1
|
Zawieja SD, Pea GA, Broyhill SE, Patro A, Bromert KH, Norton CE, Kim HJ, Sivasankaran SK, Li M, Castorena-Gonzalez JA, Drumm BT, Davis MJ. Characterization of the cellular components of mouse collecting lymphatic vessels reveals that lymphatic muscle cells are the innate pacemaker cells regulating lymphatic contractions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.24.554619. [PMID: 37662284 PMCID: PMC10473772 DOI: 10.1101/2023.08.24.554619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Collecting lymphatic vessels (cLVs) exhibit spontaneous contractions with a pressure-dependent frequency, but the identity of the lymphatic pacemaker cell is still debated. By analogy to pacemakers in the GI and lower urinary tracts, proposed cLV pacemaker cells include interstitial cells of Cajal like cells (ICLC) or the lymphatic muscle (LMCs) cells themselves. Here we combined immunofluorescence and scRNAseq analyses with electrophysiological methods to examine the cellular constituents of the mouse cLV wall and assess whether any cell type exhibited morphological and functional processes characteristic of pacemaker cells: a continuous if not contiguous network integrated into the electrical syncytium; spontaneous Ca2+ transients; and depolarization-induced propagated contractions. We employed inducible Cre (iCre) mouse models routinely used to target these specific cell populations including: c-kitCreER T2 to target ICLC; PdgfrβCreER T2 to target pericyte-like cells; PdgfrαCreER ™ to target CD34+ adventitial cells and ICLC; and Myh11CreER T2 to target LMCs directly. These specific inducible Cre lines were crossed to the fluorescent reporter ROSA26mT/mG, the genetically encoded Ca2+ sensor GCaMP6f, and the light-activated cation channel rhodopsin2 (ChR2). c-KitCreER T2 labeled both a sparse population of LECs and round adventitial cells that responded to the mast cell activator compound 48-80. PdgfrβCreER T2 drove recombination in both adventitial cells and LMCs, limiting its power to discriminate a pericyte-specific population. PdgfrαCreER ™ labeled a large population of interconnected, oak leaf-shaped cells primarily along the adventitial surface of the vessel. Of these cells, only LMCs consistently, but heterogeneously, displayed spontaneous Ca2+ events during the diastolic period of the contraction cycle, and whose frequency was modulated in a pressure-dependent manner. Optogenetic depolarization through the expression of ChR2 under control of Myh11CreER T2 , but not PdgfrαCreER ™ or c-KitCreER T2 , resulted in propagated contractions upon photo-stimulation. Membrane potential recordings in LMCs demonstrated that the rate of diastolic depolarization significantly correlated with contraction frequency. These findings support the conclusion that LMCs, or a subset of LMCs, are responsible for mouse cLV pacemaking.
Collapse
Affiliation(s)
- S D Zawieja
- Dept. of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri
| | - G A Pea
- Dept. of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri
| | - S E Broyhill
- Dept. of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri
| | - A Patro
- Dept. of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri
| | - K H Bromert
- Dept. of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri
| | - C E Norton
- Dept. of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri
| | - H J Kim
- Dept. of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri
| | - S K Sivasankaran
- Bioinformatics and Analytics Core, Division of Research, Innovation and Impact, University of Missouri, Columbia, Missouri
| | - M Li
- Dept. of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri
| | | | - B T Drumm
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, A91 K584, Ireland
| | - M J Davis
- Dept. of Medical Pharmacology & Physiology, University of Missouri, Columbia, Missouri
| |
Collapse
|
2
|
Sheng X, Zhang C, Zhao J, Xu J, Zhang P, Ding Q, Zhang J. Microvascular destabilization and intricated network of the cytokines in diabetic retinopathy: from the perspective of cellular and molecular components. Cell Biosci 2024; 14:85. [PMID: 38937783 PMCID: PMC11212265 DOI: 10.1186/s13578-024-01269-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
Microvascular destabilization is the primary cause of the inner blood-retinal barrier (iBRB) breakdown and increased vascular leakage in diabetic retinopathy (DR). Microvascular destabilization results from the combinational effects of increased levels of growth factors and cytokines, involvement of inflammation, and the changed cell-to-cell interactions, especially the loss of endothelial cells and pericytes, due to hyperglycemia and hypoxia. As the manifestation of microvascular destabilization, the fluid transports via paracellular and transcellular routes increase due to the disruption of endothelial intercellular junctional complexes and/or the altered caveolar transcellular transport across the retinal vascular endothelium. With diabetes progression, the functional and the structural changes of the iBRB components, including the cellular and noncellular components, further facilitate and aggravate microvascular destabilization, resulting in macular edema, the neuroretinal damage and the dysfunction of retinal inner neurovascular unit (iNVU). Although there have been considerable recent advances towards a better understanding of the complex cellular and molecular network underlying the microvascular destabilization, some still remain to be fully elucidated. Recent data indicate that targeting the intricate signaling pathways may allow to against the microvascular destabilization. Therefore, efforts have been made to better clarify the cellular and molecular mechanisms that are involved in the microvascular destabilization in DR. In this review, we discuss: (1) the brief introduction of DR and microvascular destabilization; (2) the cellular and molecular components of iBRB and iNVU, and the breakdown of iBRB; (3) the matrix and cell-to-cell contacts to maintain microvascular stabilization, including the endothelial glycocalyx, basement membrane, and various cell-cell interactions; (4) the molecular mechanisms mediated cell-cell contacts and vascular cell death; (5) the altered cytokines and signaling pathways as well as the intricate network of the cytokines involved in microvascular destabilization. This comprehensive review aimed to provide the insights for microvascular destabilization by targeting the key molecules or specific iBRB cells, thus restoring the function and structure of iBRB and iNVU, to treat DR.
Collapse
Affiliation(s)
- Xia Sheng
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Chunmei Zhang
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Jiwei Zhao
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Jianping Xu
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China.
| | - Peng Zhang
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China.
| | - Quanju Ding
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, National Clinical Research Center for Eye Diseases, Shanghai, China.
- The International Eye Research Institute of The Chinese University of Hong Kong (Shenzhen), Shenzhen, China.
- C-MER (Shenzhen) Dennis Lam Eye Hospital, Shenzhen, China.
- C-MER International Eye Care Group, C-MER Dennis Lam & Partners Eye Center, Hong Kong, China.
| |
Collapse
|
3
|
Ren R, Ding S, Ma K, Jiang Y, Wang Y, Chen J, Wang Y, Kou Y, Fan X, Zhu X, Qin L, Qiu C, Simons M, Wei X, Yu L. SUMOylation Fine-Tunes Endothelial HEY1 in the Regulation of Angiogenesis. Circ Res 2024; 134:203-222. [PMID: 38166414 PMCID: PMC10872267 DOI: 10.1161/circresaha.123.323398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/15/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND Angiogenesis, which plays a critical role in embryonic development and tissue repair, is controlled by a set of angiogenic signaling pathways. As a TF (transcription factor) belonging to the basic helix-loop-helix family, HEY (hairy/enhancer of split related with YRPW motif)-1 (YRPW motif, abbreviation of 4 highly conserved amino acids in the motif) has been identified as a key player in developmental angiogenesis. However, the precise mechanisms underlying HEY1's actions in angiogenesis remain largely unknown. Our previous studies have suggested a potential role for posttranslational SUMOylation in the dynamic regulation of vascular development and organization. METHODS Immunoprecipitation, mass spectrometry, and bioinformatics analysis were used to determine the biochemical characteristics of HEY1 SUMOylation. The promoter-binding capability of HEY1 was determined by chromatin immunoprecipitation, dual luciferase, and electrophoretic mobility shift assays. The dimerization pattern of HEY1 was determined by coimmunoprecipitation. The angiogenic capabilities of endothelial cells were assessed by CCK-8 (cell counting kit-8), 5-ethynyl-2-deoxyuridine staining, wound healing, transwell, and sprouting assays. Embryonic and postnatal vascular growth in mouse tissues, matrigel plug assay, cutaneous wound healing model, oxygen-induced retinopathy model, and tumor angiogenesis model were used to investigate the angiogenesis in vivo. RESULTS We identified intrinsic endothelial HEY1 SUMOylation at conserved lysines by TRIM28 (tripartite motif containing 28) as the unique E3 ligase. Functionally, SUMOylation facilitated HEY1-mediated suppression of angiogenic RTK (receptor tyrosine kinase) signaling and angiogenesis in primary human endothelial cells and mice with endothelial cell-specific expression of wild-type HEY1 or a SUMOylation-deficient HEY1 mutant. Mechanistically, SUMOylation facilitates HEY1 homodimer formation, which in turn preserves HEY1's DNA-binding capability via recognition of E-box promoter elements. Therefore, SUMOylation maintains HEY1's function as a repressive TF controlling numerous angiogenic genes, including RTKs and Notch pathway components. Proangiogenic stimuli induce HEY1 deSUMOylation, leading to heterodimerization of HEY1 with HES (hairy and enhancer of split)-1, which results in ineffective DNA binding and loss of HEY1's angiogenesis-suppressive activity. CONCLUSIONS Our findings demonstrate that reversible HEY1 SUMOylation is a molecular mechanism that coordinates endothelial angiogenic signaling and angiogenesis, both in physiological and pathological milieus, by fine-tuning the transcriptional activity of HEY1. Specifically, SUMOylation facilitates the formation of the HEY1 transcriptional complex and enhances its DNA-binding capability in endothelial cells.
