51
|
Kaposi's sarcoma-associated herpesvirus microRNAs repress breakpoint cluster region protein expression, enhance Rac1 activity, and increase in vitro angiogenesis. J Virol 2015; 89:4249-61. [PMID: 25631082 DOI: 10.1128/jvi.03687-14] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED MicroRNAs (miRNAs) are small, ∼ 22-nucleotide-long RNAs that regulate gene expression posttranscriptionally. Kaposi's sarcoma-associated herpesvirus (KSHV) encodes 12 pre-miRNAs during latency, and the functional significance of these microRNAs during KSHV infection and their cellular targets have been emerging recently. Using a previously reported microarray profiling analysis, we identified breakpoint cluster region mRNA (Bcr) as a cellular target of the KSHV miRNA miR-K12-6-5p (miR-K6-5). Bcr protein levels were repressed in human umbilical vein endothelial cells (HUVECs) upon transfection with miR-K6-5 and during KSHV infection. Luciferase assays wherein the Bcr 3' untranslated region (UTR) was cloned downstream of a luciferase reporter showed repression in the presence of miR-K6-5, and mutation of one of the two predicted miR-K6-5 binding sites relieved this repression. Furthermore, inhibition or deletion of miR-K6-5 in KSHV-infected cells showed increased Bcr protein levels. Together, these results show that Bcr is a direct target of the KSHV miRNA miR-K6-5. To understand the functional significance of Bcr knockdown in the context of KSHV-associated disease, we hypothesized that the knockdown of Bcr, a negative regulator of Rac1, might enhance Rac1-mediated angiogenesis. We found that HUVECs transfected with miR-K6-5 had increased Rac1-GTP levels and tube formation compared to HUVECs transfected with control miRNAs. Knockdown of Bcr in latently KSHV-infected BCBL-1 cells increased the levels of viral RTA, suggesting that Bcr repression by KSHV might aid lytic reactivation. Together, our results reveal a new function for both KSHV miRNAs and Bcr in KSHV infection and suggest that KSHV miRNAs, in part, promote angiogenesis and lytic reactivation. IMPORTANCE Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) infection is linked to multiple human cancers and lymphomas. KSHV encodes small nucleic acids (microRNAs) that can repress the expression of specific human genes, the biological functions of which are still emerging. This report uses a variety of approaches to show that a KSHV microRNA represses the expression of the human gene called breakpoint cluster region (Bcr). Repression of Bcr correlated with the activation of a protein previously shown to cause KS-like lesions in mice (Rac1), an increase in KS-associated phenotypes (tube formation in endothelial cells and vascular endothelial growth factor [VEGF] synthesis), and modification of the life cycle of the virus (lytic replication). Our results suggest that KSHV microRNAs suppress host proteins and contribute to KS-associated pathogenesis.
Collapse
|
52
|
Davis GE, Norden PR, Bowers SLK. Molecular control of capillary morphogenesis and maturation by recognition and remodeling of the extracellular matrix: functional roles of endothelial cells and pericytes in health and disease. Connect Tissue Res 2015; 56:392-402. [PMID: 26305158 PMCID: PMC4765926 DOI: 10.3109/03008207.2015.1066781] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
This review addresses fundamental mechanisms underlying how capillaries form in three-dimensional extracellular matrices and how endothelial cells (ECs) and pericytes co-assemble to form capillary networks. In addition to playing a critical role in supplying oxygen and nutrients to tissues, recent work suggests that blood vessels supply important signals to facilitate tissue development. Here, we hypothesize that another major function of capillaries is to supply signals to suppress major disease mechanisms including inflammation, infection, thrombosis, hemorrhage, edema, ischemic injury, fibrosis, autoimmune disease and tumor growth/progression. Capillary dysfunction plays a key pathogenic role in many human diseases, and thus, this suppressing function may be attenuated and central toward the initiation and progression of disease. We describe how capillaries form through creation of EC-lined tube networks and vascular guidance tunnels in 3D extracellular matrices. Pericytes recruit to the abluminal EC tube surface within these tunnel spaces, and work together to assemble the vascular basement membrane matrix. These processes occur under serum-free conditions in 3D collagen or fibrin matrices and in response to five key growth factors which are stem cell factor, interleukin-3, stromal-derived factor-1α, fibroblast growth factor-2 and insulin. In addition, we identified a key role for EC-derived platelet-derived growth factor-BB and heparin-binding epidermal growth factor in pericyte recruitment and proliferation to promote EC-pericyte tube co-assembly and vascular basement membrane matrix deposition. A molecular understanding of capillary morphogenesis and maturation should lead to novel therapeutic strategies to repair capillary dysfunction in major human disease contexts including cancer and diabetes.
