1
|
Bai H, Li Z, Zhang W, Thaxton C, Ohashi Y, Gonzalez L, Kano M, Yatsula B, Hwa J, Dardik A. Early thrombus formation is required for eccentric and heterogeneous neointimal hyperplasia under disturbed flow. J Thromb Haemost 2024; 22:3614-3628. [PMID: 39173878 PMCID: PMC11608155 DOI: 10.1016/j.jtha.2024.07.028] [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: 05/28/2024] [Revised: 07/06/2024] [Accepted: 07/24/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Anticoagulation and antiplatelet therapy effectively inhibit neointimal hyperplasia (NIH) in both arterial and venous systems but not in arteriovenous fistulae (AVF). The main site of AVF failure is the juxta-anastomotic area that is characterized by disturbed flow compared with laminar flow in the arterial inflow and the venous outflow. OBJECTIVES We hypothesized that early thrombus formation is required for eccentric and heterogeneous NIH in the presence of disturbed flow. METHODS Needle puncture and sutured AVF were created in C57BL/6 mice, in PF4-Cre × mT/mG reporter mice, and in Wistar rats. Human AVF samples were second-stage basilic vein transpositions. The tissues were examined by histology, immunofluorescence, immunohistochemistry, and en face staining. RESULTS In the presence of disturbed flow, both mouse and human AVF showed eccentric and heterogeneous NIH. Maladapted vein wall was characterized by eccentric and heterogeneous neointima that was composed of a different abundance of thrombus and smooth muscle cells. PF4-cre × mT/mG reporter mice AVF showed that green fluorescent protein-labeled platelets deposit on the wall directly facing the fistula exit with endothelial cell loss and continue to accumulate in the presence of disturbed flow. Neither disturbed flow with limited endothelial cell loss nor nondisturbed flow induced heterogeneous neointima in different animal models. CONCLUSION Early thrombus contributes to late heterogeneous NIH in the presence of disturbed flow. Disturbed flow, large area of endothelial cell loss, and thrombus formation are critical to form eccentric and heterogeneous NIH. Categorization of adapted or maladapted walls may be helpful for therapy targeting heterogeneous NIH.
Collapse
Affiliation(s)
- Hualong Bai
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Zhuo Li
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Weichang Zhang
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Carly Thaxton
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Yuichi Ohashi
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Masaki Kano
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut, USA; Department of Surgery, Yale School of Medicine, New Haven, Connecticut, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, Connecticut, USA; Department of Surgery, Veteran Affairs Connecticut Healthcare Systems, West Haven, Connecticut, USA.
| |
Collapse
|
2
|
Yan R, Song A, Zhang C. The Pathological Mechanisms and Therapeutic Molecular Targets in Arteriovenous Fistula Dysfunction. Int J Mol Sci 2024; 25:9519. [PMID: 39273465 PMCID: PMC11395150 DOI: 10.3390/ijms25179519] [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: 08/06/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/15/2024] Open
Abstract
The number of patients with end-stage renal disease (ESRD) requiring hemodialysis is increasing worldwide. Although arteriovenous fistula (AVF) is the best and most important vascular access (VA) for hemodialysis, its primary maturation failure rate is as high as 60%, which seriously endangers the prognosis of hemodialysis patients. After AVF establishment, the venous outflow tract undergoes hemodynamic changes, which are translated into intracellular signaling pathway cascades, resulting in an outward and inward remodeling of the vessel wall. Outward remodeling refers to the thickening of the vessel wall and the dilation of the lumen to accommodate the high blood flow in the AVF, while inward remodeling is mainly characterized by intimal hyperplasia. More and more studies have shown that the two types of remodeling are closely related in the occurrence and development of, and jointly determining the final fate of, AVF. Therefore, it is essential to investigate the underlying mechanisms involved in outward and inward remodeling for identifying the key targets in alleviating AVF dysfunction. In this review, we summarize the current clinical diagnosis, monitoring, and treatment techniques for AVF dysfunction and discuss the possible pathological mechanisms related to improper outward and inward remodeling in AVF dysfunction, as well as summarize the similarities and differences between the two remodeling types in molecular mechanisms. Finally, the representative therapeutic targets of potential clinical values are summarized.
Collapse
Affiliation(s)
- Ruiwei Yan
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Anni Song
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
3
|
Dardik A. A surgeon-scientist's approach to improving arteriovenous fistula patency. JVS Vasc Sci 2024; 5:100207. [PMID: 38975292 PMCID: PMC11225657 DOI: 10.1016/j.jvssci.2024.100207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/06/2024] [Indexed: 07/09/2024] Open
Affiliation(s)
- Alan Dardik
- Vascular Biology and Therapeutics Program, Division of Vascular Surgery and Endovascular Therapy, Yale School of Medicine, New Haven, CT
- Department of Surgery, Yale School of Medicine, New Haven, CT
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT
- Department of Surgery, VA Connecticut Healthcare System, West Haven, CT
| |
Collapse
|
4
|
Wilson CA, Batzel P, Postlethwait JH. Direct male development in chromosomally ZZ zebrafish. Front Cell Dev Biol 2024; 12:1362228. [PMID: 38529407 PMCID: PMC10961373 DOI: 10.3389/fcell.2024.1362228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/20/2024] [Indexed: 03/27/2024] Open
Abstract
The genetics of sex determination varies across taxa, sometimes even within a species. Major domesticated strains of zebrafish (Danio rerio), including AB and TU, lack a strong genetic sex determining locus, but strains more recently derived from nature, like Nadia (NA), possess a ZZ male/ZW female chromosomal sex-determination system. AB fish pass through a juvenile ovary stage, forming oocytes that survive in fish that become females but die in fish that become males. To understand mechanisms of gonad development in NA zebrafish, we studied histology and single cell transcriptomics in developing ZZ and ZW fish. ZW fish developed oocytes by 22 days post-fertilization (dpf) but ZZ fish directly formed testes, avoiding a juvenile ovary phase. Gonads of some ZW and WW fish, however, developed oocytes that died as the gonad became a testis, mimicking AB fish, suggesting that the gynogenetically derived AB strain is chromosomally WW. Single-cell RNA-seq of 19dpf gonads showed similar cell types in ZZ and ZW fish, including germ cells, precursors of gonadal support cells, steroidogenic cells, interstitial/stromal cells, and immune cells, consistent with a bipotential juvenile gonad. In contrast, scRNA-seq of 30dpf gonads revealed that cells in ZZ gonads had transcriptomes characteristic of testicular Sertoli, Leydig, and germ cells while ZW gonads had granulosa cells, theca cells, and developing oocytes. Hematopoietic and vascular cells were similar in both sex genotypes. These results show that juvenile NA zebrafish initially develop a bipotential gonad; that a factor on the NA W chromosome, or fewer than two Z chromosomes, is essential to initiate oocyte development; and without the W factor, or with two Z doses, NA gonads develop directly into testes without passing through the juvenile ovary stage. Sex determination in AB and TU strains mimics NA ZW and WW zebrafish, suggesting loss of the Z chromosome during domestication. Genetic analysis of the NA strain will facilitate our understanding of the evolution of sex determination mechanisms.
Collapse
|
5
|
Wilson CA, Batzel P, Postlethwait JH. Direct Male Development in Chromosomally ZZ Zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.27.573483. [PMID: 38234788 PMCID: PMC10793451 DOI: 10.1101/2023.12.27.573483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
The genetics of sex determination varies across taxa, sometimes even within a species. Major domesticated strains of zebrafish ( Danio rerio ), including AB and TU, lack a strong genetic sex determining locus, but strains more recently derived from nature, like Nadia (NA), possess a ZZ male/ZW female chromosomal sex-determination system. AB strain fish pass through a juvenile ovary stage, forming oocytes that survive in fish that become females but die in fish that become males. To understand mechanisms of gonad development in NA zebrafish, we studied histology and single cell transcriptomics in developing ZZ and ZW fish. ZW fish developed oocytes by 22 days post-fertilization (dpf) but ZZ fish directly formed testes, avoiding a juvenile ovary phase. Gonads of some ZW and WW fish, however, developed oocytes that died as the gonad became a testis, mimicking AB fish, suggesting that the gynogenetically derived AB strain is chromosomally WW. Single-cell RNA-seq of 19dpf gonads showed similar cell types in ZZ and ZW fish, including germ cells, precursors of gonadal support cells, steroidogenic cells, interstitial/stromal cells, and immune cells, consistent with a bipotential juvenile gonad. In contrast, scRNA-seq of 30dpf gonads revealed that cells in ZZ gonads had transcriptomes characteristic of testicular Sertoli, Leydig, and germ cells while ZW gonads had granulosa cells, theca cells, and developing oocytes. Hematopoietic and vascular cells were similar in both sex genotypes. These results show that juvenile NA zebrafish initially develop a bipotential gonad; that a factor on the NA W chromosome or fewer than two Z chromosomes is essential to initiate oocyte development; and without the W factor or with two Z doses, NA gonads develop directly into testes without passing through the juvenile ovary stage. Sex determination in AB and TU strains mimics NA ZW and WW zebrafish, suggesting loss of the Z chromosome during domestication. Genetic analysis of the NA strain will facilitate our understanding of the evolution of sex determination mechanisms.
Collapse
|
6
|
Laws KM, Bashaw GJ. Diverse roles for axon guidance pathways in adult tissue architecture and function. NATURAL SCIENCES (WEINHEIM, GERMANY) 2022; 2:e20220021. [PMID: 37456985 PMCID: PMC10346896 DOI: 10.1002/ntls.20220021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Classical axon guidance ligands and their neuronal receptors were first identified due to their fundamental roles in regulating connectivity in the developing nervous system. Since their initial discovery, it has become clear that these signaling molecules play important roles in the development of a broad array of tissue and organ systems across phylogeny. In addition to these diverse developmental roles, there is a growing appreciation that guidance signaling pathways have important functions in adult organisms, including the regulation of tissue integrity and homeostasis. These roles in adult organisms include both tissue-intrinsic activities of guidance molecules, as well as systemic effects on tissue maintenance and function mediated by the nervous and vascular systems. While many of these adult functions depend on mechanisms that mirror developmental activities, such as regulating adhesion and cell motility, there are also examples of adult roles that may reflect signaling activities that are distinct from known developmental mechanisms, including the contributions of guidance signaling pathways to lineage commitment in the intestinal epithelium and bone remodeling in vertebrates. In this review, we highlight studies of guidance receptors and their ligands in adult tissues outside of the nervous system, focusing on in vivo experimental contexts. Together, these studies lay the groundwork for future investigation into the conserved and tissue-specific mechanisms of guidance receptor signaling in adult tissues.
Collapse
Affiliation(s)
- Kaitlin M. Laws
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Current address: Department of Biology, Randolph-Macon College, Ashland, VA 23005, USA
| | - Greg J. Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
7
|
Wolf KG, Crawford EB, Wartan NM, Schneiderman SK, Riehl VE, Dambaeva SV, Beaman KD. Ephrin-B2-expressing natural killer cells induce angiogenesis. JVS Vasc Sci 2022; 3:336-344. [DOI: 10.1016/j.jvssci.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/11/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
|
8
|
Sawma T, Shaito A, Najm N, Sidani M, Orekhov A, El-Yazbi AF, Iratni R, Eid AH. Role of RhoA and Rho-associated kinase in phenotypic switching of vascular smooth muscle cells: Implications for vascular function. Atherosclerosis 2022; 358:12-28. [DOI: 10.1016/j.atherosclerosis.2022.08.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/15/2022] [Accepted: 08/11/2022] [Indexed: 12/13/2022]
|
9
|
McQueen LW, Ladak SS, Zakkar M. Acute shear stress and vein graft disease. Int J Biochem Cell Biol 2022; 144:106173. [PMID: 35151879 DOI: 10.1016/j.biocel.2022.106173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/08/2021] [Accepted: 02/08/2022] [Indexed: 11/30/2022]
Abstract
The long saphenous vein is commonly used in cardiac surgery to bypass occluded coronary arteries. Its use is complicated by late stenosis and occlusion due to the development of intimal hyperplasia. It is accepted that intimal hyperplasia is a multifactorial inflammatory process that starts immediately after surgery. The role of acute changes in haemodynamic conditions when the vein is implanted into arterial circulation, especially shear stress, is not fully appreciated. This review provides an overview of intimal hyperplasia and the effect of acute shear stress changes on the activation of pro-inflammatory mediators.