Collapse
Affiliation(s)
- Ruizhe Ren
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Sha Ding
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Kefan Ma
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Yuanqing Jiang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Yiran Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Junbo Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Yunyun Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Yaohui Kou
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Xiao Fan
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaolong Zhu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Lingfeng Qin
- Department of Surgery, Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Cong Qiu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Xiyang Wei
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Luyang Yu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| |
Collapse
|
4
|
Zhang Y, Xu S, Jiang F, Hu M, Han Y, Wang Y, Liu Z. A comprehensive insight into the role of molecular pathways affected by the Angiopoietin and Tie system involved in hematological malignancies' pathogenesis. Pathol Res Pract 2023; 248:154677. [PMID: 37467636 DOI: 10.1016/j.prp.2023.154677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/15/2023] [Accepted: 07/04/2023] [Indexed: 07/21/2023]
Abstract
Angiogenesis has been recognized as a critical factor in developing solid tumors and hematological malignancies. How angiogenesis affects the molecular pathways in malignancies is still a mystery. The angiopoietin family, one of the known molecular mediators for angiogenesis, encourages angiogenesis by attaching to Tie receptors on cell surfaces. Angiopoietin, Tie, and particularly the molecular pathways they mediate have all been the subject of recent studies that have established their diagnostic, prognostic, and therapeutic potential. Here, we've reviewed the function of molecular pathways impacted by the Angiogenin and Tie system in hematological malignancies.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Shoufang Xu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Feiyu Jiang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Mengsi Hu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Yetao Han
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Yingjian Wang
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China
| | - Zhiwei Liu
- Department of Blood Transfusion, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Hangzhou, Zhejiang 310016, China.
| |
Collapse
|
5
|
Saikia Q, Reeve H, Alzahrani A, Critchley WR, Zeqiraj E, Divan A, Harrison MA, Ponnambalam S. VEGFR endocytosis: Implications for angiogenesis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 194:109-139. [PMID: 36631189 DOI: 10.1016/bs.pmbts.2022.06.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The binding of vascular endothelial growth factor (VEGF) superfamily to VEGF receptor tyrosine kinases (VEGFRs) and co-receptors regulates vasculogenesis, angiogenesis and lymphangiogenesis. A recurring theme is that dysfunction in VEGF signaling promotes pathological angiogenesis, an important feature of cancer and pro-inflammatory disease states. Endocytosis of basal (resting) or activated VEGFRs facilitates signal attenuation and endothelial quiescence. However, increasing evidence suggest that activated VEGFRs can continue to signal from intracellular compartments such as endosomes. In this chapter, we focus on the evolving link between VEGFR endocytosis, signaling and turnover and the implications for angiogenesis. There is much interest in how such understanding of VEGFR dynamics can be harnessed therapeutically for a wide range of human disease states.
Collapse
Affiliation(s)
- Queen Saikia
- School of Molecular & Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Hannah Reeve
- School of Molecular & Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Areej Alzahrani
- School of Molecular & Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - William R Critchley
- School of Molecular & Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Elton Zeqiraj
- School of Molecular & Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Aysha Divan
- School of Molecular & Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Michael A Harrison
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | | |
Collapse
|
6
|
Cholenic acid derivative UniPR1331 impairs tumor angiogenesis via blockade of VEGF/VEGFR2 in addition to Eph/ephrin. Cancer Gene Ther 2022; 29:908-917. [PMID: 34426652 PMCID: PMC9293752 DOI: 10.1038/s41417-021-00379-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/12/2021] [Accepted: 08/10/2021] [Indexed: 12/11/2022]
Abstract
Angiogenesis, the formation of new blood vessels from preexisting ones, is crucial for tumor growth and metastatization, and is considered a promising therapeutic target. Unfortunately, drugs directed against a specific proangiogenic growth factor or receptor turned out to be of limited benefit for oncology patients, likely due to the high biochemical redundancy of the neovascularization process. In this scenario, multitarget compounds that are able to simultaneously tackle different proangiogenic pathways are eagerly awaited. UniPR1331 is a 3β-hydroxy-Δ5-cholenic acid derivative, which is already known to inhibit Eph-ephrin interaction. Here, we employed an analysis pipeline consisting of molecular modeling and simulation, surface plasmon resonance spectrometry, biochemical assays, and endothelial cell models to demonstrate that UniPR1331 directly interacts with the vascular endothelial growth factor receptor 2 (VEGFR2) too. The binding of UniPR1331 to VEGFR2 prevents its interaction with the natural ligand vascular endothelial growth factor and subsequent autophosphorylation, signal transduction, and in vitro proangiogenic activation of endothelial cells. In vivo, UniPR1331 inhibits tumor cell-driven angiogenesis in zebrafish. Taken together, these data shed light on the pleiotropic pharmacological effect of UniPR1331, and point to Δ5-cholenic acid as a promising molecular scaffold for the development of multitarget antiangiogenic compounds.
Collapse
|
7
|
Uemura A, Fruttiger M, D'Amore PA, De Falco S, Joussen AM, Sennlaub F, Brunck LR, Johnson KT, Lambrou GN, Rittenhouse KD, Langmann T. VEGFR1 signaling in retinal angiogenesis and microinflammation. Prog Retin Eye Res 2021; 84:100954. [PMID: 33640465 PMCID: PMC8385046 DOI: 10.1016/j.preteyeres.2021.100954] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
Five vascular endothelial growth factor receptor (VEGFR) ligands (VEGF-A, -B, -C, -D, and placental growth factor [PlGF]) constitute the VEGF family. VEGF-A binds VEGF receptors 1 and 2 (VEGFR1/2), whereas VEGF-B and PlGF only bind VEGFR1. Although much research has been conducted on VEGFR2 to elucidate its key role in retinal diseases, recent efforts have shown the importance and involvement of VEGFR1 and its family of ligands in angiogenesis, vascular permeability, and microinflammatory cascades within the retina. Expression of VEGFR1 depends on the microenvironment, is differentially regulated under hypoxic and inflammatory conditions, and it has been detected in retinal and choroidal endothelial cells, pericytes, retinal and choroidal mononuclear phagocytes (including microglia), Müller cells, photoreceptor cells, and the retinal pigment epithelium. Whilst the VEGF-A decoy function of VEGFR1 is well established, consequences of its direct signaling are less clear. VEGFR1 activation can affect vascular permeability and induce macrophage and microglia production of proinflammatory and proangiogenic mediators. However the ability of the VEGFR1 ligands (VEGF-A, PlGF, and VEGF-B) to compete against each other for receptor binding and to heterodimerize complicates our understanding of the relative contribution of VEGFR1 signaling alone toward the pathologic processes seen in diabetic retinopathy, retinal vascular occlusions, retinopathy of prematurity, and age-related macular degeneration. Clinically, anti-VEGF drugs have proven transformational in these pathologies and their impact on modulation of VEGFR1 signaling is still an opportunity-rich field for further research.
Collapse
Affiliation(s)
- Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Marcus Fruttiger
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, 20 Staniford Street, Boston, MA, 02114, USA.
| | - Sandro De Falco
- Angiogenesis Laboratory, Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", Via Pietro Castellino 111, 80131 Naples, Italy; ANBITION S.r.l., Via Manzoni 1, 80123, Naples, Italy.
| | - Antonia M Joussen
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12200 Berlin, and Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.
| | - Lynne R Brunck
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Kristian T Johnson
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - George N Lambrou
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Kay D Rittenhouse
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 9, 50931, Cologne, Germany.
| |
Collapse
|
8
|
Cellular Crosstalk between Endothelial and Smooth Muscle Cells in Vascular Wall Remodeling. Int J Mol Sci 2021; 22:ijms22147284. [PMID: 34298897 PMCID: PMC8306829 DOI: 10.3390/ijms22147284] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/25/2021] [Accepted: 07/01/2021] [Indexed: 12/24/2022] Open
Abstract
Pathological vascular wall remodeling refers to the structural and functional changes of the vessel wall that occur in response to injury that eventually leads to cardiovascular disease (CVD). Vessel wall are composed of two major primary cells types, endothelial cells (EC) and vascular smooth muscle cells (VSMCs). The physiological communications between these two cell types (EC–VSMCs) are crucial in the development of the vasculature and in the homeostasis of mature vessels. Moreover, aberrant EC–VSMCs communication has been associated to the promotor of various disease states including vascular wall remodeling. Paracrine regulations by bioactive molecules, communication via direct contact (junctions) or information transfer via extracellular vesicles or extracellular matrix are main crosstalk mechanisms. Identification of the nature of this EC–VSMCs crosstalk may offer strategies to develop new insights for prevention and treatment of disease that curse with vascular remodeling. Here, we will review the molecular mechanisms underlying the interplay between EC and VSMCs. Additionally, we highlight the potential applicable methodologies of the co-culture systems to identify cellular and molecular mechanisms involved in pathological vascular wall remodeling, opening questions about the future research directions.
