41598-018-32836-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 09/14/2018] [Indexed: 12/29/2022] Open
Abstract
Several members of the EPH kinase family and their ligands are involved in blood pressure regulation, and such regulation is often sex- or sex hormone-dependent, based on animal and human genetic studies. EPHB6 gene knockout (KO) in mice leads to hypertension in castrated males but not in un-manipulated KO males or females. To assess whether this finding in mice is relevant to human hypertension, we conducted a human genetic study for the association of EPHB6 and its two ligands, EFNB1 and EFNB3, with hypertension in hypogonadic patients. Seven hundred and fifty hypertensive and 750 normotensive Han Chinese patients, all of whom were hypogonadic, were genotyped for single nucleotide polymorphisms (SNPs) within the regions of the genes, plus an additional 50 kb 5′ of the genes for EPHB6, EFNB1 and EFNB3. An imputed insertion/deletion polymorphism, rs35530071, was found to be associated with hypertension at p-values below the Bonferroni-corrected significance level of 0.0024. This marker is located 5′ upstream of the EFNB3 gene start site. Previous animal studies showed that while male EFNB3 gene knockout mice were normotensive, castration of these mice resulted in hypertension, corroborating the results of the human genetic study. Considering the significant associations of EFNB3 SNPs with hypertension in hypogonadic males and supporting evidence from castrated EFNB3 KO mice, we conclude that loss-of-function variants of molecules in the EPHB6 signaling pathway in the presence of testosterone are protective against hypertension in humans.
Collapse
Affiliation(s)
- Tao Wu
- Institute of Cardiology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Bi-Qi Zhang
- Institute of Cardiology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - John Raelson
- Research Centre, Centre hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, H2X 0A9, Canada
| | - Yu-Mei Yao
- Department of Cardiology, Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310005, China
| | - Huan-Dong Wu
- Institute of Cardiology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zao-Xian Xu
- Institute of Cardiology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China
| | | | - Muhammad Ramzan Tahir
- Research Centre, Centre hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, H2X 0A9, Canada
| | - Yujia Wang
- Research Centre, Centre hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, H2X 0A9, Canada.,Children's Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310003, China
| | - W Edward Bradley
- Research Centre, Centre hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, H2X 0A9, Canada
| | - Hongyu Luo
- Research Centre, Centre hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, H2X 0A9, Canada
| | - Jiangping Wu
- Research Centre, Centre hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, H2X 0A9, Canada. .,Nephrology Service, Centre hospitalier de l'Université de Montréal (CHUM), Montreal, Quebec, H2X 0A9, Canada.
| | - Jian-Zhong Sheng
- Department of Pathology and Physiopathology, College of Medicine, Zhejiang University, Hangzhou, 310005, China.
| | - Shen-Jiang Hu
- Institute of Cardiology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
23
|
Finney AC, Orr AW. Guidance Molecules in Vascular Smooth Muscle. Front Physiol 2018; 9:1311. [PMID: 30283356 PMCID: PMC6157320 DOI: 10.3389/fphys.2018.01311] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 08/30/2018] [Indexed: 12/21/2022] Open
Abstract
Several highly conserved families of guidance molecules, including ephrins, Semaphorins, Netrins, and Slits, play conserved and distinct roles in tissue remodeling during tissue patterning and disease pathogenesis. Primarily, these guidance molecules function as either secreted or surface-bound ligands that interact with their receptors to activate a variety of downstream effects, including cell contractility, migration, adhesion, proliferation, and inflammation. Vascular smooth muscle cells, contractile cells comprising the medial layer of the vessel wall and deriving from the mural population, regulate vascular tone and blood pressure. While capillaries lack a medial layer of vascular smooth muscle, mural-derived pericytes contribute similarly to capillary tone to regulate blood flow in various tissues. Furthermore, pericyte coverage is critical in vascular development, as perturbations disrupt vascular permeability and viability. During cardiovascular disease, smooth muscle cells play a more dynamic role in which suppression of contractile markers, enhanced proliferation, and migration lead to the progression of aberrant vascular remodeling. Since many types of guidance molecules are expressed in vascular smooth muscle and pericytes, these may contribute to blood vessel formation and aberrant remodeling during vascular disease. While vascular development is a large focus of the existing literature, studies emerged to address post-developmental roles for guidance molecules in pathology and are of interest as novel therapeutic targets. In this review, we will discuss the roles of guidance molecules in vascular smooth muscle and pericyte function in development and disease.