Collapse
Affiliation(s)
- Liam W McQueen
- Department of Cardiovascular Sciences, Clinical Science Wing, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Shameem S Ladak
- Department of Cardiovascular Sciences, Clinical Science Wing, University of Leicester, Glenfield Hospital, Leicester, UK
| | - Mustafa Zakkar
- Department of Cardiovascular Sciences, Clinical Science Wing, University of Leicester, Glenfield Hospital, Leicester, UK.
| |
Collapse
|
10
|
Huang X, Guan J, Sheng Z, Wang M, Xu T, Guo G, Wan P, Tian B, Zhou J, Huang A, Hao J, Yao L. Effect of local anti-vascular endothelial growth factor therapy to prevent the formation of stenosis in outflow vein in arteriovenous fistula. J Transl Int Med 2021; 9:307-317. [PMID: 35136729 PMCID: PMC8802407 DOI: 10.2478/jtim-2021-0045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Vascular stenosis and angiogenesis are the major causes of short expectancy of arteriovenous fistula (AVF). Increased expression of vascular endothelial growth factor-A (VEGF-A) has been suggested to play an important role in the pathophysiologic process. Anti-VEGF has been proved to be effective on anti-angiogenesis and applied in clinical practice, but its effect on anti-stenosis remains to be verified before it could be applied to prevent stenosis of AVF. This study was aimed to evaluate the effect of local anti-VEGF therapy to prevent the formation of stenosis in the outflow vein in AVF and its mechanism. METHODS Bioinformatics of VEGF-A and its downstream-regulated molecules from the STRING PPI database were analyzed in this study. The biopsy samples from outflow veins of AVF in patients and C57BL/6 mouse models were analyzed to examine the mechanisms of pathologic vascular stenosis associated with VEGF pathways and their potential therapeutic targets. RESULTS We found that the reduction of VEGF-A could downregulate downstream molecules and subsequently reduce the intimal hyperplasia and abnormal vascular remodeling by analyzing the STRING PPI database. Venous wall thickening, intimal neointima formation, and apoptosis of vascular endothelial cells in the proliferative outflow vein of the AVF were significantly more obvious, and upregulation of expression of VEGF was observed in dysfunctional AVF in patients. In mouse models, the expression of VEGF, Ephrin receptor B4 (EphB4), matrix metalloproteinase (MMP)2, MMP9, tissue inhibitor of metalloproteinase (TIMP)1, TIMP2, and caspase 3 in the control-shRNA surgical group was significantly higher than in the sham group (P < 0.05), and all of these indicators were significantly lower in lentiviral transfection group and Avastin group than in control-shRNA surgical group (P < 0.05) on the 14th day after AVF operation. CONCLUSION VEGF expression is significantly increased in vascular endothelial cells in stenosed or occluded outflow veins of dysfunctional AVF. Local injection of Avastin into the adventitia of the proximal outflow vein in autologous AVF procedure has an excellent potential to prevent the subsequent local stenosis of the proximal outflow vein.
Collapse
Affiliation(s)
- Xin Huang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Jibin Guan
- College of Pharmacy, University of Minnesota, Minneapolis55455, MN, USA
| | - Zitong Sheng
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Menghua Wang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Tianhua Xu
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Guangying Guo
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Pengzhi Wan
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Binyao Tian
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Junlei Zhou
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Aoran Huang
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| | - Junfeng Hao
- Department of Nephrology, Jinqiu Hospital Liaoning Province, Shenyang110016, Liaoning Province, China
| | - Li Yao
- Department of Nephrology, The First Hospital of China Medical University, Shenyang110001, Liaoning Province, China
| |
Collapse
|
11
|
From remodeling to quiescence: The transformation of the vascular network. Cells Dev 2021; 168:203735. [PMID: 34425253 DOI: 10.1016/j.cdev.2021.203735] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/14/2021] [Accepted: 08/16/2021] [Indexed: 12/15/2022]
Abstract
The vascular system is essential for embryogenesis, healing, and homeostasis. Dysfunction or deregulated blood vessel function contributes to multiple diseases, including diabetic retinopathy, cancer, hypertension, or vascular malformations. A balance between the formation of new blood vessels, vascular remodeling, and vessel quiescence is fundamental for tissue growth and function. Whilst the major mechanisms contributing to the formation of new blood vessels have been well explored in recent years, vascular remodeling and quiescence remain poorly understood. In this review, we highlight the cellular and molecular mechanisms responsible for vessel remodeling and quiescence during angiogenesis. We further underline how impaired remodeling and/or destabilization of vessel networks can contribute to vascular pathologies. Finally, we speculate how addressing the molecular mechanisms of vascular remodeling and stabilization could help to treat vascular-related disorders.
Collapse
|
12
|
Chan SM, Weininger G, Langford J, Jane-Wit D, Dardik A. Sex Differences in Inflammation During Venous Remodeling of Arteriovenous Fistulae. Front Cardiovasc Med 2021; 8:715114. [PMID: 34368264 PMCID: PMC8335484 DOI: 10.3389/fcvm.2021.715114] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 06/24/2021] [Indexed: 12/18/2022] Open
Abstract
Vascular disorders frequently have differing clinical presentations among women and men. Sex differences exist in vascular access for hemodialysis; women have reduced rates of arteriovenous fistula (AVF) maturation as well as fistula utilization compared with men. Inflammation is increasingly implicated in both clinical studies and animal models as a potent mechanism driving AVF maturation, especially in vessel dilation and wall thickening, that allows venous remodeling to the fistula environment to support hemodialysis. Sex differences have long been recognized in arterial remodeling and diseases, with men having increased cardiovascular events compared with pre-menopausal women. Many of these arterial diseases are driven by inflammation that is similar to the inflammation during AVF maturation. Improved understanding of sex differences in inflammation during vascular remodeling may suggest sex-specific vascular therapies to improve AVF success.
Collapse
Affiliation(s)
- Shin Mei Chan
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States
| | - Gabe Weininger
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States
| | - John Langford
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States.,Department of Surgery, Yale School of Medicine, New Haven, CT, United States
| | - Daniel Jane-Wit
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States.,Division of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States.,Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, United States.,Department of Surgery, Yale School of Medicine, New Haven, CT, United States.,Department of Surgery, Veterans Affairs (VA) Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
13
|
Marziano C, Genet G, Hirschi KK. Vascular endothelial cell specification in health and disease. Angiogenesis 2021; 24:213-236. [PMID: 33844116 PMCID: PMC8205897 DOI: 10.1007/s10456-021-09785-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/17/2021] [Indexed: 02/08/2023]
Abstract
There are two vascular networks in mammals that coordinately function as the main supply and drainage systems of the body. The blood vasculature carries oxygen, nutrients, circulating cells, and soluble factors to and from every tissue. The lymphatic vasculature maintains interstitial fluid homeostasis, transports hematopoietic cells for immune surveillance, and absorbs fat from the gastrointestinal tract. These vascular systems consist of highly organized networks of specialized vessels including arteries, veins, capillaries, and lymphatic vessels that exhibit different structures and cellular composition enabling distinct functions. All vessels are composed of an inner layer of endothelial cells that are in direct contact with the circulating fluid; therefore, they are the first responders to circulating factors. However, endothelial cells are not homogenous; rather, they are a heterogenous population of specialized cells perfectly designed for the physiological demands of the vessel they constitute. This review provides an overview of the current knowledge of the specification of arterial, venous, capillary, and lymphatic endothelial cell identities during vascular development. We also discuss how the dysregulation of these processes can lead to vascular malformations, and therapeutic approaches that have been developed for their treatment.
Collapse
Affiliation(s)
- Corina Marziano
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.,Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Gael Genet
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA.,Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA
| | - Karen K Hirschi
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA. .,Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, 22908, USA. .,Department of Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT, 06520, USA.
| |
Collapse
|
14
|
Guo Y, Zhu F, Zhang X, Wu G, Fu P, Yang J. Extracellular signal-regulated kinase inhibition prevents venous adaptive remodeling via regulation of Eph-B4. Vascular 2021; 30:120-129. [PMID: 33706642 DOI: 10.1177/1708538121999854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVES Vein graft adaptation (VGA) is a process that vein as a vascular graft conduits in arterial reconstructive surgery; VGA can lead to postoperative vein graft stenosis (VGS) and complications after coronary artery bypass graft and other peripheral artery bypass surgeries. VGA is characterized by vein graft loss the venous features without exhibiting arterial features; furthermore, the activation of ERK inhibited the maintenance of venous properties of the vein graft. We hypothesized that ERK inhibition can affect vein VGS through regulating the expression of EphB4. METHODS Rat vein transplantation model was established using wild-type and EphB4+/- Sprague-Dawley rats. Hematoxylin-eosin, Masson, Verhoeff, actin staining, and immunohistochemistry were applied to observe the structure of the vein grafts. Vascular smooth muscle cells (VSMCs) were isolated from the vein and vein grafts. Western blotting was used to determine the expression of p-ERK1/2 and EphB4, and immunofluorescence was applied to detect the expression and location of EphB4. Cell wound scratch assay and CCK8 assay were used to determine the migration and proliferation of VSMCs. Real-time polymerase chain reaction was used to determine the mRNA expression of EphB4. RESULTS Western blotting in vein sample and vein graft sample detected p-ERK1/2 and ERK1/2 expression in both EphB4+/+ and EphB4+/- rats. The expression of p-ERK was increased in vein graft compared to vein. Immunofluorescence in VSMCs form EphB4+/+ and EphB4+/- rats detected EphB4 expression in both cells, and the expression of EphB4 was increased in VSMCs form EphB4+/+ rats. SCH772984 reduces the proliferation and migration of VSMCs. Inhibition of ERK suppressed the increase of vein graft wall thickness, and the expression of collagen fibers, elastic fibers, and α-actin was decreased. Vein graft from EphB4+/- rats reduces the expression of EphB4, and SCH772984 suppressed the decrease of EphB4 in vivo. Vein graft from EphB4+/- rats increased the expression of EphB4, and SCH772984 suppressed the increase of EphB4 in vivo. CONCLUSIONS The inhibition of ERK1/2 suppressed the process of VGS by decreasing the proliferation of VSMCs. The ERK-inhibitor SCH772984 suppressed the level of VGS by extending the time of EphB4 expression during the process of VGA, thus maintaining the venousization of vein graft. The mechanism may be that the inhibitor SCH772984 suppresses the level of VGS by extending the time of EphB4 expression during the process of VGA. Therefore, our research provides a new target of VGS treatment by inhibiting the expression of ERK1/2 through the process of VGA.
Collapse
Affiliation(s)
- Yuanyuan Guo
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.,Department of Vascular Surgery, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Fan Zhu
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.,Department of Vascular Surgery, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Xiong Zhang
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.,Department of Vascular Surgery, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Guangmin Wu
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.,Department of Vascular Surgery, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Pinting Fu
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.,Department of Vascular Surgery, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| | - Jun Yang
- Department of Vascular Surgery, Fuwai Yunnan Cardiovascular Hospital, Kunming, China.,Department of Vascular Surgery, Affiliated Cardiovascular Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
15
|
Cai Q, Liao W, Xue F, Wang X, Zhou W, Li Y, Zeng W. Selection of different endothelialization modes and different seed cells for tissue-engineered vascular graft. Bioact Mater 2021; 6:2557-2568. [PMID: 33665496 PMCID: PMC7887299 DOI: 10.1016/j.bioactmat.2020.12.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/09/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Tissue-engineered vascular grafts (TEVGs) have enormous potential for vascular replacement therapy. However, thrombosis and intimal hyperplasia are important problems associated with TEVGs especially small diameter TEVGs (<6 mm) after transplantation. Endothelialization of TEVGs is a key point to prevent thrombosis. Here, we discuss different types of endothelialization and different seed cells of tissue-engineered vascular grafts. Meanwhile, endothelial heterogeneity is also discussed. Based on it, we provide a new perspective for selecting suitable types of endothelialization and suitable seed cells to improve the long-term patency rate of tissue-engineered vascular grafts with different diameters and lengths. The material, diameter and length of tissue-engineered vascular graft are all key factors affecting its long-term patency. Endothelialization strategies should consider the different diameters and lengths of tissue-engineered vascular grafts. Cell heterogeneity and tissue heterogeneity should be considered in the application of seed cells.