Collapse
|
9
|
Yang Y, Yu Y, Zhou R, Yang Y, Bu Y. The effect of combined exposure of zinc and nickel on the development of zebrafish. J Appl Toxicol 2021; 41:1765-1778. [PMID: 33645740 DOI: 10.1002/jat.4159] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 02/04/2021] [Accepted: 02/19/2021] [Indexed: 12/28/2022]
Abstract
Excessive accumulation of Zn2+ or Ni2+ can cause various problems to aquatic animals. In this study, the developmental toxicity induced by individual or combined exposure of Zn2+ and Ni2+ to zebrafish embryos and larvae were evaluated to better understand the interaction between Zn2+ and Ni2+ . Both of individual and combined exposure of Zn2+ and Ni2+ could cause obvious developmental toxicity, which mainly occurred after hatching, at a concentration-dependent manner. The calculated 168-h LC50 were 2.79 mg/L for Zn2+ and 7.44 mg/L for Ni2+ . The interaction of Zn2+ and Ni2+ based on mortality was found to be an antagonism. Various malformations, including tail curving, spinal curvature, pericardial edema, and yolk sac edema, were observed with significant effects on body length and heartbeat rates after exposure of Zn2+ and Ni2+ . Meanwhile, some genes related to cardiovascular development and bone formation were mainly down-regulated by the individual and combined exposure of Zn2+ and Ni2+ . The individual exposure was more toxic than combined exposure because the interaction of Zn2+ and Ni2+ was determined to be an antagonism. The down-regulation of genes related to cardiovascular development and bone formation may contribute to the observed malformation and decreases of body length and heartbeat rates.
Collapse
Affiliation(s)
- Yongmeng Yang
- School of Environmental and Safety Engineering, Changzhou University, Changzhou, China
- Nanjing Institute of Environmental Science, Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment, Nanjing, China
- Guangdong University of Technology, Synergy Innovation Institute of GDUT, Shantou, China
| | - Yue Yu
- Nanjing Institute of Environmental Science, Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment, Nanjing, China
| | - Rong Zhou
- Nanjing Institute of Environmental Science, Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment, Nanjing, China
| | - Yan Yang
- Guangdong University of Technology, Synergy Innovation Institute of GDUT, Shantou, China
- School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou, China
| | - Yuanqing Bu
- Nanjing Institute of Environmental Science, Key Laboratory of Pesticide Environmental Assessment and Pollution Control, Ministry of Ecology and Environment, Nanjing, China
| |
Collapse
|
10
|
Rowe DW, Hong SH, Zhang C, Shin DG, Adams DJ, Youngstrom DW, Chen L, Wu Z, Zhou Y, Maye P. Skeletal screening IMPC/KOMP using μCT and computer automated cryohistology: Application to the Efna4 KO mouse line. Bone 2021; 144:115688. [PMID: 33065355 DOI: 10.1016/j.bone.2020.115688] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 01/01/2023]
Abstract
The IMPC/KOMP program provides the opportunity to screen mice harboring well defined gene-inactivation mutations in a uniform genetic background. The program performs a global tissue phenotyping survey that includes skeletal x-rays and bone density measurements. Because of the relative insensitivity of the two screening tests for detecting variance in bone architecture, we initiated a secondary screen based on μCT and a cryohistolomorphological workflow that was performed on the femur and vertebral compartments on 220 randomly selected knockouts (KOs) and 36 control bone samples over a 2 1/2 year collection period provided by one of the production/phenotyping centers. The performance of the screening protocol was designed to balance throughput and cost versus sensitivity and informativeness such that the output would be of value to the skeletal biology community. Here we report the reliability of this screening protocol to establish criteria for control skeletal variance at the architectural, dynamic and cellular histomorphometric level. Unexpected properties of the control population include unusually high variance in BV/TV in male femurs and greater bone formation and bone turnover rates in the female femur and vertebral trabeculae bone compartments. However, the manner for maintaining bone formation differed between these two bone sites. The vertebral compartment relies on maintaining a greater number of bone forming surfaces while the femoral compartment utilized more matrix production per cell. The comparison of the architectural properties obtained by μCT and histomorphology revealed significant differences in values for BV/TV, Tb.Th and Tb.N which is attributable to sampling density of the two methods. However, as a screening tool, expressing the ratio of KO to the control line as obtained by either method was remarkably similar. It identified KOs with significant variance which led to a more detailed histological analysis. Our findings are exemplified by the Efna4 KO, in which a high BV/TV was identified by μCT and confirmed by histomorphometry in the femur but not in the vertebral compartment. Dynamic labeling showed a marked increase in BFR which was attributable to increased labeling surfaces. Cellular analysis confirmed partitioning of osteoblast to labeling surfaces and a marked decrease in osteoclastic activity on both labeling and quiescent surfaces. This pattern of increased bone modeling would not be expected based on prior studies of the Ephrin-Ephrin receptor signaling pathways between osteoblasts and osteoclasts. Overall, our findings underscore why unbiased screening is needed because it can reveal unknown or unanticipated genes that impact skeletal variation.
Collapse
Affiliation(s)
- David W Rowe
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America.
| | - Seung-Hyun Hong
- Computer Science & Engineering, School of Engineering, University of Connecticut, Storrs, CT 06269, United States of America
| | - Caibin Zhang
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America
| | - Dong-Guk Shin
- Computer Science & Engineering, School of Engineering, University of Connecticut, Storrs, CT 06269, United States of America
| | - Douglas J Adams
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States of America
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, School of Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America
| | - Li Chen
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America
| | - Zhihua Wu
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America
| | - Yueying Zhou
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America
| | - Peter Maye
- Regenerative Medicine and Skeletal Development, Department of Reconstructive Sciences, Biomaterials and Skeletal Development, School of Dental Medicine, University of Connecticut Health, Farmington, CT 06030, United States of America
| |
Collapse
|
11
|
Abstract
During multicellular organism development, complex structures are sculpted to form organs and tissues, which are maintained throughout adulthood. Many of these processes require cells to fuse with one another, or with themselves. These plasma membrane fusions merge endoplasmic cellular content across external, exoplasmic, space. In the nematode Caenorhabditis elegans, such cell fusions serve as a unique sculpting force, involved in the embryonic morphogenesis of the skin-like multinuclear hypodermal cells, but also in refining delicate structures, such as valve openings and the tip of the tail. During post-embryonic development, plasma membrane fusions continue to shape complex neuron structures and organs such as the vulva, while during adulthood fusion participates in cell and tissue repair. These processes rely on two fusion proteins (fusogens): EFF-1 and AFF-1, which are part of a broader family of structurally related membrane fusion proteins, encompassing sexual reproduction, viral infection, and tissue remodeling. The established capabilities of these exoplasmic fusogens are further expanded by new findings involving EFF-1 and AFF-1 in endocytic vesicle fission and phagosome sealing. Tight regulation by cell-autonomous and non-cell autonomous mechanisms orchestrates these diverse cell fusions at the correct place and time-these processes and their significance are discussed in this review.
Collapse
|
12
|
Yuan X, Bhat OM, Samidurai A, Das A, Zhang Y, Li PL. Reversal of Endothelial Extracellular Vesicle-Induced Smooth Muscle Phenotype Transition by Hypercholesterolemia Stimulation: Role of NLRP3 Inflammasome Activation. Front Cell Dev Biol 2020; 8:597423. [PMID: 33409276 PMCID: PMC7779768 DOI: 10.3389/fcell.2020.597423] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 12/04/2020] [Indexed: 01/18/2023] Open
Abstract
Recent studies reported that vascular endothelial cells (ECs) secrete NLR family pyrin domain-containing 3 (NLRP3) inflammasome products such as interleukin-1β (IL-1β) via extracellular vesicles (EVs) under various pathological conditions. EVs represent one of the critical mechanisms mediating the cell-to-cell communication between ECs and vascular smooth muscle cells (VSMCs). However, whether or not the inflammasome-dependent EVs directly participate in the regulation of VSMC function remains unknown. In the present study, we found that in cultured carotid ECs, atherogenic stimulation by oxysterol 7-ketocholesterol (7-Ket) induced NLRP3 inflammasome formation and activation, reduced lysosome-multivesicular bodies (MVBs) fusion, and increased secretion of EVs that contain inflammasome product IL-1β. These EC-derived IL-1β-containing EVs promoted synthetic phenotype transition of co-cultured VSMCs, whereas EVs from unstimulated ECs have the opposite effects. Moreover, acid ceramidase (AC) deficiency or lysosome inhibition further exaggerated the 7-Ket-induced release of IL-1β-containing EVs in ECs. Using a Western diet (WD)-induced hypercholesterolemia mouse model, we found that endothelial-specific AC gene knockout mice (Asah1fl/fl/ECCre) exhibited augmented WD-induced EV secretion with IL-1β and more significantly decreased the interaction of MVBs with lysosomes in the carotid arterial wall compared to their wild-type littermates (WT/WT). The endothelial AC deficiency in Asah1fl/fl/ECCre mice also resulted in enhanced VSMC phenotype transition and accelerated neointima formation. Together, these results suggest that NLRP3 inflammasome-dependent IL-1β production during hypercholesterolemia promotes VSMC phenotype transition to synthetic status via EV machinery, which is controlled by lysosomal AC activity. Our findings provide novel mechanistic insights into understanding the pathogenic role of endothelial NLRP3 inflammasome in vascular injury through EV-mediated EC-to-VSMC regulation.