Collapse
Affiliation(s)
- Alexandra Christine Finney
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
| | - Anthony Wayne Orr
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
- Department of Pathology and Translational Medicine, Louisiana State University Health Sciences Center Shreveport, Shreveport, LA, United States
| |
Collapse
|
24
|
Wang Y, Shi W, Blanchette A, Peng J, Qi S, Luo H, Ledoux J, Wu J. EPHB6 and testosterone in concert regulate epinephrine release by adrenal gland chromaffin cells. Sci Rep 2018; 8:842. [PMID: 29339804 PMCID: PMC5770418 DOI: 10.1038/s41598-018-19215-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 12/27/2017] [Indexed: 12/22/2022] Open
Abstract
Erythropoietin-producing human hepatocellular receptor (EPH) B6 (EPHB6) is a member of the receptor tyrosine kinase family. We previously demonstrated that EPHB6 knockout reduces catecholamine secretion in male but not female mice, and castration reverses this phenotype. We showed here that male EPHB6 knockout adrenal gland chromaffin cells presented reduced acetylcholine-triggered Ca2+ influx. Such reduction depended on the non-genomic effect of testosterone. Increased large conductance calcium-activated potassium channel current densities were recorded in adrenal gland chromaffin cells from male EPHB6 knockout mice but not from castrated knockout or female knockout mice. Blocking of the large conductance calcium-activated potassium channel in adrenal gland chromaffin cells from male knockout mice corrected their reduced Ca2+ influx. We conclude that the absence of EPHB6 and the presence of testosterone would lead to augmented large conductance calcium-activated potassium channel currents, which limit voltage-gated calcium channel opening in adrenal gland chromaffin cells. Consequently, acetylcholine-triggered Ca2+ influx is reduced, leading to lower catecholamine release in adrenal gland chromaffin cells from male knockout mice. This explains the reduced resting-state blood catecholamine levels, and hence the blood pressure, in male but not female EPHB6 knock mice. These findings have certain clinical implications.
Collapse
Affiliation(s)
- Yujia Wang
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, H2X 0A9, Canada
- The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Wei Shi
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, H2X 0A9, Canada
| | | | - Junzheng Peng
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, H2X 0A9, Canada
| | - Shijie Qi
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, H2X 0A9, Canada
| | - Hongyu Luo
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, H2X 0A9, Canada.
| | - Jonathan Ledoux
- Montreal Heart Institute, Montreal, Quebec, H1T 1C8, Canada.
| | - Jiangping Wu
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, H2X 0A9, Canada.
- Nephrology Department, CHUM, Montreal, Quebec, H2L 4M1, Canada.
| |
Collapse
|
25
|
Kim M, Yoo HJ, Kim M, Kim J, Baek SH, Song M, Lee JH. EPHA6 rs4857055 C > T polymorphism associates with hypertension through triglyceride and LDL particle size in the Korean population. Lipids Health Dis 2017; 16:230. [PMID: 29208002 PMCID: PMC5718072 DOI: 10.1186/s12944-017-0620-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 11/22/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Erythropoietin-producing human hepatocellular (Eph) receptors might contribute to the development of atherosclerosis. A genome-wide association study indicated that the Eph receptor A6 gene (EPHA6) associated with at least 1 blood pressure (BP) phenotype. The objective of the present study was to determine whether EPHA6 is a novel candidate gene for hypertension in a Korean population. METHODS A total 2146 study participants with normotension and hypertension were included. Genotype data were obtained using a Korean Chip. To assess the association between single-nucleotide polymorphisms (SNPs) and BP, we performed a linear regression analysis, which showed that rs4850755 in the EPHA6 gene was the SNP most highly associated with both systolic and diastolic BP. RESULTS The presence of the TT genotype of the EPHA6 rs4857055 C > T SNP was associated with a higher risk of hypertension after adjusting for age, sex, body mass index (BMI), smoking, and drinking [odds ratio 1.533, P = 0.001]. In the control group, significant associations were observed between systolic BP and the rs4857055 polymorphism and between diastolic BP and the rs4857055 polymorphism. In the hypertension group, a significant association was observed between systolic BP and the rs4857055 polymorphism. In the hypertension group, subjects with the TT genotype showed significantly higher systolic BP than CC subjects. Additionally, in the hypertension group, TT carriers showed a higher tendency of serum triglyceride (P = 0.069) and significantly higher apolipoprotein B (P = 0.015) and smaller low-density lipoprotein (LDL) particle size (P < 0.001) than either TC or CC subjects. CONCLUSIONS These results could suggest that the EPHA6 rs4857055 C > T SNP is a novel candidate gene for hypertension in the Korean population. Additionally, the TT genotype could be associated with hypertriglyceridemia and small LDL particle size in hypertension.