Collapse
Affiliation(s)
- George E Davis
- a Department of Medical Pharmacology and Physiology , Dalton Cardiovascular Research Center, University of Missouri School of Medicine , Columbia , MO , USA
| | - Pieter R Norden
- a Department of Medical Pharmacology and Physiology , Dalton Cardiovascular Research Center, University of Missouri School of Medicine , Columbia , MO , USA
| | - Stephanie L K Bowers
- a Department of Medical Pharmacology and Physiology , Dalton Cardiovascular Research Center, University of Missouri School of Medicine , Columbia , MO , USA
| |
Collapse
|
53
|
Vogler G, Liu J, Iafe TW, Migh E, Mihály J, Bodmer R. Cdc42 and formin activity control non-muscle myosin dynamics during Drosophila heart morphogenesis. ACTA ACUST UNITED AC 2014; 206:909-22. [PMID: 25267295 PMCID: PMC4178965 DOI: 10.1083/jcb.201405075] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Cdc42 and the formins dDAAM and Diaphanous play pivotal roles in heart lumen formation through the spatiotemporal regulation of the actomyosin network. During heart formation, a network of transcription factors and signaling pathways guide cardiac cell fate and differentiation, but the genetic mechanisms orchestrating heart assembly and lumen formation remain unclear. Here, we show that the small GTPase Cdc42 is essential for Drosophila melanogaster heart morphogenesis and lumen formation. Cdc42 genetically interacts with the cardiogenic transcription factor tinman; with dDAAM which belongs to the family of actin organizing formins; and with zipper, which encodes nonmuscle myosin II. Zipper is required for heart lumen formation, and its spatiotemporal activity at the prospective luminal surface is controlled by Cdc42. Heart-specific expression of activated Cdc42, or the regulatory formins dDAAM and Diaphanous caused mislocalization of Zipper and induced ectopic heart lumina, as characterized by luminal markers such as the extracellular matrix protein Slit. Placement of Slit at the lumen surface depends on Cdc42 and formin function. Thus, Cdc42 and formins play pivotal roles in heart lumen formation through the spatiotemporal regulation of the actomyosin network.
Collapse
Affiliation(s)
- Georg Vogler
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Jiandong Liu
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Timothy W Iafe
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| | - Ede Migh
- Biological Research Centre, Hungarian Academy of Sciences, Institute of Genetics, H-6726 Szeged, Hungary
| | - József Mihály
- Biological Research Centre, Hungarian Academy of Sciences, Institute of Genetics, H-6726 Szeged, Hungary
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford-Burnham Medical Research Institute, La Jolla, CA 92037
| |
Collapse
|
54
|
Nehilla BJ, Nataraj N, Gaborski TR, McGrath JL. Endothelial vacuolization induced by highly permeable silicon membranes. Acta Biomater 2014; 10:4670-4677. [PMID: 25072618 DOI: 10.1016/j.actbio.2014.07.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 06/14/2014] [Accepted: 07/18/2014] [Indexed: 11/24/2022]
Abstract
Assays for initiating, controlling and studying endothelial cell behavior and blood vessel formation have applications in developmental biology, cancer and tissue engineering. In vitro vasculogenesis models typically combine complex three-dimensional gels of extracellular matrix proteins with other stimuli like growth factor supplements. Biomaterials with unique micro- and nanoscale features may provide simpler substrates to study endothelial cell morphogenesis. In this work, patterns of nanoporous, nanothin silicon membranes (porous nanocrystalline silicon, or pnc-Si) are fabricated to control the permeability of an endothelial cell culture substrate. Permeability on the basal surface of primary and immortalized endothelial cells causes vacuole formation and endothelial organization into capillary-like structures. This phenomenon is repeatable, robust and controlled entirely by patterns of free-standing, highly permeable pnc-Si membranes. Pnc-Si is a new biomaterial with precisely defined micro- and nanoscale features that can be used as a unique in vitro platform to study endothelial cell behavior and vasculogenesis.
Collapse
|
55
|
|
56
|
Seidelmann SB, Lighthouse JK, Greif DM. Development and pathologies of the arterial wall. Cell Mol Life Sci 2014; 71:1977-99. [PMID: 24071897 PMCID: PMC11113178 DOI: 10.1007/s00018-013-1478-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 09/11/2013] [Accepted: 09/12/2013] [Indexed: 01/13/2023]
Abstract
Arteries consist of an inner single layer of endothelial cells surrounded by layers of smooth muscle and an outer adventitia. The majority of vascular developmental studies focus on the construction of endothelial networks through the process of angiogenesis. Although many devastating vascular diseases involve abnormalities in components of the smooth muscle and adventitia (i.e., the vascular wall), the morphogenesis of these layers has received relatively less attention. Here, we briefly review key elements underlying endothelial layer formation and then focus on vascular wall development, specifically on smooth muscle cell origins and differentiation, patterning of the vascular wall, and the role of extracellular matrix and adventitial progenitor cells. Finally, we discuss select human diseases characterized by marked vascular wall abnormalities. We propose that continuing to apply approaches from developmental biology to the study of vascular disease will stimulate important advancements in elucidating disease mechanism and devising novel therapeutic strategies.