Collapse
Affiliation(s)
- Qingjin Cai
- Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China
| | - Wanshan Liao
- Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China
| | - Fangchao Xue
- Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China
| | - Xiaochen Wang
- Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China
| | - Weiming Zhou
- Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China
| | - Yanzhao Li
- State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing, China
| | - Wen Zeng
- Department of Cell Biology, Third Military Medical University, Chongqing, 400038, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing, China.,Departments of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
16
|
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
|
17
|
Liu F, Yao Z, Lü P, Jiao QB, Liu Q, Wu HX, You Y, Minamisawa S. Pathophysiologic Role of Molecules Determining Arteriovenous Differentiation in Adult Life. J Vasc Res 2020; 57:245-253. [PMID: 32535603 DOI: 10.1159/000507627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 03/26/2020] [Indexed: 11/19/2022] Open
Abstract
The structural differences between arteries and veins are genetically predetermined. Vascular identity markers, the molecular markers specific to veins and arteries, determine the differential development of vessels during embryogenesis and their expression persists in adult vessels. It is revealed that they can be reactivated under various pathophysiologic conditions even after vessel differentiation. Thus, once considered as quiescent in adults, vascular identity markers may actually play significant roles in vascular remodeling. Manipulation of vascular identity and the underlying molecular mechanisms might be a novel strategy to improve vascular remodeling for clinical application.
Collapse
Affiliation(s)
- Fang Liu
- Nuclear Medicine Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhong Yao
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Ping Lü
- Vascular Surgery Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,
| | - Qi-Bin Jiao
- Department of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Institute of Ageing Research, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Qin Liu
- Vascular Surgery Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong-Xiao Wu
- Vascular Surgery Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun You
- Vascular Surgery Department, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Susumu Minamisawa
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
18
|
Bai H, Wang Z, Li M, Sun P, Wei S, Wang Z, Xing Y, Dardik A. Adult Human Vein Grafts Retain Plasticity of Vessel Identity. Ann Vasc Surg 2020; 68:468-475. [PMID: 32422286 DOI: 10.1016/j.avsg.2020.04.046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/14/2020] [Accepted: 04/18/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND The spiral saphenous vein graft is an excellent choice for venous reconstruction after periphery vein injury, but only few cases have been reported. We implanted a segment of a single saphenous vein into both the popliteal vein as a venous vein graft and into the popliteal artery as an arterial vein graft at the same time in a trauma patient; we then had an extraordinary opportunity to harvest and examine both patent venous and arterial vein grafts at 2 weeks after implantation. METHODS A spiral saphenous vein graft was made as previously described and implanted into the popliteal vein and artery as interposition grafts; because of the patient's serious injuries, an amputation was performed at day 18 after vascular reconstruction. The grafts were harvested, fixed, and examined using histology and immunohistochemistry. RESULTS Both grafts were patent, and there was a larger neointimal area in the venous graft compared to the arterial graft. There were CD31- and vWF-positive cells on both neointimal endothelia, with subendothelial deposition of α-actin-, CD3-, CD45-, and CD68-positive cells. There were fewer cells in the venous graft neointima compared to the arterial graft neointima; however, there were more inflammatory cells in the neointima of the venous graft. Some of the neointimal cells were PCNA-positive, whereas very few cells were cleaved caspase-3 positive. The venous graft neointimal endothelial cells were Eph-B4 and COUP-TFII positive, while the arterial graft neointimal endothelial cells were dll-4 and Ephrin-B2 positive. CONCLUSIONS The spiral saphenous vein graft remains a reasonable choice for vessel reconstruction, especially in the presence of diameter mismatch. Both the venous and arterial grafts showed similar re-endothelialization and cellular deposition; the venous graft had more neointimal hyperplasia and inflammation. At an early time, endothelial cells showed venous identity in the venous graft, whereas endothelial cells showed arterial identity in the arterial graft. CLINICAL RELEVANCE Veins can be used as venous or arterial vein grafts but venous grafts have more neointimal hyperplasia and inflammation; vein grafts acquire different vessel identity depending on the environment into which they are implanted.
Collapse
Affiliation(s)
- Hualong Bai
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China; Department of Physiology, Medical School of Zhengzhou University, Zhengzhou, Henan, People's Republic of China.
| | - Zhiwei Wang
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Mingxing Li
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Peng Sun
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Shunbo Wei
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Zhiju Wang
- Key Vascular Physiology and Applied Research Laboratory, Zhengzhou, Henan, People's Republic of China; Department of Physiology, Medical School of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Ying Xing
- Key Vascular Physiology and Applied Research Laboratory, Zhengzhou, Henan, People's Republic of China; Department of Physiology, Medical School of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Alan Dardik
- The Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT; Department of Surgery and Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
19
|
Hashimoto T, Isaji T, Hu H, Yamamoto K, Bai H, Santana JM, Kuo A, Kuwahara G, Foster TR, Hanisch JJ, Yatsula BA, Sessa WC, Hoshina K, Dardik A. Stimulation of Caveolin-1 Signaling Improves Arteriovenous Fistula Patency. Arterioscler Thromb Vasc Biol 2020; 39:754-764. [PMID: 30786746 DOI: 10.1161/atvbaha.119.312417] [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] [Indexed: 12/14/2022]
Abstract
Objective- Arteriovenous fistulae (AVF) are the most common access created for hemodialysis; however, many AVF fail to mature and require repeated intervention, suggesting a need to improve AVF maturation. Eph-B4 (ephrin type-B receptor 4) is the embryonic venous determinant that is functional in adult veins and can regulate AVF maturation. Cav-1 (caveolin-1) is the major scaffolding protein of caveolae-a distinct microdomain that serves as a mechanosensor at the endothelial cell membrane. We hypothesized that Cav-1 function is critical for Eph-B4-mediated AVF maturation. Approach and Results- In a mouse aortocaval fistula model, both Cav-1 mRNA and protein were increased in the AVF compared with control veins. Cav-1 KO (knockout) mice showed increased fistula wall thickening ( P=0.0005) and outward remodeling ( P<0.0001), with increased eNOS (endothelial NO synthase) activity compared with WT (wild type) mice. Ephrin-B2/Fc inhibited AVF outward remodeling in WT mice but not in Cav-1 KO mice and was maintained in Cav-1 RC (Cav-1 endothelial reconstituted) mice (WT, P=0.0001; Cav-1 KO, P=0.7552; Cav-1 RC, P=0.0002). Cavtratin-a Cav-1 scaffolding domain peptide-decreased AVF wall thickness in WT mice and in Eph-B4 het mice compared with vehicle alone (WT, P=0.0235; Eph-B4 het, P=0.0431); cavtratin also increased AVF patency (day 42) in WT mice ( P=0.0275). Conclusions- Endothelial Cav-1 mediates Eph-B4-mediated AVF maturation. The Eph-B4-Cav-1 axis regulates adaptive remodeling during venous adaptation to the fistula environment. Manipulation of Cav-1 function may be a translational strategy to enhance AVF patency.
Collapse
Affiliation(s)
- Takuya Hashimoto
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery (T.H., T.I., H.H., K.Y., H.B., J.M.S., G.K., T.R.F., J.J.H., B.A.Y., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare System, West Haven (T.H., T.I., H.H., K.Y., H.B., T.R.F., A.D.).,Department of Vascular Surgery, University of Tokyo, Japan (T.H., T.I., K.Y., K.H.)
| | - Toshihiko Isaji
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery (T.H., T.I., H.H., K.Y., H.B., J.M.S., G.K., T.R.F., J.J.H., B.A.Y., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare System, West Haven (T.H., T.I., H.H., K.Y., H.B., T.R.F., A.D.).,Department of Vascular Surgery, University of Tokyo, Japan (T.H., T.I., K.Y., K.H.)
| | - Haidi Hu
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery (T.H., T.I., H.H., K.Y., H.B., J.M.S., G.K., T.R.F., J.J.H., B.A.Y., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare System, West Haven (T.H., T.I., H.H., K.Y., H.B., T.R.F., A.D.)
| | - Kota Yamamoto
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery (T.H., T.I., H.H., K.Y., H.B., J.M.S., G.K., T.R.F., J.J.H., B.A.Y., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare System, West Haven (T.H., T.I., H.H., K.Y., H.B., T.R.F., A.D.).,Department of Vascular Surgery, University of Tokyo, Japan (T.H., T.I., K.Y., K.H.)
| | - Hualong Bai
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery (T.H., T.I., H.H., K.Y., H.B., J.M.S., G.K., T.R.F., J.J.H., B.A.Y., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare System, West Haven (T.H., T.I., H.H., K.Y., H.B., T.R.F., A.D.)
| | - Jeans M Santana
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery (T.H., T.I., H.H., K.Y., H.B., J.M.S., G.K., T.R.F., J.J.H., B.A.Y., A.D.), Yale School of Medicine, New Haven, CT
| | - Andrew Kuo
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Pharmacology (A.K., W.C.S.), Yale School of Medicine, New Haven, CT
| | - Go Kuwahara
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery (T.H., T.I., H.H., K.Y., H.B., J.M.S., G.K., T.R.F., J.J.H., B.A.Y., A.D.), Yale School of Medicine, New Haven, CT
| | - Trenton R Foster
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery (T.H., T.I., H.H., K.Y., H.B., J.M.S., G.K., T.R.F., J.J.H., B.A.Y., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare System, West Haven (T.H., T.I., H.H., K.Y., H.B., T.R.F., A.D.)
| | - Jesse J Hanisch
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery (T.H., T.I., H.H., K.Y., H.B., J.M.S., G.K., T.R.F., J.J.H., B.A.Y., A.D.), Yale School of Medicine, New Haven, CT
| | - Bogdan A Yatsula
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery (T.H., T.I., H.H., K.Y., H.B., J.M.S., G.K., T.R.F., J.J.H., B.A.Y., A.D.), Yale School of Medicine, New Haven, CT
| | - William C Sessa
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Pharmacology (A.K., W.C.S.), Yale School of Medicine, New Haven, CT
| | - Katsuyuki Hoshina
- Department of Vascular Surgery, University of Tokyo, Japan (T.H., T.I., K.Y., K.H.)
| | - Alan Dardik
- From the Vascular Biology and Therapeutics Program (T.H., T.I., H.H., K.Y., H.B., J.M.S., A.K., G.K., T.R.F., J.J.H., B.A.Y., W.C.S., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery (T.H., T.I., H.H., K.Y., H.B., J.M.S., G.K., T.R.F., J.J.H., B.A.Y., A.D.), Yale School of Medicine, New Haven, CT.,Department of Surgery, VA Connecticut Healthcare System, West Haven (T.H., T.I., H.H., K.Y., H.B., T.R.F., A.D.)
| |
Collapse
|
20
|
Topography elicits distinct phenotypes and functions in human primary and stem cell derived endothelial cells. Biomaterials 2020; 234:119747. [PMID: 31951971 DOI: 10.1016/j.biomaterials.2019.119747] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 11/25/2019] [Accepted: 12/25/2019] [Indexed: 12/20/2022]
Abstract
The effective deployment of arterial (AECs), venous (VECs) and stem cell-derived endothelial cells (PSC-ECs) in clinical applications requires understanding of their distinctive phenotypic and functional characteristics, including their responses to microenvironmental cues. Efforts to mimic the in-vivo vascular basement membrane milieu have led to the design and fabrication of nano- and micro-topographical substrates. Although the basement membrane architectures of arteries and veins are different, investigations into the effects of substrate topographies have so far focused on generic EC characteristics. Thus, topographical modulation of arterial- or venous-specific EC phenotype and function remains unknown. Here, we comprehensively evaluated the effects of 16 unique topographies on primary AECs, VECs and human PSC-ECs using a Multi Architectural (MARC) Chip. Gratings and micro-lenses augmented venous-specific phenotypes and depressed arterial functions in VECs; while AECs did not respond consistently to topography. PSC-ECs exhibited phenotypic and functional maturation towards an arterial subtype with increased angiogenic potential, NOTCH1 and Ac-LDL expression on gratings. Specific topographies could elicit different phenotypic and functional changes, despite similar morphological response in different ECs, demonstrating no direct correlation between the two responses.
Collapse
|
21
|
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: 43] [Impact Index Per Article: 7.2] [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
|
22
|
Hernandez DR, Rojas MG, Martinez L, Rodriguez BL, Zigmond ZM, Vazquez-Padron RI, Lassance-Soares RM. c-Kit deficiency impairs nitric oxide signaling in smooth muscle cells. Biochem Biophys Res Commun 2019; 518:227-232. [PMID: 31416613 PMCID: PMC6732002 DOI: 10.1016/j.bbrc.2019.08.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 02/09/2023]
Abstract
INTRODUCTION Receptor tyrosine kinases have been implicated in various vascular remodeling processes and cardiovascular disease. However, their role in the regulation of vascular tone is poorly understood. Herein, we evaluate the contribution of c-Kit signaling to vasoactive responses. METHODS The vascular reactivity of mesenteric arteries was assessed under isobaric conditions in c-Kit deficient (KitW/W-v) and littermate control mice (Kit+/+) using pressure myography. Protein levels of soluble guanylyl cyclase beta 1 (sGCβ1) were quantified by Western blot. Mean arterial pressure was measured after high salt (8% NaCl) diet treatment using the tail-cuff method. RESULTS Smooth muscle cells (SMCs) from c-Kit deficient mice showed a 5-fold downregulation of sGCβ1 compared to controls. Endothelium-dependent relaxation of mesenteric arteries demonstrated a predominance of prostanoid vs. nitric oxide (NO) signaling in both animal groups. The dependence on prostanoid-induced dilation was higher in c-Kit mutant mice than in controls, as indicated by a significant impairment in vasorelaxation with indomethacin with respect to the latter. Endothelium-independent relaxation showed significant dysfunction of NO signaling in c-Kit deficient SMCs compared to controls. Mesenteric artery dilation was rescued by addition of a cGMP analog, but not with a NO donor, indicating a deficiency in cGMP production in c-Kit deficient SMCs. Finally, c-Kit deficient mice developed higher blood pressure on an 8% NaCl diet compared to their control littermates. CONCLUSION c-Kit deficiency inhibits NO signaling in SMCs. The existence of this c-Kit/sGC signaling axis may be relevant for vascular reactivity and remodeling.