Collapse
Affiliation(s)
- Minjoo Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, 03722, Korea
| | - Hye Jin Yoo
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
| | - Minkyung Kim
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, 03722, Korea
| | - Jiyoo Kim
- Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, 03722, Korea.,National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, 03722, Korea
| | - Seung Han Baek
- Institute of Convergence Technology, Yonsei University, Seoul, 03722, Korea
| | - Min Song
- Department of Library and Information Science, Yonsei University, Seoul, 03722, Korea
| | - Jong Ho Lee
- Research Center for Silver Science, Institute of Symbiotic Life-TECH, Yonsei University, Seoul, 03722, Korea. .,Department of Food and Nutrition, Brain Korea 21 PLUS Project, College of Human Ecology, Yonsei University, Seoul, 03722, Korea. .,National Leading Research Laboratory of Clinical Nutrigenetics/Nutrigenomics, Department of Food and Nutrition, College of Human Ecology, Yonsei University, Seoul, 03722, Korea.
| |
Collapse
|
26
|
Tremblay J, Wang Y, Raelson J, Marois-Blanchet FC, Wu Z, Luo H, Bradley E, Chalmers J, Woodward M, Harrap S, Hamet P, Wu J. Evidence from single nucleotide polymorphism analyses of ADVANCE study demonstrates EFNB3 as a hypertension risk gene. Sci Rep 2017; 7:44114. [PMID: 28272517 PMCID: PMC5341021 DOI: 10.1038/srep44114] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 02/03/2017] [Indexed: 01/11/2023] Open
Abstract
EPH kinases and their ligands, ephrins (EFNs), have vital and diverse biological functions. We recently reported that Efnb3 gene deletion results in hypertension in female but not male mice. These data suggest that EFNB3 regulates blood pressure in a sex- and sex hormone-dependent way. In the present study, we conducted a human genetic study to assess the association of EFNB3 single nucleotide polymorphisms with human hypertension risks, using 3,448 patients with type 2 diabetes from the ADVANCE study (Action in Diabetes and Vascular Disease: Peterax and Diamicron MR Controlled Evaluation). We have observed significant association between 2 SNPs in the 3′ untranslated region or within the adjacent region just 3′ of the EFNB3 gene with hypertension, corroborating our findings from the mouse model. Thus, our investigation has shown that EFNB3 is a hypertension risk gene in certain individuals.
Collapse
Affiliation(s)
- Johanne Tremblay
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Yujia Wang
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - John Raelson
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | | | - Zenghui Wu
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Hongyu Luo
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Edward Bradley
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - John Chalmers
- The George Institute for Global Health, University of Sydney Sydney, New South Wales, 2006, Australia
| | - Mark Woodward
- The George Institute for Global Health, University of Sydney Sydney, New South Wales, 2006, Australia.,The George Institute for Global Health, University of Oxford, Oxford, UK
| | - Stephen Harrap
- Department of Epidemiology, Johns Hopkins University, Baltimore MD, USA.,Department of Physiology, University of Melbourne, Victoria 3010, Australia
| | - Pavel Hamet
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| | - Jiangping Wu
- Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec H2X 0A9, Canada
| |
Collapse
|
27
|
The role of GRIP1 and ephrin B3 in blood pressure control and vascular smooth muscle cell contractility. Sci Rep 2016; 6:38976. [PMID: 27941904 PMCID: PMC5150233 DOI: 10.1038/srep38976] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/16/2016] [Indexed: 12/25/2022] Open
Abstract
Several erythropoietin-producing hepatocellular receptor B family (EPHB) and their ligands, ephrinBs (EFNBs), are involved in blood pressure regulation in animal models. We selected 528 single nucleotide polymorphisms (SNPs) within the genes of EPHB6, EFNB2, EFNB3 and GRIP1 in the EPH/EFN signalling system to query the International Blood Pressure Consortium dataset. A SNP within the glutamate receptor interacting protein 1 (GRIP1) gene presented a p-value of 0.000389, approaching the critical p-value of 0.000302, for association with diastolic blood pressure of 60,396 individuals. According to echocardiography, we found that Efnb3 gene knockout mice showed enhanced constriction in the carotid arteries. In vitro studies revealed that in mouse vascular smooth muscle cells, siRNA knockdown of GRIP1, which is in the EFNB3 reverse signalling pathway, resulted in increased contractility of these cells. These data suggest that molecules in the EPHB/EFNB signalling pathways, specifically EFNB3 and GRIP1, are involved blood pressure regulation.