Collapse
MESH Headings
- Angiogenic Proteins/genetics
- Angiogenic Proteins/metabolism
- Animals
- Arteries/growth & development
- Arteries/metabolism
- Arteries/pathology
- Cardiovascular Diseases/genetics
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/pathology
- Cell Differentiation
- Cell Lineage/genetics
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Endothelium, Vascular/growth & development
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Gene Expression Regulation, Developmental
- Humans
- Morphogenesis/genetics
- Muscle, Smooth, Vascular/growth & development
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Neovascularization, Pathologic
- Neovascularization, Physiologic
Collapse
Affiliation(s)
- Sara B. Seidelmann
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| | - Janet K. Lighthouse
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| | - Daniel M. Greif
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, Yale University School of Medicine, 300 George St., Rm 773J, New Haven, CT 06511 USA
| |
Collapse
|
57
|
Boas SEM, Merks RMH. Synergy of cell-cell repulsion and vacuolation in a computational model of lumen formation. J R Soc Interface 2014; 11:20131049. [PMID: 24430123 PMCID: PMC3899873 DOI: 10.1098/rsif.2013.1049] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
A key step in blood vessel development (angiogenesis) is lumen formation: the hollowing of vessels for blood perfusion. Two alternative lumen formation mechanisms are suggested to function in different types of blood vessels. The vacuolation mechanism is suggested for lumen formation in small vessels by coalescence of intracellular vacuoles, a view that was extended to extracellular lumen formation by exocytosis of vacuoles. The cell–cell repulsion mechanism is suggested to initiate extracellular lumen formation in large vessels by active repulsion of adjacent cells, and active cell shape changes extend the lumen. We used an agent-based computer model, based on the cellular Potts model, to compare and study both mechanisms separately and combined. An extensive sensitivity analysis shows that each of the mechanisms on its own can produce lumens in a narrow region of parameter space. However, combining both mechanisms makes lumen formation much more robust to the values of the parameters, suggesting that the mechanisms may work synergistically and operate in parallel, rather than in different vessel types.
Collapse
Affiliation(s)
- Sonja E M Boas
- Life Sciences Group, Centrum Wiskunde and Informatica (CWI), , Amsterdam, The Netherlands
| | | |
Collapse
|
58
|
Kim SJ, Wan Q, Cho E, Han B, Yoder MC, Voytik-Harbin SL, Na S. Matrix rigidity regulates spatiotemporal dynamics of Cdc42 activity and vacuole formation kinetics of endothelial colony forming cells. Biochem Biophys Res Commun 2014; 443:1280-5. [PMID: 24393843 DOI: 10.1016/j.bbrc.2013.12.135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 12/26/2013] [Indexed: 12/15/2022]
Abstract
Recent evidence has shown that endothelial colony forming cells (ECFCs) may serve as a cell therapy for improving blood vessel formation in subjects with vascular injury, largely due to their robust vasculogenic potential. The Rho family GTPase Cdc42 is known to play a primary role in this vasculogenesis process, but little is known about how extracellular matrix (ECM) rigidity affects Cdc42 activity during the process. In this study, we addressed two questions: Does matrix rigidity affect Cdc42 activity in ECFC undergoing early vacuole formation? How is the spatiotemporal activation of Cdc42 related to ECFC vacuole formation? A fluorescence resonance energy transfer (FRET)-based Cdc42 biosensor was used to examine the effects of the rigidity of three-dimensional (3D) collagen matrices on spatiotemporal activity of Cdc42 in ECFCs. Collagen matrix stiffness was modulated by varying the collagen concentration and therefore fibril density. The results showed that soft (150 Pa) matrices induced an increased level of Cdc42 activity compared to stiff (1 kPa) matrices. Time-course imaging and colocalization analysis of Cdc42 activity and vacuole formation revealed that Cdc42 activity was colocalized to the periphery of cytoplasmic vacuoles. Moreover, soft matrices generated faster and larger vacuoles than stiff matrices. The matrix-driven vacuole formation was enhanced by a constitutively active Cdc42 mutant, but significantly inhibited by a dominant-negative Cdc42 mutant. Collectively, the results suggest that matrix rigidity is a strong regulator of Cdc42 activity and vacuole formation kinetics, and that enhanced activity of Cdc42 is an important step in early vacuole formation in ECFCs.