Collapse
Affiliation(s)
- Diana R Hernandez
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Miguel G Rojas
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Laisel Martinez
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Boris L Rodriguez
- Department of Surgery, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | - Zachary M Zigmond
- Department of Molecular and Cellular Pharmacology, University of Miami, Miller School of Medicine, Miami, FL, 33136, USA
| | | | | |
Collapse
|
23
|
Gorecka J, Fereydooni A, Gonzalez L, Lee SR, Liu S, Ono S, Xu J, Liu J, Taniguchi R, Matsubara Y, Gao X, Gao M, Langford J, Yatsula B, Dardik A. Molecular Targets for Improving Arteriovenous Fistula Maturation and Patency. VASCULAR INVESTIGATION AND THERAPY 2019; 2:33-41. [PMID: 31608322 DOI: 10.4103/vit.vit_9_19] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The increasing prevalence of chronic and end-stage renal disease creates an increased need for reliable vascular access, and although arteriovenous fistulae (AVF) are the preferred mode of hemodialysis access, 60% fail to mature and only 50% remain patent at one year. Fistulae mature by diameter expansion and wall thickening; this outward remodeling of the venous wall in the fistula environment relies on a delicate balance of extracellular matrix (ECM) remodeling, inflammation, growth factor secretion, and cell adhesion molecule upregulation in the venous wall. AVF failure occurs via two distinct mechanisms with early failure secondary to lack of outward remodeling, that is insufficient diameter expansion or wall thickening, whereas late failure occurs with excessive wall thickening due to neointimal hyperplasia (NIH) and insufficient diameter expansion in a previously functional fistula. In recent years, the molecular basis of AVF maturation and failure are becoming understood in order to develop potential therapeutic targets to aide maturation and prevent access loss. Erythropoietin-producing hepatocellular carcinoma (Eph) receptors, along with their ligands, ephrins, determine vascular identity and are critical for vascular remodeling in the embryo. Manipulation of Eph receptor signaling in adults, as well as downstream pathways, is a potential treatment strategy to improve the rates of AVF maturation and patency. This review examines our current understanding of molecular changes occurring following fistula creation, factors predictive of fistula success, and potential areas of intervention to decrease AVF failure.
Collapse
Affiliation(s)
- Jolanta Gorecka
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Arash Fereydooni
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Luis Gonzalez
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Shin Rong Lee
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Shirley Liu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Shun Ono
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Jianbiao Xu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Jia Liu
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA.,The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Vascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ryosuke Taniguchi
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Yutaka Matsubara
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Xixiang Gao
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA.,Department of Vascular Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Mingjie Gao
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA.,Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - John Langford
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Bogdan Yatsula
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, Yale University, New Haven, USA.,Section of Vascular and Endovascular Surgery, VA Connecticut Healthcare System, West Haven, USA
| |
Collapse
|
24
|
Arora S, Yim EKF, Toh YC. Environmental Specification of Pluripotent Stem Cell Derived Endothelial Cells Toward Arterial and Venous Subtypes. Front Bioeng Biotechnol 2019; 7:143. [PMID: 31259171 PMCID: PMC6587665 DOI: 10.3389/fbioe.2019.00143] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/28/2019] [Indexed: 12/25/2022] Open
Abstract
Endothelial cells (ECs) are required for a multitude of cardiovascular clinical applications, such as revascularization of ischemic tissues or endothelialization of tissue engineered grafts. Patient derived primary ECs are limited in number, have donor variabilities and their in vitro phenotypes and functions can deteriorate over time. This necessitates the exploration of alternative EC sources. Although there has been a recent surge in the use of pluripotent stem cell derived endothelial cells (PSC-ECs) for various cardiovascular clinical applications, current differentiation protocols yield a heterogeneous EC population, where their specification into arterial or venous subtypes is undefined. Since arterial and venous ECs are phenotypically and functionally different, inappropriate matching of exogenous ECs to host sites can potentially affect clinical efficacy, as exemplified by venous graft mismatch when placed into an arterial environment. Therefore, there is a need to design and employ environmental cues that can effectively modulate PSC-ECs into a more homogeneous arterial or venous phenotype for better adaptation to the host environment, which will in turn contribute to better application efficacy. In this review, we will first give an overview of the developmental and functional differences between arterial and venous ECs. This provides the foundation for our subsequent discussion on the different bioengineering strategies that have been investigated to varying extent in providing biochemical and biophysical environmental cues to mature PSC-ECs into arterial or venous subtypes. The ability to efficiently leverage on a combination of biochemical and biophysical environmental cues to modulate intrinsic arterio-venous specification programs in ECs will greatly facilitate future translational applications of PSC-ECs. Since the development and maintenance of arterial and venous ECs in vivo occur in disparate physio-chemical microenvironments, it is conceivable that the application of these environmental factors in customized combinations or magnitudes can be used to selectively mature PSC-ECs into an arterial or venous subtype.
Collapse
Affiliation(s)
- Seep Arora
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, Singapore
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, Singapore.,Biomedical Institute for Global Health Research and Technology (BIGHEART), National University of Singapore, Singapore, Singapore.,NUS Tissue Engineering Program, National University of Singapore, Singapore, Singapore
| |
Collapse
|
25
|
Wolf K, Hu H, Isaji T, Dardik A. Molecular identity of arteries, veins, and lymphatics. J Vasc Surg 2019; 69:253-262. [PMID: 30154011 PMCID: PMC6309638 DOI: 10.1016/j.jvs.2018.06.195] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 06/25/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Arteries, veins, and lymphatic vessels are distinguished by structural differences that correspond to their different functions. Each of these vessels is also defined by specific molecular markers that persist throughout adult life; these markers are some of the molecular determinants that control the differentiation of embryonic undifferentiated cells into arteries, veins, or lymphatics. METHODS This is a review of experimental literature. RESULTS The Eph-B4 receptor and its ligand, ephrin-B2, are critical molecular determinants of vessel identity, arising on endothelial cells early in embryonic development. Eph-B4 and ephrin-B2 continue to be expressed on adult vessels and mark vessel identity. However, after vascular surgery, vessel identity can change and is marked by altered Eph-B4 and ephrin-B2 expression. Vein grafts show loss of venous identity, with less Eph-B4 expression. Arteriovenous fistulas show gain of dual arterial-venous identity, with both Eph-B4 and ephrin-B2 expression, and manipulation of Eph-B4 improves arteriovenous fistula patency. Patches used to close arteries and veins exhibit context-dependent gain of identity, that is, patches in the arterial environment gain arterial identity, whereas patches in the venous environment gain venous identity; these results show the importance of the host infiltrating cells in determining vascular identity after vascular surgery. CONCLUSIONS Changes in the vessel's molecular identity after vascular surgery correspond to structural changes that depend on the host's postsurgical environment. Regulation of vascular identity and the underlying molecular mechanisms may allow new therapeutic approaches to improve vascular surgical procedures.
Collapse
Affiliation(s)
- Katharine Wolf
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Haidi Hu
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn; VA Connecticut Healthcare System, West Haven, Conn.
| |
Collapse
|
26
|
Dorsey TB, Kim D, Grath A, James D, Dai G. Multivalent biomaterial platform to control the distinct arterial venous differentiation of pluripotent stem cells. Biomaterials 2018; 185:1-12. [PMID: 30216805 DOI: 10.1016/j.biomaterials.2018.09.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/30/2018] [Accepted: 09/02/2018] [Indexed: 11/25/2022]
Abstract
Vascular endothelial cells (ECs) differentiated from pluripotent stem cells have enormous potential to be used in a variety of therapeutic areas such as tissue engineering of vascular grafts and re-vascularization of ischemic tissues. To date, various protocols have been developed to differentiate stem cells toward vascular ECs. However, current methods are still not sufficient to drive the distinct arterial venous differentiation. Therefore, developing refined method of arterial-venous differentiation is critically needed to address this gap. Here, we developed a biomaterial platform to mimic multivalent ephrin-B2/EphB4 signaling and investigated its role in the early arterial and venous specification of pluripotent stem cells. Our results show immobilized ephrinB2 or EphB4 on hydrogel substrates have a distinct effect on arterial venous differentiation by regulating several arterial venous markers. When in combination with Wnt pathway agonist or BMP4 signaling, the ephrin-B2/EphB4 biomaterial platform can create diverging EC progenitor populations, demonstrating differential gene expression pattern across a wide range of arterial and venous markers, as well as phenotypic markers such as anti-thrombotic, pro-atherogenic and osteogenic genes, that are consistent with the in vivo expression patterns of arterial and venous ECs. Importantly, this distinct EC progenitor population cannot be achieved by current methods of applying soluble factors or hemodynamic stimuli alone, illustrating that fine-tuning of developmental signals using the biomaterial platform offers a new approach to better control the arterial venous differentiation of stem cells.
Collapse
Affiliation(s)
- Taylor B Dorsey
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY 12180, United States; Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 1623 15th, St, Troy, NY 12180, United States; Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
| | - Diana Kim
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY 12180, United States; Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 1623 15th, St, Troy, NY 12180, United States; Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
| | - Alexander Grath
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY 12180, United States; Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 1623 15th, St, Troy, NY 12180, United States; Department of Bioengineering, Northeastern University, Boston, MA 02115, United States
| | - Daylon James
- Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY, 10065, United States
| | - Guohao Dai
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th St., Troy, NY 12180, United States; Rensselaer Polytechnic Institute, Center for Biotechnology and Interdisciplinary Studies, 1623 15th, St, Troy, NY 12180, United States; Department of Bioengineering, Northeastern University, Boston, MA 02115, United States.
| |
Collapse
|
27
|
Abstract
The development processes of arteries and veins are fundamentally different, leading to distinct differences in anatomy, structure, and function as well as molecular profiles. Understanding the complex interaction between genetic and epigenetic pathways, as well as extracellular and biomechanical signals that orchestrate arterial venous differentiation, is not only critical for the understanding of vascular diseases of arteries and veins but also valuable for vascular tissue engineering strategies. Recent research has suggested that certain transcriptional factors not only control arterial venous differentiation during development but also play a critical role in adult vessel function and disease processes. This review summarizes the signaling pathways and critical transcription factors that are important for arterial versus venous specification. We focus on those signals that have a direct relation to the structure and function of arteries and veins, and have implications for vascular disease processes and tissue engineering applications.
Collapse
Affiliation(s)
- Laura Niklason
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, Connecticut 06520, USA.,Departments of Anesthesiology and Biomedical Engineering, Yale University, New Haven, Connecticut 06519, USA
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, Massachusetts 02115, USA;
| |
Collapse
|
28
|
Podemska-Jedrzejczak Z, Malinska A, Sujka-Kordowska P, Nowicki M, Puslecki M, Jemielity M, Perek B. Vascular restenosis in coronary artery bypass grafting might be associated with VEGF-C/VEGFR-3 signaling pathway. Heart Vessels 2018; 33:1106-1120. [PMID: 29557990 PMCID: PMC6096743 DOI: 10.1007/s00380-018-1158-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 03/16/2018] [Indexed: 11/29/2022]
Abstract
The vascular endothelial growth factor (VEGF) family of peptides and caveolins (CAVs) are reported to contribute, in early graft failure in patients, a coronary artery bypass grafting (CABG). To investigate the possible association of ultimate luminal occlusion to VEGFs and CAVs expression, a functional analysis (based on the molecular biology, bioinformatics, histology, and clinical studies) was performed. Twenty-four hundred and sixty-eight CABG patients diagnosed with multivessel stable coronary artery disease (CAD) were enrolled into prospective study and assigned to two subgroups: double- and triple-vessel CAD subjects. Distal parts of all the harvested saphenous vein (SV) and internal thoracic artery (ITA) segments were used for further tests. ITA graft failure did not differ between double-vessel and triple-vessel CAD patients. The number of SV occlusions was significantly higher in triple-vessel CAD subjects. The microarray analysis performed on SV and ITA samples obtained exclusively from triple-vessel CAD patients who developed early graft occlusion revealed 383 genes with increased and 301 genes with decreased expression in ITA samples as compared to SV grafts. This was followed by functional analysis of ‘blood vessel development’ group of genes. Average VEGF-C expression in ITA grafts was higher than in corresponding SV grafts; FLT4 expression was significantly higher in SV than in ITA transplants. VEGFR-3 and CAV3 expression demonstrated immunohistochemically in SMCs of the tunica media of SV grafts predicted their early restenosis in triple-vessel CAD patients. CAV2 protein expression in SMCs of ITA grafts indicated the risk of early graft failure both in double-vessel and triple-vessel CAD subjects.