Collapse
Affiliation(s)
- Xinxu Yuan
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Owais M Bhat
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Arun Samidurai
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Anindita Das
- Pauley Heart Center, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Yang Zhang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| |
Collapse
|
13
|
Chen Z, Morales JE, Avci N, Guerrero PA, Rao G, Seo JH, McCarty JH. The vascular endothelial cell-expressed prion protein doppel promotes angiogenesis and blood-brain barrier development. Development 2020; 147:dev.193094. [PMID: 32895288 DOI: 10.1242/dev.193094] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/17/2020] [Indexed: 12/14/2022]
Abstract
The central nervous system (CNS) contains a complex network of blood vessels that promote normal tissue development and physiology. Abnormal control of blood vessel morphogenesis and maturation is linked to the pathogenesis of various neurodevelopmental diseases. The CNS-specific genes that regulate blood vessel morphogenesis in development and disease remain largely unknown. Here, we have characterized functions for the gene encoding prion protein 2 (Prnd) in CNS blood vessel development and physiology. Prnd encodes the glycosylphosphatidylinositol (GPI)-linked protein doppel, which is expressed on the surface of angiogenic vascular endothelial cells, but is absent in quiescent endothelial cells of the adult CNS. During CNS vascular development, doppel interacts with receptor tyrosine kinases and activates cytoplasmic signaling pathways involved in endothelial cell survival, metabolism and migration. Analysis of mice genetically null for Prnd revealed impaired CNS blood vessel morphogenesis and associated endothelial cell sprouting defects. Prnd-/- mice also displayed defects in endothelial barrier integrity. Collectively, these data reveal novel mechanisms underlying doppel control of angiogenesis in the developing CNS, and may provide new insights about dysfunctional pathways that cause vascular-related CNS disorders.
Collapse
Affiliation(s)
- Zhihua Chen
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - John E Morales
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Naze Avci
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Paola A Guerrero
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Ganesh Rao
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | - Je Hoon Seo
- Department of Anatomy, Chungbuk National University College of Medicine, Chungbuk 28644, Republic of Korea
| | - Joseph H McCarty
- Department of Neurosurgery, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
14
|
Coucha M, Barrett AC, Bailey J, Abdelghani M, Abdelsaid M. Increased Ephrin-B2 expression in pericytes contributes to retinal vascular death in rodents. Vascul Pharmacol 2020; 131:106761. [PMID: 32585189 DOI: 10.1016/j.vph.2020.106761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/29/2022]
Abstract
AIMS Diabetes-induced retinal vascular cell death aggravates diabetic retinopathy (DR) to the proliferative stage and blindness. Pericytes play a crucial role in retinal capillaries survival, stability, and angiogenesis. Ephrin-B2 is a tyrosine kinase that regulates pericytes/endothelial cells communication during angiogenesis; yet, its role in DR is still unclear. We hypothesize that diabetes increases Ephrin-B2 signaling in pericytes, which contributes to inflammation and retinal vascular cell death. METHODS Selective inhibition of the Ephrin-B2 expression in the retinal pericytes was achieved using an intraocular injection of adeno-associated virus (AAV) with a specific pericyte promotor. Vascular death was determined by retinal trypsin digest. Pathological angiogenesis was assessed using the oxygen-induced retinopathy model in pericyte-Ephrin-B2 knockout mice, wild type, and wild type injected with AAV. Angiogenic properties, inflammatory, and apoptotic markers were measured in human retinal pericytes (HRP) grown under diabetic conditions. KEY FINDING Diabetes significantly increased the expression of Ephrin-B2, inflammatory, and apoptotic markers in the diabetic retinas and HRP. These effects were prevented by silencing Ephrin-B2 in HRP. Moreover, Ephrin-B2 silencing in retinal pericytes decreased pathological angiogenesis and acellular capillaries formation in diabetic retinas. SIGNIFICANCE Increased Ephrin-B2 expression in the pericytes contributed to diabetes-induced retinal inflammation and vascular death. These results identify pericytes-Ephrin-B2 as a therapeutic target for DR.
Collapse
Affiliation(s)
- Maha Coucha
- Department of Pharmaceutical Sciences, School of Pharmacy, South University, Savannah, GA, USA
| | - Amy C Barrett
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA
| | - Joseph Bailey
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA
| | - Maryam Abdelghani
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA
| | - Mohammed Abdelsaid
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA.
| |
Collapse
|
15
|
Arteriogenesis of the Spinal Cord-The Network Challenge. Cells 2020; 9:cells9020501. [PMID: 32098337 PMCID: PMC7072838 DOI: 10.3390/cells9020501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 12/27/2022] Open
Abstract
Spinal cord ischemia (SCI) is a clinical complication following aortic repair that significantly impairs the quality and expectancy of life. Despite some strategies, like cerebrospinal fluid drainage, the occurrence of neurological symptoms, such as paraplegia and paraparesis, remains unpredictable. Beside the major blood supply through conduit arteries, a huge collateral network protects the central nervous system from ischemia—the paraspinous and the intraspinal compartment. The intraspinal arcades maintain perfusion pressure following a sudden inflow interruption, whereas the paraspinal system first needs to undergo arteriogenesis to ensure sufficient blood supply after an acute ischemic insult. The so-called steal phenomenon can even worsen the postoperative situation by causing the hypoperfusion of the spine when, shortly after thoracoabdominal aortic aneurysm (TAAA) surgery, muscles connected with the network divert blood and cause additional stress. Vessels are a conglomeration of different cell types involved in adapting to stress, like endothelial cells, smooth muscle cells, and pericytes. This adaption to stress is subdivided in three phases—initiation, growth, and the maturation phase. In fields of endovascular aortic aneurysm repair, pre-operative selective segmental artery occlusion may enable the development of a sufficient collateral network by stimulating collateral vessel growth, which, again, may prevent spinal cord ischemia. Among others, the major signaling pathways include the phosphoinositide 3 kinase (PI3K) pathway/the antiapoptotic kinase (AKT) pathway/the endothelial nitric oxide synthase (eNOS) pathway, the Erk1, the delta-like ligand (DII), the jagged (Jag)/NOTCH pathway, and the midkine regulatory cytokine signaling pathways.