Collapse
Affiliation(s)
- Seung Joon Kim
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Qiaoqiao Wan
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Eunhye Cho
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Bumsoo Han
- School of Mechanical Engineering, Purdue University, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Mervin C Yoder
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sherry L Voytik-Harbin
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA; Department of Basic Medical Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Sungsoo Na
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA.
| |
Collapse
|
59
|
Charpentier MS, Conlon FL. Cellular and molecular mechanisms underlying blood vessel lumen formation. Bioessays 2013; 36:251-9. [PMID: 24323945 DOI: 10.1002/bies.201300133] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The establishment of a functional vascular system requires multiple complex steps throughout embryogenesis, from endothelial cell (EC) specification to vascular patterning into venous and arterial hierarchies. Following the initial assembly of ECs into a network of cord-like structures, vascular expansion and remodeling occur rapidly through morphogenetic events including vessel sprouting, fusion, and pruning. In addition, vascular morphogenesis encompasses the process of lumen formation, critical for the transformation of cords into perfusable vascular tubes. Studies in mouse, zebrafish, frog, and human endothelial cells have begun to outline the cellular and molecular requirements underlying lumen formation. Although the lumen can be generated through diverse mechanisms, the coordinated participation of multiple conserved molecules including transcription factors, small GTPases, and adhesion and polarity proteins remains a fundamental principle, leading us closer to a more thorough understanding of this complex event.
Collapse
Affiliation(s)
- Marta S Charpentier
- McAllister Heart Institute, Departments of Biology and Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|
60
|
Barreto-Ortiz SF, Zhang S, Davenport M, Fradkin J, Ginn B, Mao HQ, Gerecht S. A novel in vitro model for microvasculature reveals regulation of circumferential ECM organization by curvature. PLoS One 2013; 8:e81061. [PMID: 24278378 PMCID: PMC3836741 DOI: 10.1371/journal.pone.0081061] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 10/09/2013] [Indexed: 12/25/2022] Open
Abstract
In microvascular vessels, endothelial cells are aligned longitudinally whereas several components of the extracellular matrix (ECM) are organized circumferentially. While current three-dimensional (3D) in vitro models for microvasculature have allowed the study of ECM-regulated tubulogenesis, they have limited control over topographical cues presented by the ECM and impart a barrier for the high-resolution and dynamic study of multicellular and extracellular organization. Here we exploit a 3D fibrin microfiber scaffold to develop a novel in vitro model of the microvasculature that recapitulates endothelial alignment and ECM deposition in a setting that also allows the sequential co-culture of mural cells. We show that the microfibers' nanotopography induces longitudinal adhesion and alignment of endothelial colony-forming cells (ECFCs), and that these deposit circumferentially organized ECM. We found that ECM wrapping on the microfibers is independent of ECFCs' actin and microtubule organization, but it is dependent on the curvature of the microfiber. Microfibers with smaller diameters (100–400 µm) guided circumferential ECM deposition, whereas microfibers with larger diameters (450 µm) failed to support wrapping ECM. Finally, we demonstrate that vascular smooth muscle cells attached on ECFC-seeded microfibers, depositing collagen I and elastin. Collectively, we establish a novel in vitro model for the sequential control and study of microvasculature development and reveal the unprecedented role of the endothelium in organized ECM deposition regulated by the microfiber curvature.
Collapse
Affiliation(s)
- Sebastian F. Barreto-Ortiz
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Shuming Zhang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Matthew Davenport
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jamie Fradkin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Brian Ginn
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Hai-Quan Mao
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Sharon Gerecht
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Sciences-Oncology Center and Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
61
|
Deletion of Cdc42 enhances ADAM17-mediated vascular endothelial growth factor receptor 2 shedding and impairs vascular endothelial cell survival and vasculogenesis. Mol Cell Biol 2013; 33:4181-97. [PMID: 23979594 DOI: 10.1128/mcb.00650-13] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cdc42 is a Ras-related GTPase that plays an important role in the regulation of a range of cellular functions, including cell migration, proliferation, and survival. Consistent with its critical functions in vitro, the inactivation of Cdc42 in mice has been shown to result in embryonic lethality at embryonic day 6.5 (E6.5) before blood vessel formation. To determine the role of Cdc42 in new blood vessel formation, we have generated vascular endothelial cell (EC)-specific Cdc42 knockout mice by crossing Cdc42(flox/flox) mice with Tie2-Cre mice. The deletion of Cdc42 in ECs caused embryonic lethality with vasculogenesis and angiogenesis defects. We observed that Cdc42 is critical for EC migration and survival but not for cell cycle progression. Moreover, we found that the inactivation of Cdc42 in ECs decreased the level of vascular endothelial growth factor receptor 2 (VEGFR2) protein on the EC surface and promoted the production of a 75-kDa membrane-associated C-terminal VEGFR2 fragment. Using cultured primary mouse ECs and human umbilical vein ECs, we have demonstrated that the deletion of Cdc42 increased ADAM17-mediated VEGFR2 shedding. Notably, inhibition of ADAM17 or overexpression of VEGFR2 can partially reverse Cdc42 deletion-induced EC apoptosis. These data indicate that Cdc42 is essential for VEGFR2-mediated signal transduction in blood vessel formation.