Collapse
Affiliation(s)
- Zuzanna Podemska-Jedrzejczak
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecickiego 6 St, 60-781, Poznan, Poland
| | - Agnieszka Malinska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecickiego 6 St, 60-781, Poznan, Poland.,Department of Anatomy and Histology, University of Zielona Gora, Zyty 28 St, 65-046, Zielona Gora, Poland
| | - Patrycja Sujka-Kordowska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecickiego 6 St, 60-781, Poznan, Poland. .,Department of Anatomy and Histology, University of Zielona Gora, Zyty 28 St, 65-046, Zielona Gora, Poland.
| | - Michal Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Swiecickiego 6 St, 60-781, Poznan, Poland
| | - Mateusz Puslecki
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, Dluga 1/2 St, 60-101, Poznan, Poland
| | - Marek Jemielity
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, Dluga 1/2 St, 60-101, Poznan, Poland
| | - Bartlomiej Perek
- Department of Cardiac Surgery and Transplantology, Poznan University of Medical Sciences, Dluga 1/2 St, 60-101, Poznan, Poland
| |
Collapse
|
29
|
Lü P, Jiao Q, Shimura D, Kusakari Y, Liu F, Minamisawa S. Distinct Vascular Remodeling Pattern of Adult Rats with Carotid-Jugular Shunt. Ann Vasc Surg 2018; 49:168-178. [PMID: 29501904 DOI: 10.1016/j.avsg.2017.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/26/2017] [Accepted: 12/17/2017] [Indexed: 11/18/2022]
Abstract
BACKGROUND Previous research has revealed that patent vein grafts lose their venous identity Eph-B4 but do not gain arterial identity ephrin-B2 during adaptation to the arterial circulation, and vascular identity marker, for example, the Eph-B4 signaling is a critical determinant of venous wall thickness of vein grafts. But what is the remodeling pattern, especially the remodeling pattern of vascular identity in the venous segment of arteriovenous shunt at a late stage postoperation has not been fully explored. This study was conducted to characterize the remodeling pattern of shear stress, vascular identity, structural composition and morphology, and transcriptional profiles in jugular segment of carotid-jugular (CJ) shunt and/or pulmonary artery (PA), which delivers an increased amount of mixed blood at a late stage postoperation in adult rats. METHODS CJ shunt was created in adult Wistar rats via end-to-end anastomosis of carotid artery (CA) and jugular vein (JV). At the time of 15 weeks, after hemodynamics test, remodeled jugular segment of CJ shunt, PA, and sham-operated corresponding vessels were isolated. Reverse transcription polymerase chain reaction, microarray, western blot, immunohistochemistry experiments, and morphology analyses were performed. RESULTS CJ shunt shear stresses have been patterned to some sort of balance with no significant difference in shear stress between carotid segment and jugular segment (P > 0.05). Immunohistochemical analysis reveals that venous identity marker Eph-B4 is lost, but arterial identity markers ephrin-B2 and regulator of G-protein signaling 5 are gained in jugular segment of CJ shunt (P < 0.01), and these 2 arterial identity markers further strengthened in PA (P < 0.01) in shunted rats compared with controls. Jugular segment of CJ shunt undergoes significant intimal hyperplasia with strong expression of smooth muscle cell markers (P < 0.05) and demonstrates a distinct transcriptional profiles which reveals that transcripts of 5 arterial markers are significantly upregulated (P < 0.05 or < 0.01) compared with sham-operated JV; among them, G-protein signaling 5 is exactly the gene with the largest fold change (10.14-fold) in all genes tested by microarray experiment. CONCLUSIONS Venous identity is lost, but arterial identity is gained in jugular segment of CJ shunt and arterial identity further strengthened in PA in adult shunted rats during late adaptation.
Collapse
MESH Headings
- Anastomosis, Surgical
- Animals
- Biopsy
- Blotting, Western
- Carotid Arteries/metabolism
- Carotid Arteries/pathology
- Carotid Arteries/physiopathology
- Carotid Arteries/surgery
- Gene Expression Profiling/methods
- Gene Expression Regulation
- Hemodynamics
- Immunohistochemistry
- Jugular Veins/metabolism
- Jugular Veins/pathology
- Jugular Veins/physiopathology
- Jugular Veins/surgery
- Male
- Models, Animal
- Oligonucleotide Array Sequence Analysis
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Pulmonary Artery/surgery
- RGS Proteins/genetics
- RGS Proteins/metabolism
- Rats, Wistar
- Real-Time Polymerase Chain Reaction
- Receptor, EphB2/genetics
- Receptor, EphB2/metabolism
- Receptor, EphB4/genetics
- Receptor, EphB4/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Stress, Mechanical
- Time Factors
- Transcriptome
- Ultrasonography, Doppler, Color
- Vascular Remodeling
Collapse
Affiliation(s)
- Ping Lü
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qibin Jiao
- Department of Cardiology, The Affiliated Hospital of Hangzhou Normal University, Institute of Ageing Research, School of Medicine, Hangzhou Normal University, Hangzhou, China
| | - Daisuke Shimura
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoichiro Kusakari
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| | - Fang Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Susumu Minamisawa
- Department of Cell Physiology, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
30
|
de Vries MR, Quax PHA. Inflammation in Vein Graft Disease. Front Cardiovasc Med 2018; 5:3. [PMID: 29417051 PMCID: PMC5787541 DOI: 10.3389/fcvm.2018.00003] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/08/2018] [Indexed: 12/23/2022] Open
Abstract
Bypass surgery is one of the most frequently used strategies to revascularize tissues downstream occlusive atherosclerotic lesions. For venous bypass surgery the great saphenous vein is the most commonly used vessel. Unfortunately, graft efficacy is low due to the development of vascular inflammation, intimal hyperplasia and accelerated atherosclerosis. Moreover, failure of grafts leads to significant adverse outcomes and even mortality. The last couple of decades not much has changed in the treatment of vein graft disease (VGD). However, insight is the cellular and molecular mechanisms of VGD has increased. In this review, we discuss the latest insights on VGD and the role of inflammation in this. We discuss vein graft pathophysiology including hemodynamic changes, the role of vessel wall constitutions and vascular remodeling. We show that profound systemic and local inflammatory responses, including inflammation of the perivascular fat, involve both the innate and adaptive immune system.
Collapse
Affiliation(s)
- Margreet R de Vries
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Paul H A Quax
- Department of Surgery, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
31
|
Protack CD, Foster TR, Hashimoto T, Yamamoto K, Lee MY, Kraehling JR, Bai H, Hu H, Isaji T, Santana JM, Wang M, Sessa WC, Dardik A. Eph-B4 regulates adaptive venous remodeling to improve arteriovenous fistula patency. Sci Rep 2017; 7:15386. [PMID: 29133876 PMCID: PMC5684317 DOI: 10.1038/s41598-017-13071-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 09/13/2017] [Indexed: 12/29/2022] Open
Abstract
Low rates of arteriovenous fistula (AVF) maturation prevent optimal fistula use for hemodialysis; however, the mechanism of venous remodeling in the fistula environment is not well understood. We hypothesized that the embryonic venous determinant Eph-B4 mediates AVF maturation. In human AVF and a mouse aortocaval fistula model, Eph-B4 protein expression increased in the fistula vein; expression of the arterial determinant Ephrin-B2 also increased. Stimulation of Eph-B-mediated signaling with Ephrin-B2/Fc showed improved fistula patency with less wall thickness. Mutagenesis studies showed that tyrosine-774 is critical for Eph-B4 signaling and administration of inactive Eph-B4-Y774F increased fistula wall thickness. Akt1 expression also increased in AVF; Akt1 knockout mice showed reduced fistula diameter and wall thickness. In Akt1 knockout mice, stimulation of Eph-B signaling with Ephrin-B2/Fc showed no effect on remodeling. These results show that AVF maturation is associated with acquisition of dual arteriovenous identity; increased Eph-B activity improves AVF patency. Inhibition of Akt1 function abolishes Eph-B-mediated venous remodeling suggesting that Eph-B4 regulates AVF venous adaptation through an Akt1-mediated mechanism.
Collapse
Affiliation(s)
- Clinton D Protack
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Trenton R Foster
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Takuya Hashimoto
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, VA Connecticut Healthcare System, West Haven, CT, USA.,Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan
| | - Kota Yamamoto
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, VA Connecticut Healthcare System, West Haven, CT, USA.,Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan
| | - Monica Y Lee
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Jan R Kraehling
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Hualong Bai
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Haidi Hu
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Toshihiko Isaji
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, Yale School of Medicine, New Haven, CT, USA.,Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan
| | - Jeans M Santana
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Mo Wang
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - William C Sessa
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA.,Department of Pharmacology, Yale School of Medicine, New Haven, CT, USA
| | - Alan Dardik
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA. .,Department of Surgery, Yale School of Medicine, New Haven, CT, USA. .,Department of Surgery, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
32
|
Hernandez-Lopez R, Chavez-Gonzalez A, Torres-Barrera P, Moreno-Lorenzana D, Lopez-DiazGuerrero N, Santiago-German D, Isordia-Salas I, Smadja D, C. Yoder M, Majluf-Cruz A, Alvarado-Moreno JA. Reduced proliferation of endothelial colony-forming cells in unprovoked venous thromboembolic disease as a consequence of endothelial dysfunction. PLoS One 2017; 12:e0183827. [PMID: 28910333 PMCID: PMC5598948 DOI: 10.1371/journal.pone.0183827] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 08/11/2017] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Venous thromboembolic disease (VTD) is a public health problem. We recently reported that endothelial colony-forming cells (ECFCs) derived from endothelial cells (EC) (ECFC-ECs) from patients with VTD have a dysfunctional state. For this study, we proposed that a dysfunctional status of these cells generates a reduction of its proliferative ability, which is also associated with senescence and reactive oxygen species (ROS). METHODS AND RESULTS Human mononuclear cells (MNCs) were obtained from peripheral blood from 40 healthy human volunteers (controls) and 50 patients with VTD matched by age (20-50 years) and sex to obtain ECFCs. We assayed their proliferative ability with plasma of patients and controls and supernatants of cultures from ECFC-ECs, senescence-associated β-galactosidase (SA-β-gal), ROS, and expression of ephrin-B2/Eph-B4 receptor. Compared with cells from controls, cells from VTD patients showed an 8-fold increase of ECFCs that emerged 1 week earlier, reduced proliferation at long term (39%) and, in passages 4 and 10, a highly senescent rate (30±1.05% vs. 91.3±15.07%, respectively) with an increase of ROS and impaired expression of ephrin-B2/Eph-4 genes. Proliferation potential of cells from VTD patients was reduced in endothelial medium [1.4±0.22 doubling population (DP)], control plasma (1.18±0.31 DP), or plasma from VTD patients (1.65±0.27 DP). CONCLUSIONS As compared with controls, ECFC-ECs from individuals with VTD have higher oxidative stress, proliferation stress, cellular senescence, and low proliferative potential. These findings suggest that patients with a history of VTD are ECFC-ECs dysfunctional that could be associated to permanent risk for new thrombotic events.