Collapse
|
16
|
Zhou C, Qian X, Hu M, Zhang R, Liu N, Huang Y, Yang J, Zhang J, Bai H, Yang Y, Wang Y, Ali D, Michalak M, Chen XZ, Tang J. STYK1 promotes autophagy through enhancing the assembly of autophagy-specific class III phosphatidylinositol 3-kinase complex I. Autophagy 2019; 16:1786-1806. [PMID: 31696776 DOI: 10.1080/15548627.2019.1687212] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Macroautophagy/autophagy plays key roles in development, oncogenesis, and cardiovascular and metabolic diseases. Autophagy-specific class III phosphatidylinositol 3-kinase complex I (PtdIns3K-C1) is essential for autophagosome formation. However, the regulation of this complex formation requires further investigation. Here, we discovered that STYK1 (serine/threonine/tyrosine kinase 1), a member of the receptor tyrosine kinases (RTKs) family, is a new upstream regulator of autophagy. We discovered that STYK1 facilitated autophagosome formation in human cells and zebrafish, which was characterized by elevated LC3-II and lowered SQSTM1/p62 levels and increased puncta formation by several marker proteins, such as ATG14, WIPI1, and ZFYVE1. Moreover, we observed that STYK1 directly binds to the PtdIns3K-C1 complex as a homodimer. The binding with this complex was promoted by Tyr191 phosphorylation, by means of which the kinase activity of STYK1 was elevated. We also demonstrated that STYK1 elevated the serine phosphorylation of BECN1, thereby decreasing the interaction between BECN1 and BCL2. Furthermore, we found that STYK1 preferentially facilitated the assembly of the PtdIns3K-C1 complex and was required for PtdIns3K-C1 complex kinase activity. Taken together, our findings provide new insights into autophagy induction and reveal evidence of novel crosstalk between the components of RTK signaling and autophagy. Abbreviations: AICAR: 5-aminoimidazole-4-carboxamide ribonucleotide; AMPK: adenosine 5'-monophosphate (AMP)-activated protein kinase; ATG: autophagy related; ATP: adenosine triphosphate; BCL2: BCL2 apoptosis regulator; BECN1: beclin 1; Bre A: brefeldin A; Co-IP: co-immunoprecipitation; CRISPR: clustered regularly interspaced short palindromic repeats; DAPI: 4',6-diamidino-2-phenylindole; EBSS: Earle's balanced salt solution; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; GSEA: gene set enrichment analysis; MAP1LC3/LC3, microtubule associated protein 1 light chain 3; MAPK8/JNK1: mitogen-activated protein kinase 8; mRFP: monomeric red fluorescent protein; MTOR: mechanistic target of rapamycin kinase; MTT: 3-(4,5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide; PIK3C3: phosphatidylinositol 3-kinase catalytic subunit type 3; PIK3R4: phosphoinositide-3-kinase regulatory subunit 4; qRT-PCR: quantitative reverse transcription PCR; RACK1: receptor for activated C kinase 1; RUBCN: rubicon autophagy regulator; siRNA: small interfering RNA; SQSTM1: sequestosome 1; STYK1/NOK: serine/threonine/tyrosine kinase 1; TCGA: The Cancer Genome Atlas; Ub: ubiquitin; ULK1: unc-51 like autophagy activating kinase 1; UVRAG: UV radiation resistance associated; WIPI1: WD repeat domain, phosphoinositide interacting 1; ZFYVE1: zinc finger FYVE-type containing 1.
Collapse
Affiliation(s)
- Cefan Zhou
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology , Wuhan, China.,The State Key Laboratory of Virology, College of Life Sciences, Wuhan University , Wuhan, Hubei, China
| | - Xuehong Qian
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology , Wuhan, China
| | - Miao Hu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology , Wuhan, China
| | - Rui Zhang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology , Wuhan, China
| | - Nanxi Liu
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology , Wuhan, China
| | - Yuan Huang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology , Wuhan, China
| | - Jing Yang
- Department of Biological Sciences, Boler-Parseghian Center for Rare and Neglected Diseases, Harper Cancer Research Institute, University of Notre Dame , Notre Dame, IN, USA
| | - Juan Zhang
- Department of Gastroenterology, First Affiliated Hospital of Xi`an Jiaotong University , Xi`an, Shanxi, China
| | - Hua Bai
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology , Wuhan, China
| | - Yuyan Yang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology , Wuhan, China
| | - Yefu Wang
- The State Key Laboratory of Virology, College of Life Sciences, Wuhan University , Wuhan, Hubei, China
| | - Declan Ali
- Department of Biological Sciences, University of Alberta , Edmonton, Alberta, Canada
| | - Marek Michalak
- Department of Biochemistry, University of Alberta , Edmonton, Alberta, Canada
| | - Xing-Zhen Chen
- Membrane Protein Disease Research Group, Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta , Edmonton, AB, Canada
| | - Jingfeng Tang
- National "111" Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology , Wuhan, China
| |
Collapse
|
17
|
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: 16] [Impact Index Per Article: 3.2] [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
|
18
|
Drexler HCA, Vockel M, Polaschegg C, Frye M, Peters K, Vestweber D. Vascular Endothelial Receptor Tyrosine Phosphatase: Identification of Novel Substrates Related to Junctions and a Ternary Complex with EPHB4 and TIE2. Mol Cell Proteomics 2019; 18:2058-2077. [PMID: 31427368 PMCID: PMC6773558 DOI: 10.1074/mcp.ra119.001716] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Indexed: 12/30/2022] Open
Abstract
Vascular endothelial protein tyrosine phosphatase (VE-PTP, PTPRB) is a receptor type phosphatase that is crucial for the regulation of endothelial junctions and blood vessel development. We and others have shown recently that VE-PTP regulates vascular integrity by dephosphorylating substrates that are key players in endothelial junction stability, such as the angiopoietin receptor TIE2, the endothelial adherens junction protein VE-cadherin and the vascular endothelial growth factor receptor VEGFR2. Here, we have systematically searched for novel substrates of VE-PTP in endothelial cells by utilizing two approaches. First, we studied changes in the endothelial phosphoproteome on exposing cells to a highly VE-PTP-specific phosphatase inhibitor followed by affinity isolation and mass-spectrometric analysis of phosphorylated proteins by phosphotyrosine-specific antibodies. Second, we used a substrate trapping mutant of VE-PTP to pull down phosphorylated substrates in combination with SILAC-based quantitative mass spectrometry measurements. We identified a set of substrate candidates of VE-PTP, of which a remarkably large fraction (29%) is related to cell junctions. Several of those were found in both screens and displayed very high connectivity in predicted functional interaction networks. The receptor protein tyrosine kinase EPHB4 was the most prominently phosphorylated protein on VE-PTP inhibition among those VE-PTP targets that were identified by both proteomic approaches. Further analysis revealed that EPHB4 forms a ternary complex with VE-PTP and TIE2 in endothelial cells. VE-PTP controls the phosphorylation of each of these two tyrosine kinase receptors. Despite their simultaneous presence in a ternary complex, stimulating each of the receptors with their own specific ligand did not cross-activate the respective partner receptor. Our systematic approach has led to the identification of novel substrates of VE-PTP, of which many are relevant for the control of cellular junctions further promoting the importance of VE-PTP as a key player of junctional signaling.
Collapse
Affiliation(s)
- Hannes C A Drexler
- Bioanalytical Mass Spectrometry, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149 Münster, Germany.
| | - Matthias Vockel
- Department of Vascular Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149 Münster, Germany; Institute of Human Genetics, University Hospital Münster, 48149 Münster, Germany
| | - Christian Polaschegg
- Department of Vascular Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149 Münster, Germany
| | - Maike Frye
- Department of Vascular Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149 Münster, Germany
| | | | - Dietmar Vestweber
- Department of Vascular Biology, Max Planck Institute for Molecular Biomedicine, Röntgenstr. 20, 48149 Münster, Germany.
| |
Collapse
|
19
|
Kyuno D, Zhao K, Schnölzer M, Provaznik J, Hackert T, Zöller M. Claudin7-dependent exosome-promoted reprogramming of nonmetastasizing tumor cells. Int J Cancer 2019; 145:2182-2200. [PMID: 30945750 DOI: 10.1002/ijc.32312] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 03/10/2019] [Accepted: 03/27/2019] [Indexed: 12/17/2022]
Abstract
Claudin7 (cld7) is a cancer-initiating cell (CIC) marker in gastrointestinal tumors, a cld7-knockdown (kd) being accompanied by loss of tumor progression. Tumor exosomes (TEX) restoring CIC activities, we explored the contribution of cld7. This became particularly interesting, as tight junction (TJ)- and glycolipid-enriched membrane domain (GEM)-derived cld7 is recruited into distinct TEX. TEXs were derived from CIC or cld7kd cells of a rat pancreatic and a human colon cancer line. TEX derived from pancreatic cancer cld7kd cells rescued with palmitoylation site-deficient cld7 (cld7mP) allowed selectively evaluating the contribution of GEM-derived TEX, only palmitoylated cld7 being integrated into GEM. Cld7 CIC-TEX promoted tumor cell dissemination and metastatic growth without a major impact on proliferation, apoptosis resistance and epithelial-mesenchymal transition. Instead, migration, invasion and (lymph)angiogenesis were strongly supported, only migration being selectively fostered by GEM-derived cld7 TEX. CIC-TEX coculture of cld7kd cells uncovered significant changes in the cld7kd cell protein and miRNA profiles. However, changes did not correspond to the CIC-TEX profile, CIC-TEX rather initiating integrin, protease and RTK, particularly lymphangiogenic receptor activation. CIC-TEX preferentially rescuing cld7kd-associated defects in signal transduction was backed up by an RTK inhibitor neutralizing the impact of CIC-TEX on tumor progression. In conclusion, cld7 contributes to selective steps of the metastatic cascade. Defects of cld7kd and cld7mP cells in migration, invasion and (lymph)angiogenesis are effaced by CIC-TEX that act by signaling cascade activation. Accordingly, RTK inhibitors are an efficient therapeutic defeating CIC-TEX.