Collapse
|
62
|
Suzuki R, Yamamoto H, Ngan CY, Ohtsuka M, Kitani K, Uemura M, Nishimura J, Takemasa I, Mizushima T, Sekimoto M, Minamoto T, Doki Y, Mori M. Inhibition of angiopoietin 2 attenuates lumen formation of tumour-associated vessels in vivo. Int J Oncol 2013; 43:1447-55. [PMID: 23982687 DOI: 10.3892/ijo.2013.2076] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 06/14/2013] [Indexed: 11/06/2022] Open
Abstract
Anti-angiogenic therapy, inhibition of a co-operative process with vascular endothelial cells and pericytes could be an effective strategy to treat malignant tumours. Apart from vascular endothelial growth factor (VEGF), angiopoietin 2 (Ang2) is a promising target of anti-angiogenic therapy. Although inhibition of Ang2 has been shown to decrease tumour size in preclinical and phase I trials, its mechanisms of action remain largely unknown. To elucidate the mechanisms of Ang2 inhibition, we have focused on differentiation of the vessels as well as on growth of the vessels, especially in vivo. L1-10, a selective Ang2 inhibitor was used. The in vitro effects of Ang2 inhibition or addition of Ang2 using HUVECs were also examined. Growth and differentiation of tumour-associated vessels were investigated in xenografts derived from a colon cancer treated by L1-10. Effects of VEGF inhibition were also examined to discriminate Ang2-specific action on the tumour-associated vessels. In vitro studies showed that VEGF enhanced proliferation and tube formation of HUVECs, and caused a significant increase in Rac1 and CDC42 expression when cultured in the collagen matrix gel, whereas neither Ang2 nor L1-10 affected in vitro behaviour of HUVECs or levels of the proteins. In vivo, on the other hand, we found that Ang2 inhibition with treatment of L1-10 dose‑dependently decreased tumour growth. Furthermore, we found that L1-10 treatment extends the tumour-associated vessels whilst it suppressed a sound lumen formation. Histological analysis on xenografts suggests that Ang2 inhibition could have disturbed in vivo vascular differentiation. Our data provide a novel aspect that Ang2 may play an essential role in in vivo vascular differentiation, thus supporting a rationale for Ang2-targeted therapy against colon cancer.
Collapse
Affiliation(s)
- Rei Suzuki
- Department of Surgery, Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
63
|
Whittington CF, Yoder MC, Voytik-Harbin SL. Collagen-polymer guidance of vessel network formation and stabilization by endothelial colony forming cells in vitro. Macromol Biosci 2013; 13:1135-49. [PMID: 23832790 DOI: 10.1002/mabi.201300128] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 04/11/2013] [Indexed: 01/13/2023]
Abstract
Vessel morphogenesis is vital to regenerative medicine strategies. Here, collagen polymers, specified by intermolecular cross-link composition, are used to independently vary microstructure (fibril density, interfibril branching) and physical properties (stiffness) to guide 3D vessel network formation by endothelial colony forming cells (ECFC) in vitro. Increasing stiffness, by modulation of fibril density or interfibril branching, increases vessel diameter, length and branching. Oligomer matrices also induce vessel stabilization via type IV collagen deposition. This work shows that ECFC vessel formation depends on the interplay of collagen microstructure and physical properties and names oligomers and intermolecular cross-links as key design parameters for vascular-inductive matrices.