Collapse
Affiliation(s)
- Rubicel Hernandez-Lopez
- Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
- Posgrado en Biologia Experimental, Universidad Autonoma Metropolitana, Iztapalapa. Mexico City, Mexico
| | - Antonieta Chavez-Gonzalez
- Unidad de Investigacion Medica en Enfermedades Oncologicas, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Patricia Torres-Barrera
- Unidad de Investigacion Medica en Enfermedades Oncologicas, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Dafne Moreno-Lorenzana
- Unidad de Investigacion Medica en Enfermedades Oncologicas, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Norma Lopez-DiazGuerrero
- Posgrado en Biologia Experimental, Universidad Autonoma Metropolitana, Iztapalapa. Mexico City, Mexico
| | | | - Irma Isordia-Salas
- Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - David Smadja
- Paris Descartes University, INSERM UMR-S 1140, Faculté de Pharmacie de Paris, Paris, France
- AP-HP, Hôpital Européen Georges Pompidou, Hematology department, Paris, France
| | - Mervin C. Yoder
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Abraham Majluf-Cruz
- Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - J. Antonio Alvarado-Moreno
- Unidad de Investigacion Medica en Trombosis, Hemostasia y Aterogenesis, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| |
Collapse
|
33
|
Isaji T, Hashimoto T, Yamamoto K, Santana JM, Yatsula B, Hu H, Bai H, Jianming G, Kudze T, Nishibe T, Dardik A. Improving the Outcome of Vein Grafts: Should Vascular Surgeons Turn Veins into Arteries? Ann Vasc Dis 2017; 10:8-16. [PMID: 29034014 PMCID: PMC5579803 DOI: 10.3400/avd.ra.17-00008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 01/26/2017] [Indexed: 01/21/2023] Open
Abstract
Autogenous vein grafts remain the gold standard conduit for arterial bypass, particularly for the treatment of critical limb ischemia. Vein graft adaptation to the arterial environment, i.e., adequate dilation and wall thickening, contributes to the superior performance of vein grafts. However, abnormal venous wall remodeling with excessive neointimal hyperplasia commonly causes vein graft failure. Since the PREVENT trials failed to improve vein graft outcomes, new strategies focus on the adaptive response of the venous endothelial cells to the post-surgical arterial environment. Eph-B4, the determinant of venous endothelium during embryonic development, remains expressed and functional in adult venous tissue. After surgery, vein grafts lose their venous identity, with loss of Eph-B4 expression; however, arterial identity is not gained, consistent with loss of all vessel identity. In mouse vein grafts, stimulation of venous Eph-B4 signaling promotes retention of venous identity in endothelial cells and is associated with vein graft walls that are not thickened. Eph-B4 regulates downstream signaling pathways of relevance to vascular biology, including caveolin-1, Akt, and endothelial nitric oxide synthase (eNOS). Regulation of the Eph-B4 signaling pathway may be a novel therapeutic target to prevent vein graft failure.
Collapse
Affiliation(s)
- Toshihiko Isaji
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA.,Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan
| | - Takuya Hashimoto
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA.,Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven, Connecticut, USA
| | - Kota Yamamoto
- Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan
| | - Jeans M Santana
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA
| | - Bogdan Yatsula
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA
| | - Haidi Hu
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA
| | - Hualong Bai
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA
| | - Guo Jianming
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA
| | - Tambudzai Kudze
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA
| | - Toshiya Nishibe
- Department of Cardiovascular Surgery, Tokyo Medical University, Tokyo, Japan
| | - Alan Dardik
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut, USA.,Department of Surgery, VA Connecticut Healthcare Systems, West Haven, Connecticut, USA
| |
Collapse
|
34
|
Zhao J, Jourd'heuil FL, Xue M, Conti D, Lopez-Soler RI, Ginnan R, Asif A, Singer HA, Jourd'heuil D, Long X. Dual Function for Mature Vascular Smooth Muscle Cells During Arteriovenous Fistula Remodeling. J Am Heart Assoc 2017; 6:e004891. [PMID: 28360226 PMCID: PMC5533005 DOI: 10.1161/jaha.116.004891] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/19/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND The arteriovenous fistula (AVF) is the preferred form of hemodialysis access for patients with chronic kidney disease. However, AVFs are associated with significant problems including high incidence of both early and late failures, usually attributed to inadequate venous arterialization and neointimal hyperplasia, respectively. Understanding the cellular basis of venous remodeling in the setting of AVF could provide targets for improving AVF patency rates. METHODS AND RESULTS A novel vascular smooth muscle cell (VSMC) lineage tracing reporter mouse, Myh11-Cre/ERT2-mTmG, was used to track mature VSMCs in a clinically relevant AVF mouse model created by a jugular vein branch end to carotid artery side anastomosis. Prior to AVF surgery, differentiated medial layer VSMCs were labeled with membrane green fluorescent protein (GFP) following tamoxifen induction. Four weeks after AVF surgery, we observed medial VSMC layer thickening in the middle region of the arterialized vein branch. This thickened medial VSMC layer was solely composed of differentiated VSMCs that were GFP+/MYH11+/Ki67-. Extensive neointimal hyperplasia occurred in the AVF region proximal to the anastomosis site. Dedifferentiated VSMCs (GFP+/MYH11-) were a major cellular component of the neointima. Examination of failed human AVF samples revealed that the processes of VSMC phenotypic modulation and intimal hyperplasia, as well as medial VSMC layer thickening, also occurred in human AVFs. CONCLUSIONS We demonstrated a dual function for mature VSMCs in AVF remodeling, with differentiated VSMCs contributing to medial wall thickening towards venous maturation and dedifferentiated VSMCs contributing to neointimal hyperplasia. These results provide valuable insights into the mechanisms underlying venous adaptations during AVF remodeling.
Collapse
Affiliation(s)
- Jinjing Zhao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY
| | | | - Min Xue
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY
- Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, China
| | - David Conti
- Department of Surgery, Transplantation Group, Albany Medical College, Albany, NY
| | | | - Roman Ginnan
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY
| | - Arif Asif
- Jersey Shore University Medical Center, Hackensack-Meridian Health Seton Hall-Hackensack Meridian School of Medicine, Neptune, NJ
| | - Harold A Singer
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY
| | - David Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY
| | - Xiaochun Long
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY
| |
Collapse
|
35
|
Bai H, Li X, Hashimoto T, Hu H, Foster TR, Hanisch JJ, Santana JM, Dardik A. Patch Angioplasty in the Rat Aorta or Inferior Vena Cava. J Vis Exp 2017. [PMID: 28287579 DOI: 10.3791/55253] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pericardial patches are commonly used in vascular surgery to close vessels. To facilitate studies of the neointimal hyperplasia that forms on the patch, we developed a rat model of patch angioplasty that can be used in either a vein or an artery, creating a patch venoplasty or a patch arterioplasty, respectively. Technical aspects of this model are discussed. The infra-renal IVC or aorta are dissected and then clamped proximally and distally. A 3 mm venotomy or arteriotomy is performed in the infrarenal inferior vena cava or aorta of 6 to 8 week-old Wistar rats. A bovine pericardial patch (3 mm x 1.5 mm x 0.6 mm) is then used to close the site using a 10-0 nylon suture. Compared to arterial patches, venous patches show increased neointimal thickness on postoperative day 7. This novel model of pericardial patch angioplasty can be used to examine neointimal hyperplasia on vascular biomaterials, as well as to compare the differences between the arterial and venous environments.
Collapse
Affiliation(s)
- Hualong Bai
- Department of Surgery, Yale University; Vascular Biology and Therapeutics Program, Yale University; Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University; Basic Medical College of Zhengzhou University; VA Connecticut Healthcare Systems, West Haven, CT
| | - Xin Li
- Department of Vascular Surgery, Xiangya Second Hospital of Central South University, Changsha, China
| | - Takuya Hashimoto
- Department of Surgery, Yale University; Vascular Biology and Therapeutics Program, Yale University; VA Connecticut Healthcare Systems, West Haven, CT
| | - Haidi Hu
- Department of Surgery, Yale University; Vascular Biology and Therapeutics Program, Yale University; VA Connecticut Healthcare Systems, West Haven, CT
| | - Trenton R Foster
- Department of Surgery, Yale University; Vascular Biology and Therapeutics Program, Yale University; VA Connecticut Healthcare Systems, West Haven, CT
| | - Jesse J Hanisch
- Department of Surgery, Yale University; Vascular Biology and Therapeutics Program, Yale University; VA Connecticut Healthcare Systems, West Haven, CT
| | - Jeans M Santana
- Department of Surgery, Yale University; Vascular Biology and Therapeutics Program, Yale University; VA Connecticut Healthcare Systems, West Haven, CT
| | - Alan Dardik
- Department of Surgery, Yale University; Vascular Biology and Therapeutics Program, Yale University; VA Connecticut Healthcare Systems, West Haven, CT;
| |
Collapse
|
36
|
Bai H, Wang M, Foster TR, Hu H, He H, Hashimoto T, Hanisch JJ, Santana JM, Xing Y, Dardik A. Pericardial patch venoplasty heals via attraction of venous progenitor cells. Physiol Rep 2016; 4:e12841. [PMID: 27354544 PMCID: PMC4923240 DOI: 10.14814/phy2.12841] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 06/02/2016] [Indexed: 11/24/2022] Open
Abstract
Pericardial patches are commonly used during cardiovascular surgery to close blood vessels. In arteries, patches accumulate arterial progenitor cells; we hypothesized that venous patches would accumulate venous progenitor cells, in the absence of arterial pressure. We developed a novel rat inferior vena cava (IVC) venotomy model and repaired it with a pericardial patch. Cells infiltrated the patch to form a thick neointima by day 7; some cells were CD34(+)/VEGFR2(+) and CD31(+)/Eph-B4(+) consistent with development of venous identity in the healing patch. Compared to arterial patches, the venous patches had increased neointimal thickness at day 7 without any pseudoaneurysms. Addition of an arteriovenous fistula (AVF) to increase blood flow on the patch resulted in reduced patch neointimal thickness and proliferation, but neointimal thickness was not reversible with AVF ligation. These results show that rat patch venoplasty is a novel model of aggressive venous neointimal hyperplasia.
Collapse
Affiliation(s)
- Hualong Bai
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, Connecticut Basic Medical College of Zhengzhou University, Henan, China Department of Vascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Mo Wang
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, Connecticut Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Trenton R Foster
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Haidi Hu
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, Connecticut Department of Surgery, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Hao He
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, Connecticut Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Takuya Hashimoto
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, Connecticut Department of Surgery, VA Connecticut Healthcare System, West Haven, Connecticut
| | - Jesse J Hanisch
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Jeans M Santana
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, Connecticut
| | - Ying Xing
- Basic Medical College of Zhengzhou University, Henan, China
| | - Alan Dardik
- The Vascular Biology and Therapeutics Program and the Department of Surgery, Yale University School of Medicine, New Haven, Connecticut Department of Surgery, VA Connecticut Healthcare System, West Haven, Connecticut
| |
Collapse
|
37
|
Wong DJ, Lu DY, Protack CD, Kuwahara G, Bai H, Sadaghianloo N, Tellides G, Dardik A. Ephrin type-B receptor 4 activation reduces neointimal hyperplasia in human saphenous vein in vitro. J Vasc Surg 2016; 63:795-804. [PMID: 25446283 PMCID: PMC4409444 DOI: 10.1016/j.jvs.2014.09.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 09/24/2014] [Indexed: 11/21/2022]
Abstract
BACKGROUND Vein bypass is an essential therapy for patients with advanced peripheral and coronary artery disease despite development of neointimal hyperplasia. We have shown that stimulation of the receptor tyrosine kinase ephrin type-B receptor 4 (Eph-B4) with its ligand ephrin-B2 prevents neointimal hyperplasia in murine vein grafts. This study determines whether Eph-B4 in adult human veins is capable of phosphorylation and activation of downstream signaling pathways, as well as functional to release nitric oxide (NO) and prevent neointimal hyperplasia in vitro. METHODS Discarded human saphenous veins were taken from the operating room and placed in organ culture without or with ephrin-B2/Fc (2 μg/mL) for 14 days, and the neointima/media ratio was measured in matched veins. Primary human umbilical vein endothelial cells were treated with ephrin-B2/Fc (2 μg/mL) and examined with quantitative polymerase chain reaction, Western blot, immunoassays, and for release of NO. Ephrin-B2/Fc (2 μg/mL) was placed on the adventitia of saphenous veins treated with arterial shear stress for 24 hours in a bioreactor and activated Eph-B4 examined with immunofluorescence. RESULTS The baseline intima/media ratio in saphenous vein rings was 0.456 ± 0.097, which increased to 0.726 ± 0.142 in untreated veins after 14 days in organ culture but only to 0.630 ± 0.132 in veins treated with ephrin-B2/Fc (n = 19, P = .017). Ephrin-B2/Fc stimulated Akt, endothelial NO synthase and caveolin-1 phosphorylation, and NO release (P = .007) from human umbilical vein endothelial cells (n = 6). Ephrin-B2/Fc delivered to the adventitia stimulated endothelial Eph-B4 phosphorylation after 24 hours of arterial stress in a bioreactor (n = 3). CONCLUSIONS Eph-B4 is present and functional in adult human saphenous veins, with intact downstream signaling pathways capable of NO release and prevention of neointimal hyperplasia in vitro. Adventitial delivery of ephrin-B2/Fc activates endothelial Eph-B4 in saphenous veins treated with arterial shear stress in vitro. These results suggest that stimulation of Eph-B4 function may be a candidate strategy for translation to human clinical trials designed to inhibit venous neointimal hyperplasia.