Collapse
Affiliation(s)
- Daisuke Kyuno
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany.,Department of Surgery, Surgical Oncology and Science, Sapporo Medical University, Sapporo, Japan
| | - Kun Zhao
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Martina Schnölzer
- Functional Proteome Analysis, German Cancer Research Center, Heidelberg, Germany
| | | | - Thilo Hackert
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Margot Zöller
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
20
|
Coucha M, Barrett AC, Elgebaly M, Ergul A, Abdelsaid M. Inhibition of Ephrin-B2 in brain pericytes decreases cerebral pathological neovascularization in diabetic rats. PLoS One 2019; 14:e0210523. [PMID: 30620753 PMCID: PMC6324788 DOI: 10.1371/journal.pone.0210523] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 12/22/2018] [Indexed: 12/20/2022] Open
Abstract
We have previously shown that diabetes causes dysfunctional cerebral neovascularization that increases the risk for cerebrovascular disorders such as stroke and cognitive impairment. Pericytes (PCs) play a pivotal role in the angiogenic process through their interaction with the endothelial cells (EC). Yet, the role of PCs in dysfunctional cerebral neovascularization in diabetes is unclear. In the present study, we tested the hypothesis that the increased proangiogenic Ephrin-B2 signaling in PCs contributes to the dysfunctional cerebral neovascularization in diabetes. Type-II diabetes was induced by a combination of high fat diet and low dose streptozotocin injection in male Wistar rats. Selective in vivo Ephrin-B2 silencing in brain PCs was achieved using the stereotactic injection of adeno-associated virus (AAV) with NG2-promoter that expresses Ephrin-B2 shRNA. Neovascularization was assessed using vascular fluorescent dye stain. Novel object recognition (NOR) test was used to determine cognitive functions. Human brain microvascular pericytes HBMVPCs were grown in high glucose 25 mM and palmitate 200 uM (HG/Pal) to mimic diabetic conditions. Scratch migration and tube formation assays were conducted to evaluate PC/EC interaction and angiogenic functions in PC/EC co-culture. Diabetes increased the expression of Ephrin-B2 in the cerebrovasculature and pericytes. Concomitant increases in cerebral neovascularization parameters including vascular density, tortuosity and branching density in diabetic rats were accompanied by deterioration of cognitive function. Inhibition of Ephrin-B2 expression in PCs significantly restored cerebral vascularization and improved cognitive functions. HG/Pal increased PC/EC angiogenic properties in co-culture. Silencing Ephrin-B2 in PCs significantly reduced PC migration and PC/EC co-culture angiogenic properties. This study emphasizes the significant contribution of PCs to the pathological neovascularization in diabetes. Our findings introduce Ephrin-B2 signaling as a promising therapeutic target to improve cerebrovascular integrity in diabetes.
Collapse
Affiliation(s)
- Maha Coucha
- Department of Pharmaceutical Sciences, School of Pharmacy, South University, Savannah, Georgia, United States of America
| | - Amy C. Barrett
- Biomedical Sciences Department, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| | - Mostafa Elgebaly
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, Florida, United States of America
| | - Adviye Ergul
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, Georgia, United States of America
| | - Mohammed Abdelsaid
- Biomedical Sciences Department, School of Medicine, Mercer University, Savannah, Georgia, United States of America
| |
Collapse
|
21
|
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: 40] [Impact Index Per Article: 8.0] [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
|
22
|
Zhao K, Wang Z, Hackert T, Pitzer C, Zöller M. Tspan8 and Tspan8/CD151 knockout mice unravel the contribution of tumor and host exosomes to tumor progression. J Exp Clin Cancer Res 2018; 37:312. [PMID: 30541597 PMCID: PMC6292129 DOI: 10.1186/s13046-018-0961-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/14/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The tetraspanins Tspan8 and CD151 promote metastasis, exosomes (Exo) being suggested to be important in the crosstalk between tumor and host. The contribution of Tspan8 and CD151 to host versus tumor-derived exosome (TEX) activities being not defined, we approached the questions using 3-methylcholanthrene-induced (MCA) tumors from wt, Tspan8ko, CD151ko and Tspan8/CD151 (db)ko mice, implanted into tetraspanin-competent and deficient hosts. METHODS Tumor growth and dissemination, hematopoiesis and angiogenesis were surveyed in wild type (wt), Tspan8ko, CD151ko and dbko mice bearing tetraspanin-competent and -deficient MCA tumors. In vitro studies using tumor cells, bone marrow cells (BMC) and endothelial cells (EC) elaborated the mechanism of serum (s)Exo- and TEX-induced target modulation. RESULTS Tumors grew in autochthonous and syngeneic hosts differing in Tspan8- and/or CD151-competence. However, Tspan8ko- and/or CD151ko-tumor cell dissemination and settlement in metastatic organs was significantly reduced in the autochthonous host, and less severely in the wt-host. Impaired wt-MCA tumor dissemination in the ko-host confirmed a contribution of host- and tumor-Tspan8/-CD151 to tumor cell dissemination, delivery of sExo and TEX being severely impaired by a Tspan8ko/CD151ko. Coculturing tumor cells, BMC and EC with sExo and TEX revealed minor defects in epithelial mesenchymal transition and apoptosis resistance of ko tumors. Strongly reduced migratory and invasive capacity of Tspan8ko/CD151ko-MCA relies on distorted associations with integrins and CAM and missing Tspan8/CD151-promoted recruitment of proteases. The defects, differing between Tspan8ko- and CD151ko-MCA, were rescued by wt-TEX and, less efficiently Tspan8ko- and CD151ko-TEX. Minor defects in hematopoietic progenitor maturation were based on the missing association of hematopoietic growth factors /- receptors with CD151 and, less pronounced, Tspan8. Rescue of impaired angiogenesis in ko mice by wt-sExo and promotion of angiogenesis by TEX depended on the association of Tspan8 and CD151 with GPCR and RTK in EC and tumor cells. CONCLUSIONS Tspan8-/CD151-TEX play central roles in tumor progression. Tspan8-/CD151-sExo and TEX contribute by stimulating angiogenesis. Tspan8 and CD151 fulfill these tasks by associating with function-relevant proteins, the additive impact of Tspan8 and CD151 relying on differences in preferred associations. The distinct Tspan8 and CD151 contributions suggest a blockade of TEX-Tspan8 and -CD151 promising for therapeutic intervention.
Collapse
Affiliation(s)
- Kun Zhao
- Pancreas Section, University Hospital of Surgery, Ruprecht-Karls-University, Heidelberg, Germany
| | - Zhe Wang
- Pancreas Section, University Hospital of Surgery, Ruprecht-Karls-University, Heidelberg, Germany
- Present Address: Department of Oncology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong, China
| | - Thilo Hackert
- Pancreas Section, University Hospital of Surgery, Ruprecht-Karls-University, Heidelberg, Germany
| | - Claudia Pitzer
- Interdisciplinary Neurobehavioral Core, Institute of Pharmacology, Ruprecht-Karls-University, Heidelberg, Germany
| | - Margot Zöller
- Pancreas Section, University Hospital of Surgery, Ruprecht-Karls-University, Heidelberg, Germany
| |
Collapse
|
23
|
Li M, Qian M, Kyler K, Xu J. Endothelial-Vascular Smooth Muscle Cells Interactions in Atherosclerosis. Front Cardiovasc Med 2018; 5:151. [PMID: 30406116 PMCID: PMC6207093 DOI: 10.3389/fcvm.2018.00151] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 10/04/2018] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammatory process that can eventually lead to cardiovascular disease (CVD). Despite available treatment, the prevalence of atherosclerotic CVD, which has become the leading cause of death worldwide, persists. Identification of new mechanisms of atherogenesis are highly needed in order to develop an effective therapeutic treatment. The blood vessels contain two primary major cell types: endothelial cells (EC) and vascular smooth muscle cells (VSMC). Each of these performs an essential function in sustaining vascular homeostasis. EC-VSMC communication is essential not only to development, but also to the homeostasis of mature blood vessels. Aberrant EC-VSMC interaction could promote atherogenesis. Identification of the mode of EC-VSMC crosstalk that regulates vascular functionality and sustains homeostasis may offer strategic insights for prevention and treatment of atherosclerotic CVD. Here we will review the molecular mechanisms underlying the interplay between EC and VSMC that could contribute to atherosclerosis. We also highlight open questions for future research directions.