Collapse
Affiliation(s)
- Catherine F Whittington
- Weldon School of Biomedical Engineering, College of Engineering, Purdue University, 206 Martin Jischke Drive, West Lafayette, Indiana, 47907, USA
| | | | | |
Collapse
|
64
|
Charpentier MS, Christine KS, Amin NM, Dorr KM, Kushner EJ, Bautch VL, Taylor JM, Conlon FL. CASZ1 promotes vascular assembly and morphogenesis through the direct regulation of an EGFL7/RhoA-mediated pathway. Dev Cell 2013; 25:132-43. [PMID: 23639441 DOI: 10.1016/j.devcel.2013.03.003] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 01/22/2013] [Accepted: 03/01/2013] [Indexed: 01/11/2023]
Abstract
The formation of the vascular system is essential for embryonic development and homeostasis. However, transcriptional control of this process is not fully understood. Here we report an evolutionarily conserved role for the transcription factor CASZ1 (CASTOR) in blood vessel assembly and morphogenesis. In the absence of CASZ1, Xenopus embryos fail to develop a branched and lumenized vascular system, and CASZ1-depleted human endothelial cells display dramatic alterations in adhesion, morphology, and sprouting. Mechanistically, we show that CASZ1 directly regulates Epidermal Growth Factor-Like Domain 7 (Egfl7). We further demonstrate that defects of CASZ1- or EGFL7-depleted cells are in part due to diminished RhoA expression and impaired focal adhesion localization. Moreover, these abnormal endothelial cell behaviors in CASZ1-depleted cells can be rescued by restoration of Egfl7. Collectively, these studies show that CASZ1 is required to directly regulate an EGFL7/RhoA-mediated pathway to promote vertebrate vascular development.
Collapse
Affiliation(s)
- Marta S Charpentier
- McAllister Heart Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-3280, USA
| | | | | | | | | | | | | | | |
Collapse
|
65
|
Abstract
Interconnection of epithelial tubules is a crucial process during organogenesis. Organisms have evolved sets of molecular and cellular strategies to generate an interconnected tubular network during animal development. Spatiotemporal control of common cellular strategies includes dissolution of the basement membrane, apoptosis, rearrangements of cell adhesion junctions, and mesenchymal-like invasive cellular behaviors prior to tubular interconnection. Different model systems exhibit varying degrees of active invasive-like behaviors that precede tubular interconnection, which may reflect changes in cell polarity or differential adhesive cell states. Studies in this newly-emerging field of tubular interconnections will provide a greater understanding of pediatric diseases and cancer metastasis, as well as generate fundamentally new insights into lumen formation pathology, or lumopathies.
Collapse
Affiliation(s)
- Robert M Kao
- Departments of Molecular and Cellular Biology and Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
66
|
Davis GE, Kim DJ, Meng CX, Norden PR, Speichinger KR, Davis MT, Smith AO, Bowers SLK, Stratman AN. Control of vascular tube morphogenesis and maturation in 3D extracellular matrices by endothelial cells and pericytes. Methods Mol Biol 2013; 1066:17-28. [PMID: 23955730 DOI: 10.1007/978-1-62703-604-7_2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
An important advance using in vitro EC tube morphogenesis and maturation models has been the development of systems using serum-free defined media. Using this approach, the growth factors and cytokines which are actually necessary for these events can be determined. The first model developed by our laboratory was such a system where we showed that phorbol ester was needed in order to promote survival and tube morphogenesis in 3D collagen matrices. Recently, we have developed a new system in which the hematopoietic stem cell cytokines, stem cell factor (SCF), interleukin-3 (IL-3), and stromal derived factor-1α (SDF-1α) were added in conjunction with FGF-2 to promote human EC tube morphogenesis in 3D collagen matrices under serum-free defined conditions. This new model using SCF, IL-3, SDF-1α, and FGF-2 also works well following the addition of pericytes where EC tube formation occurs, pericytes are recruited to the tubes, and vascular basement membrane matrix assembly occurs following EC-pericyte interactions. In this chapter, we describe several in vitro assay models that we routinely utilize to investigate the molecular requirements that are critical to EC tube formation and maturation events in 3D extracellular matrix environments.
Collapse
Affiliation(s)
- George E Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine and Dalton Cardiovascular Research Center, Columbia, MO, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
67
|
Ribatti D, Crivellato E. “Sprouting angiogenesis”, a reappraisal. Dev Biol 2012; 372:157-65. [DOI: 10.1016/j.ydbio.2012.09.018] [Citation(s) in RCA: 209] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 09/22/2012] [Accepted: 09/24/2012] [Indexed: 01/15/2023]
|
68
|
Abstract
Peripheral arterial disease (PAD) is a common vascular disease that reduces blood flow capacity to the legs of patients. PAD leads to exercise intolerance that can progress in severity to greatly limit mobility, and in advanced cases leads to frank ischemia with pain at rest. It is estimated that 12 to 15 million people in the United States are diagnosed with PAD, with a much larger population that is undiagnosed. The presence of PAD predicts a 50% to 1500% increase in morbidity and mortality, depending on severity. Treatment of patients with PAD is limited to modification of cardiovascular disease risk factors, pharmacological intervention, surgery, and exercise therapy. Extended exercise programs that involve walking approximately five times per week, at a significant intensity that requires frequent rest periods, are most significant. Preclinical studies and virtually all clinical trials demonstrate the benefits of exercise therapy, including improved walking tolerance, modified inflammatory/hemostatic markers, enhanced vasoresponsiveness, adaptations within the limb (angiogenesis, arteriogenesis, and mitochondrial synthesis) that enhance oxygen delivery and metabolic responses, potentially delayed progression of the disease, enhanced quality of life indices, and extended longevity. A synthesis is provided as to how these adaptations can develop in the context of our current state of knowledge and events known to be orchestrated by exercise. The benefits are so compelling that exercise prescription should be an essential option presented to patients with PAD in the absence of contraindications. Obviously, selecting for a lifestyle pattern that includes enhanced physical activity prior to the advance of PAD limitations is the most desirable and beneficial.