Collapse
Affiliation(s)
- Daniel J Wong
- Vascular Biology and Therapeutics (VBT) Program and the Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Daniel Y Lu
- Vascular Biology and Therapeutics (VBT) Program and the Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Clinton D Protack
- Vascular Biology and Therapeutics (VBT) Program and the Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Go Kuwahara
- Vascular Biology and Therapeutics (VBT) Program and the Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Hualong Bai
- Vascular Biology and Therapeutics (VBT) Program and the Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Nirvana Sadaghianloo
- Vascular Biology and Therapeutics (VBT) Program and the Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - George Tellides
- Vascular Biology and Therapeutics (VBT) Program and the Department of Surgery, Yale University School of Medicine, New Haven, Conn; Department of Surgery, VA Connecticut Healthcare System, West Haven, Conn
| | - Alan Dardik
- Vascular Biology and Therapeutics (VBT) Program and the Department of Surgery, Yale University School of Medicine, New Haven, Conn; Department of Surgery, VA Connecticut Healthcare System, West Haven, Conn.
| |
Collapse
|
38
|
Hashimoto T, Tsuneki M, Foster TR, Santana JM, Bai H, Wang M, Hu H, Hanisch JJ, Dardik A. Membrane-mediated regulation of vascular identity. BIRTH DEFECTS RESEARCH. PART C, EMBRYO TODAY : REVIEWS 2016; 108:65-84. [PMID: 26992081 PMCID: PMC5310768 DOI: 10.1002/bdrc.21123] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 02/22/2016] [Indexed: 02/06/2023]
Abstract
Vascular diseases span diverse pathology, but frequently arise from aberrant signaling attributed to specific membrane-associated molecules, particularly the Eph-ephrin family. Originally recognized as markers of embryonic vessel identity, Eph receptors and their membrane-associated ligands, ephrins, are now known to have a range of vital functions in vascular physiology. Interactions of Ephs with ephrins at cell-to-cell interfaces promote a variety of cellular responses such as repulsion, adhesion, attraction, and migration, and frequently occur during organ development, including vessel formation. Elaborate coordination of Eph- and ephrin-related signaling among different cell populations is required for proper formation of the embryonic vessel network. There is growing evidence supporting the idea that Eph and ephrin proteins also have postnatal interactions with a number of other membrane-associated signal transduction pathways, coordinating translation of environmental signals into cells. This article provides an overview of membrane-bound signaling mechanisms that define vascular identity in both the embryo and the adult, focusing on Eph- and ephrin-related signaling. We also discuss the role and clinical significance of this signaling system in normal organ development, neoplasms, and vascular pathologies.
Collapse
Affiliation(s)
- Takuya Hashimoto
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, Connecticut
- Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan
| | - Masayuki Tsuneki
- Division of Cancer Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Trenton R. Foster
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Jeans M. Santana
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Hualong Bai
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
- Department of Vascular Surgery, The 1st Affiliated Hospital of Zhengzhou University, Henan, China
| | - Mo Wang
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Haidi Hu
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Jesse J. Hanisch
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Alan Dardik
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, Connecticut
| |
Collapse
|
39
|
Wang M, Collins MJ, Foster TR, Bai H, Hashimoto T, Santana JM, Shu C, Dardik A. Eph-B4 mediates vein graft adaptation by regulation of endothelial nitric oxide synthase. J Vasc Surg 2016; 65:179-189. [PMID: 26817610 DOI: 10.1016/j.jvs.2015.11.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/10/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vein graft adaptation is characterized by loss of expression of the tyrosine kinase receptor Eph-B4, the embryonic determinant of venous identity, without increased expression of its ligand ephrin-B2, the embryonic determinant of arterial identity. Endothelial nitric oxide synthase (eNOS) is an important mediator of vessel remodeling. We hypothesized that the mechanism of action of Eph-B4 during vein graft adaptation might be through regulation of downstream eNOS activity. METHODS Mouse lung endothelial cells were stimulated with ephrin-B2/Fc, without and with preclustering, without and with the eNOS inhibitor Nω-nitro-l-arginine methyl ester hydrochloride or the Eph-B4 inhibitor NVP-BHG712, and assessed by Western blot and immunofluorescence for eNOS and Eph-B4 phosphorylation. Nitric oxide (NO) production was assessed using an NO-specific chemiluminescence analyzer. Cell migration was assessed using a Transwell assay. Human and mouse vein graft specimens were examined for eNOS activity by Western blot, and vessel remodeling was assessed in vein grafts in wild-type or eNOS knockout mice. RESULTS Ephrin-B2/Fc stimulated both Eph-B4 and eNOS phosphorylation in a bimodal temporal distribution (n = 4; P < .05), with preclustered ephrin-B2/Fc causing prolonged peak Eph-B4 and eNOS phosphorylation as well as altered subcellular localization (n = 4; P < .05). Ephrin-B2/Fc increased NO release (n = 3; P < .01) as well as increased endothelial cell migration (n = 6; P < .05) in an eNOS-dependent fashion. Both human and mouse vein grafts showed increased eNOS phosphorylation compared with normal veins (n = 3; P < .05). Vein grafts from eNOS knockout mice showed less dilation and less wall thickening compared with wild-type vein grafts (n = 7; P < .05). CONCLUSIONS eNOS is a mediator of vein graft adaptation to the arterial environment. Eph-B4 stimulates eNOS phosphorylation in vitro and may mediate vein graft adaptation by regulation of eNOS activity in vivo.
Collapse
Affiliation(s)
- Mo Wang
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China; Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Michael J Collins
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Trenton R Foster
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Hualong Bai
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Takuya Hashimoto
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Jeans M Santana
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn
| | - Chang Shu
- Department of Vascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Alan Dardik
- Vascular Biology and Therapeutics Program and Department of Surgery, Yale University School of Medicine, New Haven, Conn; Department of Surgery, VA Connecticut Healthcare System, West Haven, Conn.
| |
Collapse
|
40
|
Cui X, Lu YW, Lee V, Kim D, Dorsey T, Wang Q, Lee Y, Vincent P, Schwarz J, Dai G. Venous Endothelial Marker COUP-TFII Regulates the Distinct Pathologic Potentials of Adult Arteries and Veins. Sci Rep 2015; 5:16193. [PMID: 26537113 PMCID: PMC4633649 DOI: 10.1038/srep16193] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 10/05/2015] [Indexed: 12/21/2022] Open
Abstract
Arteries and veins have very different susceptibility to certain vascular diseases such as atherosclerosis and vascular calcification. The molecular mechanisms of these differences are not fully understood. In this study, we discovered that COUP-TFII, a transcription factor critical for establishing the venous identity during embryonic vascular development, also regulates the pathophysiological functions of adult blood vessels, especially those directly related to vascular diseases. Specifically, we found that suppression of COUP-TFII in venous ECs switched its phenotype toward pro-atherogenic by up-regulating the expression of inflammatory genes and down-regulating anti-thrombotic genes. ECs with COUP-TFII knockdown also readily undergo endothelial-to-mesenchymal transition (EndoMT) and subsequent osteogenic differentiation with dramatically increased osteogenic transcriptional program and calcium deposition. Consistently, over-expression of COUP-TFII led to the completely opposite effects. In vivo validation of these pro-atherogenic and osteogenic genes also demonstrates a broad consistent differential expression pattern in mouse aorta vs. vena cava ECs, which cannot be explained by the difference in hemodynamic flow. These data reveal phenotypic modulation by different levels of COUP-TFII in arterial and venous ECs, and suggest COUP-TFII may play an important role in the different susceptibilities of arteries and veins to vascular diseases such as atherosclerosis and vascular calcification.
Collapse
Affiliation(s)
- Xiaofeng Cui
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, China 430070.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Yao Wei Lu
- The Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - Vivian Lee
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Diana Kim
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Taylor Dorsey
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Qingjie Wang
- Neural Stem Cell Institute, Rensselaer, NY 12144, USA
| | - Young Lee
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | - Peter Vincent
- The Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - John Schwarz
- The Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - Guohao Dai
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA.,Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| |
Collapse
|
41
|
Abstract
Formation of arterial vasculature, here termed arteriogenesis, is a central process in embryonic vascular development as well as in adult tissues. Although the process of capillary formation, angiogenesis, is relatively well understood, much remains to be learned about arteriogenesis. Recent discoveries point to the key role played by vascular endothelial growth factor receptor 2 in control of this process and to newly identified control circuits that dramatically influence its activity. The latter can present particularly attractive targets for a new class of therapeutic agents capable of activation of this signaling cascade in a ligand-independent manner, thereby promoting arteriogenesis in diseased tissues.
Collapse
Affiliation(s)
- Michael Simons
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (M.S., A.E.) and Departments of Cell Biology (M.S.) and Molecular Physiology (A.E.), Yale University School of Medicine, New Haven, CT.
| | - Anne Eichmann
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (M.S., A.E.) and Departments of Cell Biology (M.S.) and Molecular Physiology (A.E.), Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
42
|
Temporal regulation of venous extracellular matrix components during arteriovenous fistula maturation. J Vasc Access 2014; 16:93-106. [PMID: 25262757 DOI: 10.5301/jva.5000290] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2014] [Indexed: 01/08/2023] Open
Abstract
PURPOSE The venous limb of arteriovenous fistulae (AVF) adapts to the arterial environment by dilation and wall thickening; however, the temporal regulation of the expression of extracellular matrix (ECM) components in the venous limb of the maturing AVF has not been well characterized. We used a murine model of AVF maturation that recapitulates human AVF maturation to determine the temporal pattern of expression of these ECM components. METHODS Aortocaval fistulae were created in C57BL/6J mice and the venous limb was analyzed on postoperative days 1, 3, 7, 21, and 42. A gene microarray analysis was performed on day 7; results were confirmed by qPCR, histology, and immunohistochemistry. Proteases, protease inhibitors, collagens, glycoproteins, and other non-collagenous proteins were characterized. RESULTS The maturing AVF has increased expression of many ECM components, including increased collagen and elastin. Matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinase 1 (TIMP1) showed increased mRNA and protein expression during the first 7 days of maturation. Increased collagen and elastin expression was also significant at day 7. Expression of structural proteins was increased later during AVF maturation. Osteopontin (OPN) expression was increased at day 1 and sustained during AVF maturation. CONCLUSIONS During AVF maturation, there is significantly increased expression of ECM components, each of which shows distinct temporal patterns during AVF maturation. Increased expression of regulatory proteins such as MMP and TIMP precedes increased expression of structural proteins such as collagen and elastin, potentially mediating a controlled pattern of ECM degradation and vessel remodeling without structural failure.
Collapse
|
43
|
Model LS, Hall MR, Wong DJ, Muto A, Kondo Y, Ziegler KR, Feigel A, Quint C, Niklason L, Dardik A. Arterial shear stress reduces eph-b4 expression in adult human veins. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2014; 87:359-71. [PMID: 25191151 PMCID: PMC4144290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Vein graft adaptation to the arterial environment is characterized by loss of venous identity, with reduced Ephrin type-B receptor 4 (Eph-B4) expression but without increased Ephrin-B2 expression. We examined changes of vessel identity of human saphenous veins in a flow circuit in which shear stress could be precisely controlled. Medium circulated at arterial or venous magnitudes of laminar shear stress for 24 hours; histologic, protein, and RNA analyses of vein segments were performed. Vein endothelium remained viable and functional, with platelet endothelial cell adhesion molecule (PECAM)-expressing cells on the luminal surface. Venous Eph-B4 expression diminished (p = .002), Ephrin-B2 expression was not induced (p = .268), and expression of osteopontin (p = .002) was increased with exposure to arterial magnitudes of shear stress. Similar changes were not found in veins placed under venous flow or static conditions. These data show that human saphenous veins remain viable during ex vivo application of shear stress in a bioreactor, without loss of the venous endothelium. Arterial magnitudes of shear stress cause loss of venous identity without gain of arterial identity in human veins perfused ex vivo. Shear stress alone, without immunologic or hormonal influence, is capable of inducing changes in vessel identity and, specifically, loss of venous identity.