Collapse
|
28
|
Wang Y, Hamet P, Thorin E, Tremblay J, Raelson J, Wu Z, Luo H, Jin W, Lavoie JL, Peng J, Marois-Blanchet FC, Tahir MR, Chalmers J, Woodward M, Harrap S, Qi S, Li CY, Wu J. Reduced blood pressure after smooth muscle EFNB2 deletion and the potential association of EFNB2 mutation with human hypertension risk. Eur J Hum Genet 2016; 24:1817-1825. [PMID: 27530629 DOI: 10.1038/ejhg.2016.105] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 06/04/2016] [Accepted: 07/05/2016] [Indexed: 11/09/2022] Open
Abstract
Ephrin B2 (EFNB2) is a ligand for erythropoietin-producing hepatocellular kinases (EPH), the largest family of receptor tyrosine kinases. It has critical functions in many biological systems, but is not known to regulate blood pressure. We generated mice with a smooth muscle cell (SMC)-specific deletion of EFNB2 and investigated its roles in blood pressure regulation and vascular SMC (VSMC) contractility. Male Efnb2 knockout (KO) mice presented reduced blood pressure, whereas female KO mice had no such reduction. Both forward signaling from EFNB2 to EPHs and reverse signaling from EPHs to EFNB2 were involved in regulating VSMC contractility, with EPHB4 serving as a critical molecule for forward signaling, based on crosslinking studies. We also found that a region from aa 313 to aa 331 in the intracellular tail of EFNB2 was essential for reverse signaling regulating VSMC contractility, based on deletion mutation studies. In a human genetic study, we identified five SNPs in the 3' region of the EFNB2 gene, which were in linkage disequilibrium and were significantly associated with hypertension for male but not female subjects, consistent with our findings in mice. The coding (minor) alleles of these five SNPs were protective in males. We have thus discovered a previously unknown blood pressure-lowering mechanism mediated by EFNB2 and identified EFNB2 as a gene associated with hypertension risk in humans.
Collapse
Affiliation(s)
- Yujia Wang
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Pavel Hamet
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Eric Thorin
- Department of Surgery, Université de Montréal and Université Montreal Heart Institute, Montreal, QC, Canada
| | - Johanne Tremblay
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - John Raelson
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.,PGX-Services, Montreal, QC, Canada
| | - Zenghui Wu
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Hongyu Luo
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Wei Jin
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Julie L Lavoie
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.,Department of Kinesiology, University of Montreal, Montreal, QC, Canada
| | - Junzheng Peng
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | | | - Muhammad Ramzan Tahir
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - John Chalmers
- Department of Biostatistics, The George Institute for Global Health, University of Sydney, Sydney, NSW, Australia
| | - Mark Woodward
- Department of Biostatistics, The George Institute for Global Health, University of Sydney, Sydney, NSW, Australia
| | - Stephen Harrap
- Department of Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Shijie Qi
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Charles Yibin Li
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Jiangping Wu
- Research Centre, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.,Department of Nephrology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| |
Collapse
|
29
|
Functional and Proteomic Investigations Reveal Major Royal Jelly Protein 1 Associated with Anti-hypertension Activity in Mouse Vascular Smooth Muscle Cells. Sci Rep 2016; 6:30230. [PMID: 27444336 PMCID: PMC4957218 DOI: 10.1038/srep30230] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 07/01/2016] [Indexed: 12/14/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) are a major cell type of the arterial wall and their functionality is associated with blood pressure regulation. Although royal jelly (RJ) has reported effects on anti-hypertension, the mechanism of blood pressure regulation by major royal jelly protein 1 (MRJP1), the most abundant RJ protein, is still unknown. The mrjp1 gene was inserted into mouse VSMCs to investigate how MRJP1 influences VSMC functionality by functional and proteomic analysis. The expression of MRJP1 in VSMCs significantly reduced cell contraction, migration, and proliferation, suggesting a potential role in decreasing hypertension via action on VSMCs. These anti-hypertension activities were further observed in the changes of the proteome setting of mouse VSMCs. Among 675 different proteins after MRJP1 expression, 646 were down-regulated and significantly enriched in pathways implicated in VSMC contraction and migration, which suggest MRJP1 lowers VSMC contraction and migration by inhibiting muscle filament movement. The down-regulated proteins also enriched pathways in proliferation, indicating that MRJP1 hinders VSMC proliferation by reducing the supply of energy and genetic material. This is the first report integrating MRJP1 into VSMC, revealing the function and mechanism correlated with anti-hypertensive activity. This offers a therapeutic potential to control hypertension by gene-therapy using bee-products.