Collapse
Affiliation(s)
- Tara L Haas
- Angiogenesis Research Group, Muscle Health Research Centre, Faculty of Health, York University, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
69
|
|
70
|
Lei Y, Zouani OF, Rémy M, Ayela C, Durrieu MC. Geometrical microfeature cues for directing tubulogenesis of endothelial cells. PLoS One 2012; 7:e41163. [PMID: 22829923 PMCID: PMC3400641 DOI: 10.1371/journal.pone.0041163] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Accepted: 06/18/2012] [Indexed: 12/21/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels by sprouting from pre-existing ones, is critical for the establishment and maintenance of complex tissues. Angiogenesis is usually triggered by soluble growth factors such as VEGF. However, geometrical cues also play an important role in this process. Here we report the induction of angiogenesis solely by SVVYGLR peptide micropatterning on polymer surfaces. SVVYGLR peptide stripes were micropatterned onto polymer surfaces by photolithography to study their effects on endothelial cell (EC) behavior. Our results showed that the EC behaviors (cell spreading, orientation and migration) were significantly more guided and regulated on narrower SVVYGLR micropatterns (10 and 50 µm) than on larger stripes (100 µm). Also, EC morphogenesis into tube formation was switched on onto the smaller patterns. We illustrated that the central lumen of tubular structures can be formed by only one-to-four cells due to geometrical constraints on the micropatterns which mediated cell-substrate adhesion and generated a correct maturation of adherens junctions. In addition, sprouting of ECs and vascular networks were also induced by geometrical cues on surfaces micropatterned with SVVYGLR peptides. These micropatterned surfaces provide opportunities for mimicking angiogenesis by peptide modification instead of exogenous growth factors. The organization of ECs into tubular structures and the induction of sprouting angiogenesis are important towards the fabrication of vascularized tissues, and this work has great potential applications in tissue engineering and tissue regeneration.
Collapse
Affiliation(s)
- Yifeng Lei
- INSERM U1026, Université Victor Segalen Bordeaux 2, Bordeaux, France
- CBMN, UMR CNRS 5248, Université Bordeaux 1, Pessac, France
- * E-mail: (YL); (OFZ)
| | - Omar F. Zouani
- INSERM U1026, Université Victor Segalen Bordeaux 2, Bordeaux, France
- CBMN, UMR CNRS 5248, Université Bordeaux 1, Pessac, France
- * E-mail: (YL); (OFZ)
| | - Murielle Rémy
- INSERM U1026, Université Victor Segalen Bordeaux 2, Bordeaux, France
| | - Cédric Ayela
- IMS, UMR CNRS 5218, Université de Bordeaux, Talence, France
| | - Marie-Christine Durrieu
- INSERM U1026, Université Victor Segalen Bordeaux 2, Bordeaux, France
- CBMN, UMR CNRS 5248, Université Bordeaux 1, Pessac, France
| |
Collapse
|
71
|
Sukmana I. Microvascular guidance: a challenge to support the development of vascularised tissue engineering construct. ScientificWorldJournal 2012; 2012:201352. [PMID: 22623881 PMCID: PMC3349125 DOI: 10.1100/2012/201352] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 11/20/2011] [Indexed: 01/11/2023] Open
Abstract
The guidance of endothelial cell organization into a capillary network has been a long-standing challenge in tissue engineering. Some research efforts have been made to develop methods to promote capillary networks inside engineered tissue constructs. Capillary and vascular networks that would mimic blood microvessel function can be used to subsequently facilitate oxygen and nutrient transfer as well as waste removal. Vascularization of engineering tissue construct is one of the most favorable strategies to overpass nutrient and oxygen supply limitation, which is often the major hurdle in developing thick and complex tissue and artificial organ. This paper addresses recent advances and future challenges in developing three-dimensional culture systems to promote tissue construct vascularization allowing mimicking blood microvessel development and function encountered in vivo. Bioreactors systems that have been used to create fully vascularized functional tissue constructs will also be outlined.