Collapse
Affiliation(s)
- Manna Li
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Ming Qian
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Kathy Kyler
- Office of Research Administration, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| | - Jian Xu
- Department of Medicine, Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma, OK, United States
| |
Collapse
|
24
|
Malik VA, Di Benedetto B. The Blood-Brain Barrier and the EphR/Ephrin System: Perspectives on a Link Between Neurovascular and Neuropsychiatric Disorders. Front Mol Neurosci 2018; 11:127. [PMID: 29706868 PMCID: PMC5906525 DOI: 10.3389/fnmol.2018.00127] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 03/29/2018] [Indexed: 01/06/2023] Open
Abstract
Interactions among endothelial cells (EC) forming blood vessels and their surrounding cell types are essential to establish the blood-brain barrier (BBB), an integral part of the neurovascular unit (NVU). Research on the NVU has recently seen a renaissance to especially understand the neurobiology of vascular and brain pathologies and their frequently occurring comorbidities. Diverse signaling molecules activated in the near proximity of blood vessels trigger paracellular pathways which regulate the formation and stabilization of tight junctions (TJ) between EC and thereby influence BBB permeability. Among regulatory molecules, the erythropoietin-producing-hepatocellular carcinoma receptors (EphR) and their Eph receptor-interacting signals (ephrins) play a pivotal role in EC differentiation, angiogenesis and BBB integrity. Multiple EphR-ligand interactions between EC and other cell types influence different aspects of angiogenesis and BBB formation. Such interactions additionally control BBB sealing properties and thus the penetration of substances into the brain parenchyma. Thus, they play critical roles in the healthy brain and during the pathogenesis of brain disorders. In this mini-review article, we aim at integrating the constantly growing literature about the functional roles of the EphR/ephrin system for the development of the vascular system and the BBB and in the pathogenesis of neurovascular and neuropsychiatric disorders. We suggest the hypothesis that a disrupted EphR/ephrin signaling at the BBB might represent an underappreciated molecular hub of disease comorbidity. Finally, we propose the possibility that the EphR/ephrin system bears the potential of becoming a novel target for the development of alternative therapeutic treatments, focusing on such comorbidities.
Collapse
Affiliation(s)
- Victoria A Malik
- RG Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Barbara Di Benedetto
- RG Neuro-Glia Pharmacology, Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany.,Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany
| |
Collapse
|
25
|
Abstract
The zebrafish is an outstanding model for studying vascular biology in vivo. Pericytes and vascular smooth muscle cells can be imaged as they associate with vessels and provide stability and integrity to the vasculature. In zebrafish, pericytes associate with the cerebral and trunk vasculature on the second day of development, as assayed by pdgfrβ and notch3 markers. In the head, cerebral pericytes are neural crest derived, except for the pericytes of the hindbrain vasculature, which are mesoderm derived. Similar to the hindbrain, pericytes on the trunk vasculature are also mesoderm derived. Regardless of their location, pericyte development depends on a complex interaction between blood flow and signalling pathways, such as Notch, SONIC HEDGEHOG and BMP signalling, all of which positively regulate pericyte numbers.Pericyte numbers rapidly increase as development proceeds in order to stabilize both the blood-brain barrier and the vasculature and hence, prevent haemorrhage. Consequently, compromised pericyte development results in compromised vascular integrity, which then evolves into detrimental pathologies. Some of these pathologies have been modelled in zebrafish by inducing mutations in the notch3, foxc1 and foxf2 genes. These zebrafish models provide insights into the mechanisms of disease as associated with pericyte biology. Going forward, these models may be key contributors in elucidating the role of vascular mural cells in regulating vessel diameter and hence, blood flow.
Collapse
Affiliation(s)
- Nabila Bahrami
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Sarah J Childs
- Department of Biochemistry and Molecular Biology, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
26
|
Norrin-induced Frizzled4 endocytosis and endo-lysosomal trafficking control retinal angiogenesis and barrier function. Nat Commun 2017; 8:16050. [PMID: 28675177 PMCID: PMC5500887 DOI: 10.1038/ncomms16050] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 05/17/2017] [Indexed: 02/06/2023] Open
Abstract
Angiogenesis and blood–brain barrier formation are required for normal central nervous system (CNS) function. Both processes are controlled by Wnt or Norrin (NDP) ligands, Frizzled (FZD) receptors, and β-catenin-dependent signalling in vascular endothelial cells. In the retina, FZD4 and the ligand NDP are critical mediators of signalling and are mutated in familial exudative vitreoretinopathy. Here, we report that NDP is a potent trigger of FZD4 ubiquitination and induces internalization of the NDP receptor complex into the endo-lysosomal compartment. Inhibition of ubiquitinated cargo transport through the multivesicular body (MVB) pathway using a dominant negative ESCRT (endosomal sorting complexes required for transport) component VPS4 EQ strongly impairs NDP/FZD4 signalling in vitro and recapitulates CNS angiogenesis and blood-CNS-barrier defects caused by impaired vascular β-catenin signalling in mice. These findings provide evidence for an important role of FZD4 endocytosis in NDP/FZD4 signalling and in CNS vascular biology and disease. Multiple mechanisms regulate Wnt/ß-catenin signalling. Zhang et al. describe a novel regulatory pathway and show that the activator of canonical Wnt signalling, Norrin, triggers endocytosis of its receptor Frizzled4 by promoting Frizzled4 ubiquitination.
Collapse
|
27
|
Hwangbo C, Lee HW, Kang H, Ju H, Wiley DS, Papangeli I, Han J, Kim JD, Dunworth WP, Hu X, Lee S, El-Hely O, Sofer A, Pak B, Peterson L, Comhair S, Hwang EM, Park JY, Thomas JL, Bautch VL, Erzurum SC, Chun HJ, Jin SW. Modulation of Endothelial Bone Morphogenetic Protein Receptor Type 2 Activity by Vascular Endothelial Growth Factor Receptor 3 in Pulmonary Arterial Hypertension. Circulation 2017; 135:2288-2298. [PMID: 28356442 DOI: 10.1161/circulationaha.116.025390] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 03/17/2017] [Indexed: 12/21/2022]
Abstract
BACKGROUND Bone morphogenetic protein (BMP) signaling has multiple roles in the development and function of the blood vessels. In humans, mutations in BMP receptor type 2 (BMPR2), a key component of BMP signaling, have been identified in the majority of patients with familial pulmonary arterial hypertension (PAH). However, only a small subset of individuals with BMPR2 mutation develops PAH, suggesting that additional modifiers of BMPR2 function play an important role in the onset and progression of PAH. METHODS We used a combination of studies in zebrafish embryos and genetically engineered mice lacking endothelial expression of Vegfr3 to determine the interaction between vascular endothelial growth factor receptor 3 (VEGFR3) and BMPR2. Additional in vitro studies were performed by using human endothelial cells, including primary lung endothelial cells from subjects with PAH. RESULTS Attenuation of Vegfr3 in zebrafish embryos abrogated Bmp2b-induced ectopic angiogenesis. Endothelial cells with disrupted VEGFR3 expression failed to respond to exogenous BMP stimulation. Mechanistically, VEGFR3 is physically associated with BMPR2 and facilitates ligand-induced endocytosis of BMPR2 to promote phosphorylation of SMADs and transcription of ID genes. Conditional, endothelial-specific deletion of Vegfr3 in mice resulted in impaired BMP signaling responses, and significantly worsened hypoxia-induced pulmonary hypertension. Consistent with these data, we found significant decrease in VEGFR3 expression in pulmonary arterial endothelial cells from human PAH subjects, and reconstitution of VEGFR3 expression in PAH pulmonary arterial endothelial cells restored BMP signaling responses. CONCLUSIONS Our findings identify VEGFR3 as a key regulator of endothelial BMPR2 signaling and a potential determinant of PAH penetrance in humans.
Collapse
Affiliation(s)
- Cheol Hwangbo
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Heon-Woo Lee
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Hyeseon Kang
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Hyekyung Ju
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - David S Wiley
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Irinna Papangeli
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Jinah Han
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Jun-Dae Kim
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - William P Dunworth
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Xiaoyue Hu
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Seyoung Lee
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Omar El-Hely
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Avraham Sofer
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Boryeong Pak
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Laura Peterson
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Suzy Comhair
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Eun Mi Hwang
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Jae-Yong Park
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Jean-Leon Thomas
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Victoria L Bautch
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Serpil C Erzurum
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.)
| | - Hyung J Chun
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.).
| | - Suk-Won Jin
- From Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (C.H., H.-W.L., H.K., H.J., I.P., J.H., J.-D.K., W.P.D., X.H., S.L., O.E.-H., A.S., H.J.C., S.-W.J.); Department of Biology, University of North Carolina, Chapel Hill (D.S.W., V.L.B.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (B.P., S.-W.J.); Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, OH (L.P., S.C., S.C.E.); Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul (E.M.H., J.-Y.P.); School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul (J.-Y.P.); Department of Neurology, Yale University School of Medicine, New Haven, CT (J.-L.T.); and Université Pierre and Marie Curie-Paris 6, CRICM, Groupe Hospitalier Pitié-Salpètrière, France; INSERM, UMRS 975, Groupe Hospitalier Pitié-Salpètrière, Paris, France; APHP, Groupe Hospitalier Pitié-Salpètrière, Paris, France (J.-L.T.).