Collapse
|
30
|
Brozovich FV, Nicholson CJ, Degen CV, Gao YZ, Aggarwal M, Morgan KG. Mechanisms of Vascular Smooth Muscle Contraction and the Basis for Pharmacologic Treatment of Smooth Muscle Disorders. Pharmacol Rev 2016; 68:476-532. [PMID: 27037223 PMCID: PMC4819215 DOI: 10.1124/pr.115.010652] [Citation(s) in RCA: 316] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The smooth muscle cell directly drives the contraction of the vascular wall and hence regulates the size of the blood vessel lumen. We review here the current understanding of the molecular mechanisms by which agonists, therapeutics, and diseases regulate contractility of the vascular smooth muscle cell and we place this within the context of whole body function. We also discuss the implications for personalized medicine and highlight specific potential target molecules that may provide opportunities for the future development of new therapeutics to regulate vascular function.
Collapse
Affiliation(s)
- F V Brozovich
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - C J Nicholson
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - C V Degen
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - Yuan Z Gao
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - M Aggarwal
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| | - K G Morgan
- Department of Health Sciences, Boston University, Boston, Massachusetts (C.J.N., Y.Z.G., M.A., K.G.M.); Department of Medicine, Mayo Clinic, Rochester, Minnesota (F.V.B.); and Paracelsus Medical University Salzburg, Salzburg, Austria (C.V.D.)
| |
Collapse
|
31
|
Wang Y, Wu Z, Thorin E, Tremblay J, Lavoie JL, Luo H, Peng J, Qi S, Wu T, Chen F, Shen J, Hu S, Wu J. Estrogen and testosterone in concert with EFNB3 regulate vascular smooth muscle cell contractility and blood pressure. Am J Physiol Heart Circ Physiol 2016; 310:H861-72. [PMID: 26851246 DOI: 10.1152/ajpheart.00873.2015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/02/2016] [Indexed: 12/20/2022]
Abstract
EPH kinases and their ligands, ephrins (EFNs), have vital and diverse biological functions, although their function in blood pressure (BP) control has not been studied in detail. In the present study, we report that Efnb3 gene knockout (KO) led to increased BP in female but not male mice. Vascular smooth muscle cells (VSMCs) were target cells for EFNB3 function in BP regulation. The deletion of EFNB3 augmented contractility of VSMCs from female but not male KO mice, compared with their wild-type (WT) counterparts. Estrogen augmented VSMC contractility while testosterone reduced it in the absence of EFNB3, although these sex hormones had no effect on the contractility of VSMCs from WT mice. The effect of estrogen on KO VSMC contractility was via a nongenomic pathway involving GPER, while that of testosterone was likely via a genomic pathway, according to VSMC contractility assays and GPER knockdown assays. The sex hormone-dependent contraction phenotypes in KO VSMCs were reflected in BP in vivo. Ovariectomy rendered female KO mice normotensive. At the molecular level, EFNB3 KO in VSMCs resulted in reduced myosin light chain kinase phosphorylation, an event enhancing sensitivity to Ca(2+)flux in VSMCs. Our investigation has revealed previously unknown EFNB3 functions in BP regulation and show that EFNB3 might be a hypertension risk gene in certain individuals.
Collapse
Affiliation(s)
- Yujia Wang
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Zenghui Wu
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada;
| | - Eric Thorin
- Montreal Heart Institute, Montreal, Quebec, Canada
| | - Johanne Tremblay
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Julie L Lavoie
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada; Département de Kinésiologie, Université de Montréal, Montreal, Quebec, Canada
| | - Hongyu Luo
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Junzheng Peng
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Shijie Qi
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Tao Wu
- Institute of Cardiology, First Affiliated Hospital, Zhejiang University Medical College, Hangzhou, China; and
| | - Fei Chen
- Institute of Cardiology, First Affiliated Hospital, Zhejiang University Medical College, Hangzhou, China; and
| | - Jianzhong Shen
- Institute of Cardiology, First Affiliated Hospital, Zhejiang University Medical College, Hangzhou, China; and
| | - Shenjiang Hu
- Institute of Cardiology, First Affiliated Hospital, Zhejiang University Medical College, Hangzhou, China; and
| | - Jiangping Wu
- Research Centre, Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada; Nephrology Service, CRCHUM, Montreal, Quebec, Canada
| |
Collapse
|