Collapse
Affiliation(s)
- Lynn S. Model
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut
| | - Michael R. Hall
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut
| | - Daniel J. Wong
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut
| | - Akihito Muto
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut
| | - Yuka Kondo
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut
| | - Kenneth R. Ziegler
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut
| | - Amanda Feigel
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut
| | - Clay Quint
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut,Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut
| | - Laura Niklason
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut,Department of Biomedical Engineering, Yale School of Engineering & Applied Science, New Haven, Connecticut
| | - Alan Dardik
- Department of Surgery, Yale School of Medicine, New Haven, Connecticut,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, Connecticut,VA Connecticut Healthcare System, West Haven, Connecticut,To whom all correspondence should be addressed: Alan Dardik, Yale School of Medicine, 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089; Tele: 203-737-2082; Fax: 203-737-2290;
| |
Collapse
|
44
|
Zhu H, Hou CC, Luo LF, Hu YJ, Yang WX. Endometrial stromal cells and decidualized stromal cells: origins, transformation and functions. Gene 2014; 551:1-14. [PMID: 25168894 DOI: 10.1016/j.gene.2014.08.047] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 07/24/2014] [Accepted: 08/24/2014] [Indexed: 10/24/2022]
Abstract
Decidualization of endometrium, which is characterized by endometrial stromal cell (ESC) decidualization, vascular reconstruction, immune cell recruitment, and plentiful molecule production, is a crucial step for uterus to become receptive for embryo. When implantation takes place, ESCs surround and directly interact with embryo. Decidualized stromal cells (DSCs) are of great importance in endometrial decidualization, having a broad function in regulating immune activity and vascular remodeling of uterus. DSCs are shown to have a higher metabolic level and looser cytoskeleton than ESCs. What's the origin of ESCs and how ESCs successfully transform into DSCs had puzzled scientists in the last decades. Breakthrough had been achieved recently, and many studies had elucidated some of the characters and functions of DSCs. However, several questions still remain unclear. This paper reviews current understanding of where ESCs come from and how ESCs differentiate into DSCs, summarizes some characters and functions of DSCs, analyzes current studies and their limitations and points out research areas that need further investigation.
Collapse
Affiliation(s)
- Ha Zhu
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Cong-Cong Hou
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ling-Feng Luo
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yan-Jun Hu
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, China.
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
45
|
Lee VK, Kim DY, Ngo H, Lee Y, Seo L, Yoo SS, Vincent PA, Dai G. Creating perfused functional vascular channels using 3D bio-printing technology. Biomaterials 2014; 35:8092-102. [PMID: 24965886 DOI: 10.1016/j.biomaterials.2014.05.083] [Citation(s) in RCA: 318] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 05/28/2014] [Indexed: 11/25/2022]
Abstract
We developed a methodology using 3D bio-printing technology to create a functional in vitro vascular channel with perfused open lumen using only cells and biological matrices. The fabricated vasculature has a tight, confluent endothelium lining, presenting barrier function for both plasma protein and high-molecular weight dextran molecule. The fluidic vascular channel is capable of supporting the viability of tissue up to 5 mm in distance at 5 million cells/mL density under the physiological flow condition. In static-cultured vascular channels, active angiogenic sprouting from the vessel surface was observed whereas physiological flow strongly suppressed this process. Gene expression analysis was reported in this study to show the potential of this vessel model in vascular biology research. The methods have great potential in vascularized tissue fabrication using 3D bio-printing technology as the vascular channel is simultaneously created while cells and matrix are printed around the channel in desired 3D patterns. It can also serve as a unique experimental tool for investigating fundamental mechanisms of vascular remodeling with extracellular matrix and maturation process under 3D flow condition.
Collapse
Affiliation(s)
- Vivian K Lee
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| | - Diana Y Kim
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| | - Haygan Ngo
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| | - Young Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Lan Seo
- Department of Biological Sciences, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| | - Seung-Schik Yoo
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Peter A Vincent
- Center for Cardiovascular Sciences, Albany Medical College, Albany, NY 12208, USA
| | - Guohao Dai
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA.
| |
Collapse
|
46
|
Shi HT, Wang Y, Jia LX, Qin YW, Liu Y, Li HH, Qi YF, Du J. Cathepsin S contributes to macrophage migration via degradation of elastic fibre integrity to facilitate vein graft neointimal hyperplasia. Cardiovasc Res 2014; 101:454-463. [DOI: 10.1093/cvr/cvt273] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
47
|
Yamamoto K, Li X, Shu C, Miyata T, Dardik A. Technical aspects of the mouse aortocaval fistula. J Vis Exp 2013:e50449. [PMID: 23892387 DOI: 10.3791/50449] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Technical aspects of creating an arteriovenous fistula in the mouse are discussed. Under general anesthesia, an abdominal incision is made, and the aorta and inferior vena cava (IVC) are exposed. The proximal infrarenal aorta and the distal aorta are dissected for clamp placement and needle puncture, respectively. Special attention is paid to avoid dissection between the aorta and the IVC. After clamping the aorta, a 25 G needle is used to puncture both walls of the aorta into the IVC. The surrounding connective tissue is used for hemostatic compression. Successful creation of the AVF will show pulsatile arterial blood flow in the IVC. Further confirmation of successful AVF can be achieved by post-operative Doppler ultrasound.
Collapse
Affiliation(s)
- Kota Yamamoto
- Department of Surgery and the Interdepartmental Program in Vascular Biology and Therapeutics, Yale University, USA
| | | | | | | | | |
Collapse
|
48
|
Yang C, Guo Y, Jadlowiec CC, Li X, Lv W, Model LS, Collins MJ, Kondo Y, Muto A, Shu C, Dardik A. Vascular endothelial growth factor-A inhibits EphB4 and stimulates delta-like ligand 4 expression in adult endothelial cells. J Surg Res 2013; 183:478-86. [PMID: 23394931 DOI: 10.1016/j.jss.2013.01.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Revised: 12/18/2012] [Accepted: 01/03/2013] [Indexed: 10/27/2022]
Abstract
BACKGROUND During vein graft adaptation to the arterial circulation, vascular endothelial growth factor (VEGF) A expression transiently increases before becoming downregulated; however, the role of VEGF-A in venous remodeling is not clear. In addition, although VEGF-A stimulates angiogenesis and determines arterial identity in nascent arterial endothelial cells (EC), the role of VEGF-A in regulating identity in adult venous EC is also not clear. MATERIALS AND METHODS EC, wild type (EphB4+/+) or heterozygous knockout (EphB4+/-), were stimulated with VEGF-A (0-100 ng/mL) and examined with quantitative polymerase chain reaction and western blotting. RESULTS VEGF-A (100 ng/mL) inhibited expression of EphB4 and stimulated expression of delta-like ligand 4 (dll4) but did not stimulate either notch or EphrinB2 expression in adult venous EC. Pretreatment with VEGF receptor 2-neutralizing antibody abolished VEGF-stimulated downregulation of EphB4 but not the upregulation of dll4. Pretreatment with PD98059 or wortmannin showed that VEGF-A downregulation of EphB4 and upregulation of dll4 are mitogen-activated protein kinase kinase and extracellular signal-regulated kinase dependent but phosphatidylinositol 3 kinase-Akt independent. Compared with VEGF-induced EphB4 downregulation and dll4 upregulation in control EC, reduced EphB4 signaling in EphB4+/- EC showed even further downregulation of EphB4 and upregulation of dll4. CONCLUSIONS Despite the genetic programming of arterial and venous EC fate, VEGF-A can repress venous identity in adult venous EC without induction of arterial identity. These changes in adult EC in vitro recapitulate the changes in identity described during vein graft adaptation to the arterial environment in vivo.
Collapse
Affiliation(s)
- Chenzi Yang
- Interdepartmental Program in Vascular Biology and Therapeutics, Yale University School of Medicine, New Haven, Connecticut 06520-8089, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Berard X, Déglise S, Alonso F, Saucy F, Meda P, Bordenave L, Corpataux JM, Haefliger JA. Role of hemodynamic forces in the ex vivo arterialization of human saphenous veins. J Vasc Surg 2013; 57:1371-82. [PMID: 23351647 DOI: 10.1016/j.jvs.2012.09.041] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/31/2012] [Accepted: 09/10/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND Human saphenous vein grafts are one of the salvage bypass conduits when endovascular procedures are not feasible or fail. Understanding the remodeling process that venous grafts undergo during exposure to arterial conditions is crucial to improve their patency, which is often compromised by intimal hyperplasia. The precise role of hemodynamic forces such as shear stress and arterial pressure in this remodeling is not fully characterized. The aim of this study was to determine the involvement of arterial shear stress and pressure on vein wall remodeling and to unravel the underlying molecular mechanisms. METHODS An ex vivo vein support system was modified for chronic (up to 1 week), pulsatile perfusion of human saphenous veins under controlled conditions that permitted the separate control of arterial shear stress and different arterial pressure (7 mm Hg or 70 mm Hg). RESULTS Veins perfused for 7 days under high pressure (70 mm Hg) underwent significant development of a neointima compared with veins exposed to low pressure (7 mm Hg). These structural changes were associated with altered expression of several molecular markers. Exposure to an arterial shear stress under low pressure increased the expression of matrix metalloproteinase (MMP)-2 and MMP-9 and tissue inhibitor of metalloproteinase (TIMP)-1 at the transcript, protein, and activity levels. This increase was enhanced by high pressure, which also increased TIMP-2 protein expression despite decreased levels of the cognate transcript. In contrast, the expression of plasminogen activator inhibitor-1 increased with shear stress but was not modified by pressure. Levels of the venous marker Eph-B4 were decreased under arterial shear stress, and levels of the arterial marker Ephrin-B2 were downregulated under high-pressure conditions. CONCLUSIONS This model is a valuable tool to identify the role of hemodynamic forces and to decipher the molecular mechanisms leading to failure of human saphenous vein grafts. Under ex vivo conditions, arterial perfusion is sufficient to activate the remodeling of human veins, a change that is associated with the loss of specific vein markers. Elevation of pressure generates intimal hyperplasia, even though veins do not acquire arterial markers. CLINICAL RELEVANCE The pathological remodeling of the venous wall, which leads to stenosis and ultimately graft failure, is the main limiting factor of human saphenous vein graft bypass. This remodeling is due to the hemodynamic adaptation of the vein to the arterial environment and cannot be prevented by conventional therapy. To develop a more targeted therapy, a better understanding of the molecular mechanisms involved in intimal hyperplasia is essential, which requires the development of ex vivo models of chronic perfusion of human veins.
Collapse
Affiliation(s)
- Xavier Berard
- Department of Vascular Surgery, Pellegrin Hospital, University of Bordeaux, Bordeaux, France
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Jadlowiec CC, Feigel A, Yang C, Feinstein AJ, Kim ST, Collins MJ, Kondo Y, Muto A, Dardik A. Reduced adult endothelial cell EphB4 function promotes venous remodeling. Am J Physiol Cell Physiol 2012; 304:C627-35. [PMID: 23269240 DOI: 10.1152/ajpcell.00333.2012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Reduced EphB4 expression is observed during vein graft adaptation and is associated with increased venous wall thickening. These findings suggest that EphB4 may mediate normal adult venous endothelial cell (EC) function and vein graft adaptation. We therefore tested the functional significance of EphB4 using EC with genetically reduced EphB4 signaling. EC were isolated from EphB4(+/+) and EphB4(+/-) mice. In vitro function was assessed through EC proliferation, migration, nitric oxide (NO) synthesis, and chemokine production. A mouse vein graft model was used to correlate in vitro findings with in vivo vein grafts. Smooth muscle cells (SMC) were subjected to proliferation and migration assays using EphB4(+/+) and EphB4(+/-) EC-conditioned medium. EphB4(+/-) EC exhibited diminished proliferation (P < 0.0001, n = 6), migration (P < 0.0001, n = 3), and NO production (P = 0.0012, n = 3). EphB4(+/-) EC had increased VEGF-A mRNA (P = 0.0006, n = 6) and protein (P = 0.0106, n = 3) as well as increased secretion of VEGF-A (P = 0.0010, n = 5), PDGF-BB (P < 0.0001, n = 6), and TGF-β1 (P < 0.0001, n = 6). EphB4(+/-)-conditioned medium promoted SMC proliferation (P < 0.0001, n = 7) and migration (P = 0.0358, n = 3). Vein grafts and EphB4(+/-) EC showed similarity with regard to VEGF-A and eNOS mRNA and protein expression. In conclusion, reduced venous EC EphB4 function is associated with a proangiogenic and mitogenic phenotype. EphB4(+/-) EC have increased secretion of SMC mitogens and reduced NO production that correlate with the thickened neointima formed during vein graft adaptation. These findings suggest that EphB4 remains active in adult venous EC and that loss of EphB4 plays a role in vein graft adaptation.
Collapse
Affiliation(s)
- Caroline C Jadlowiec
- The Interdepartmental Program in Vascular Biology and Therapeutics and the Department of Surgery, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | | | |
Collapse
|