| |
Collapse
|
28
|
Jarad M, Kuczynski EA, Morrison J, Viloria-Petit AM, Coomber BL. Release of endothelial cell associated VEGFR2 during TGF-β modulated angiogenesis in vitro. BMC Cell Biol 2017; 18:10. [PMID: 28114883 PMCID: PMC5260130 DOI: 10.1186/s12860-017-0127-y] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/18/2017] [Indexed: 01/11/2023] Open
Abstract
Background Sprouting angiogenesis requires vascular endothelial proliferation, migration and morphogenesis. The process is regulated by soluble factors, principally vascular endothelial growth factor (VEGF), and via bidirectional signaling through the Jagged/Notch system, leading to assignment of tip cell and stalk cell identity. The cytokine transforming growth factor beta (TGF-β) can either stimulate or inhibit angiogenesis via its differential surface receptor signaling. Here we evaluate changes in expression of angiogenic signaling receptors when bovine aortic endothelial cells were exposed to TGF-β1 under low serum conditions. Results TGF-β1 induced a dose dependent inhibition of tip cell assignment and subsequent angiogenesis on Matrigel, maximal at 5.0 ng/ml. This occurred via ALK5-dependent pathways and was accompanied by significant upregulation of the TGF-β co-receptor endoglin, and SMAD2 phosphorylation, but no alteration in Smad1/5 activation. TGF-β1 also induced ALK5-dependent downregulation of Notch1 but not of its ligand delta-like ligand 4. Cell associated VEGFR2 (but not VEGFR1) was significantly downregulated and accompanied by reciprocal upregulation of VEGFR2 in conditioned medium. Quantitative polymerase chain reaction analysis revealed that this soluble VEGFR2 was not generated by a selective shift in mRNA isoform transcription. This VEGFR2 in conditioned medium was full-length protein and was associated with increased soluble HSP-90, consistent with a possible shedding of microvesicles/exosomes. Conclusions Taken together, our results suggest that endothelial cells exposed to TGF-β1 lose both tip and stalk cell identity, possibly mediated by loss of VEGFR2 signaling. The role of these events in physiological and pathological angiogenesis requires further investigation. Electronic supplementary material The online version of this article (doi:10.1186/s12860-017-0127-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M Jarad
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada
| | - E A Kuczynski
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada
| | - J Morrison
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada
| | - A M Viloria-Petit
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada
| | - B L Coomber
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada.
| |
Collapse
|
29
|
Howell DW, Duran CL, Tsai SP, Bondos SE, Bayless KJ. Functionalization of Ultrabithorax Materials with Vascular Endothelial Growth Factor Enhances Angiogenic Activity. Biomacromolecules 2016; 17:3558-3569. [DOI: 10.1021/acs.biomac.6b01068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- David W. Howell
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas 77843, United States
| | - Camille L. Duran
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas 77843, United States
| | - Shang-Pu Tsai
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas 77843, United States
| | - Sarah E. Bondos
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas 77843, United States
- Department
of Biochemistry and Cell Biology, Rice University, Houston, Texas 77005, United States
| | - Kayla J. Bayless
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas 77843, United States
| |
Collapse
|
30
|
Claesson-Welsh L. VEGF receptor signal transduction - A brief update. Vascul Pharmacol 2016; 86:14-17. [PMID: 27268035 DOI: 10.1016/j.vph.2016.05.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 03/20/2016] [Accepted: 05/28/2016] [Indexed: 12/31/2022]
Abstract
Vascular endothelial growth factor (VEGF) signal transduction through receptor tyrosine kinases VEGF receptor-1, -2 and -3 is of crucial importance for monocytes/macrophages, blood vascular endothelial and lymphatic endothelial cells both in physiology and in a number of pathologies notably cancer. This brief review summarizes the current status of VEGF receptor signaling with emphasis on in vivo data.
Collapse
Affiliation(s)
- Lena Claesson-Welsh
- Uppsala University, Dept. Immunology, Genetics and Pathology, Rudbeck Laboratory, Dag Hammarskjöldsv 20, 751 85 Uppsala, Sweden.
| |
Collapse
|
31
|
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: 1.6] [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
|
32
|
Song J, Li Z, Yu A. Effects of preserving different veins on flow-through flap survival: An experimental study. Exp Ther Med 2015; 11:318-324. [PMID: 26889261 DOI: 10.3892/etm.2015.2869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 04/07/2015] [Indexed: 11/06/2022] Open
Abstract
Flow-through skin flap grafting is becoming widely used for the reconstruction of skin and soft tissue defects, particularly for patients with poor blood supply around the defect. However, the treatment of the veins remains controversial. In the present study, 5×2-cm skin flaps were created on the left inner thighs of rabbits in order to investigate the effects of various treatments of the veins on the survival of a flow-through skin flap. A femoral artery perforator running through the flap was preserved. Five groups were established in which no veins, one superficial vein (SV), one accompanying vein (AV), one SV plus one AV, or all trunk veins (control) were retained. The percentage of flap area survival was determined on day 10. On days 3, 5, 7 and 9, tissues were harvested from the skin flaps and immunohistochemical analysis was performed in order to count the number of microvessels. Western blot analysis was subsequently completed in order to determine the expression levels of vascular endothelial growth factor (VEGF). The flap areas in which no veins were retained demonstrated significantly reduced survival rates on day 10, as compared with those in the other four groups (P<0.01). Furthermore, flaps with the retention of a SV also demonstrated reduced survival rates, as compared with the AV, AV plus SV and all veins groups (P<0.01); however, there no significant differences were detected between the latter three groups (P>0.05). Immunohistochemical analysis on day 3 detected a greater number of microvessels in the flaps of the control group, as compared with the flaps of the other groups. Furthermore, the AV and AV plus SV groups demonstrated a greater number of microvessels, as compared with the SV and no vein groups, and he no vein group demonstrated the fewest microvessels. No significant differences were found between the AV, AV plus SV and control groups on day 5, 7 and 9. Western blot analysis on day 5 demonstrated that the expression levles of VEGF were significantly increased in the flaps of the AV, AV plus SV and control groups, as compared with those in the SV and no vein groups. No significant differences were detected between the former three groups, and increased VEGF expression levels were detected in the flaps of the SV group, as compared with the no vein group. The flow-through flap grafts with no retained veins barely survived. Anastomosing one AV was adequate for flap survival; however, further studies are required in order to investigate the survival of flow-through flaps in more detail.
Collapse
Affiliation(s)
- Jian Song
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Zonghuan Li
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| | - Aixi Yu
- Department of Orthopedics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430071, P.R. China
| |
Collapse
|
33
|
Hasan SS, Siekmann AF. The same but different: signaling pathways in control of endothelial cell migration. Curr Opin Cell Biol 2015; 36:86-92. [DOI: 10.1016/j.ceb.2015.07.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Revised: 07/16/2015] [Accepted: 07/19/2015] [Indexed: 11/24/2022]
|
34
|
Sawada J, Li F, Komatsu M. R-Ras protein inhibits autophosphorylation of vascular endothelial growth factor receptor 2 in endothelial cells and suppresses receptor activation in tumor vasculature. J Biol Chem 2015; 290:8133-45. [PMID: 25645912 DOI: 10.1074/jbc.m114.591511] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Abnormal angiogenesis is associated with a broad range of medical conditions, including cancer. The formation of neovasculature with functionally defective blood vessels significantly impacts tumor progression, metastasis, and the efficacy of anticancer therapies. Vascular endothelial growth factor (VEGF) potently induces vascular permeability and vessel growth in the tumor microenvironment, and its inhibition normalizes tumor vasculature. In contrast, the signaling of the small GTPase R-Ras inhibits excessive angiogenic growth and promotes the maturation of regenerating blood vessels. R-Ras signaling counteracts VEGF-induced vessel sprouting, permeability, and invasive activities of endothelial cells. In this study, we investigated the effect of R-Ras on VEGF receptor 2 (VEGFR2) activation by VEGF, the key mechanism for angiogenic stimulation. We show that tyrosine phosphorylation of VEGFR2 is significantly elevated in the tumor vasculature and dermal microvessels of VEGF-injected skin in R-Ras knockout mice. In cultured endothelial cells, R-Ras suppressed the internalization of VEGFR2, which is required for full activation of the receptor by VEGF. Consequently, R-Ras strongly suppressed autophosphorylation of the receptor at all five major tyrosine phosphorylation sites. Conversely, silencing of R-Ras resulted in increased VEGFR2 phosphorylation. This effect of R-Ras on VEGFR2 was, at least in part, dependent on vascular endothelial cadherin. These findings identify a novel function of R-Ras to control the response of endothelial cells to VEGF and suggest an underlying mechanism by which R-Ras regulates angiogenesis.
Collapse
Affiliation(s)
- Junko Sawada
- From the Cardiovascular Pathobiology Program and Tumor Microenvironment and Metastasis Program, Sanford-Burnham Medical Research Institute at Lake Nona, Orlando, Florida 32827
| | - Fangfei Li
- From the Cardiovascular Pathobiology Program and Tumor Microenvironment and Metastasis Program, Sanford-Burnham Medical Research Institute at Lake Nona, Orlando, Florida 32827
| | - Masanobu Komatsu
- From the Cardiovascular Pathobiology Program and Tumor Microenvironment and Metastasis Program, Sanford-Burnham Medical Research Institute at Lake Nona, Orlando, Florida 32827
| |
Collapse
|