Collapse
Affiliation(s)
- Irza Sukmana
- Medical Implant Technology-MediTeg Research Group, Department of Biomechanics and Biomedical Materials, Universiti Teknologi Malaysia, P23 UTM Skudai, Johore, Johor Bahru, Malaysia.
| |
Collapse
|
72
|
Kurogane Y, Miyata M, Kubo Y, Nagamatsu Y, Kundu RK, Uemura A, Ishida T, Quertermous T, Hirata KI, Rikitake Y. FGD5 mediates proangiogenic action of vascular endothelial growth factor in human vascular endothelial cells. Arterioscler Thromb Vasc Biol 2012; 32:988-96. [PMID: 22328776 DOI: 10.1161/atvbaha.111.244004] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular endothelial growth factor (VEGF) exerts proangiogenic action and induces activation of a variety of proangiogenic signaling pathways, including the Rho family small G proteins. However, regulators of the Rho family small G proteins in vascular endothelial cells (ECs) are poorly understood. Here we attempted to clarify the expression, subcellular localization, downstream effectors, and proangiogenic role of FGD5, a member of the FGD family of guanine nucleotide exchange factors. METHODS AND RESULTS FGD5 was shown to be selectively expressed in cultured human vascular ECs. Immunofluorescence microscopy showed that the signal for FGD5 was observed at peripheral membrane ruffles and perinuclear regions in human umbilical vein ECs. Overexpression of FGD5 increased Cdc42 activity, whereas knockdown of FGD5 by small interfering RNAs inhibited the VEGF-induced activation of Cdc42 and extracellular signal-regulated kinase. VEGF-promoted capillary-like network formation, permeability, directional movement, and proliferation of human umbilical vein ECs and the reorientation of the Golgi complex during directional cell movement were attenuated by knockdown of FGD5. CONCLUSIONS This study provides the first demonstration of expression, subcellular localization, and function of FGD5 in vascular ECs. The results suggest that FGD5 regulates proangiogenic action of VEGF in vascular ECs, including network formation, permeability, directional movement, and proliferation.
Collapse
Affiliation(s)
- Yusuke Kurogane
- Division of Cardiovascular Medicine, Department of Internal Medicine, Kobe University Graduate School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
73
|
Sánchez NS, Barnett JV. TGFβ and BMP-2 regulate epicardial cell invasion via TGFβR3 activation of the Par6/Smurf1/RhoA pathway. Cell Signal 2012; 24:539-548. [PMID: 22033038 PMCID: PMC3237859 DOI: 10.1016/j.cellsig.2011.10.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Accepted: 10/10/2011] [Indexed: 01/19/2023]
Abstract
Coronary vessel development requires transfer of mesothelial cells to the heart surface to form the epicardium where some cells subsequently undergo epithelial-mesenchymal transformation (EMT) and invade the subepicardial matrix. Tgfbr3(-/-) mice die due to failed coronary vessel formation associated with decreased epicardial cell invasion but the mediators downstream of TGFβR3 are not well described. TGFβR3-dependent endocardial EMT stimulated by either TGFβ2 or BMP-2 requires activation of the Par6/Smurf1/RhoA 1pathway where Activin Receptor Like Kinase (ALK5) signals Par6 to act downstream of TGFβ to recruit Smurf1 to target RhoA for degradation to regulate apical-basal polarity and tight junction dissolution. Here we asked if this pathway was operant in epicardial cells and if TGFβR3 was required to access this pathway. Targeting of ALK5 in Tgfbr3(+/+) cells inhibited loss of epithelial character and invasion. Overexpression of wild-type (wt) Par6, but not dominant negative (dn) Par6, induced EMT and invasion while targeting Par6 by siRNA inhibited EMT and invasion. Overexpression of Smurf1 and dnRhoA induced loss of epithelial character and invasion. Targeting of Smurf1 by siRNA or overexpression of constitutively active (ca) RhoA inhibited EMT and invasion. In Tgfbr3(-/-) epicardial cells which have a decreased ability to invade collagen gels in response to TGFβ2, overexpression of wtPar6, Smurf1, or dnRhoA had a diminished ability to induce invasion. Overexpression of TGFβR3 in Tgfbr3(-/-) cells, followed by siRNA targeting of Par6 or Smurf1, diminished the ability of TGFβR3 to rescue invasion demonstrating that the Par6/Smurf1/RhoA pathway is activated downstream of TGFβR3 in epicardial cells.
Collapse
Affiliation(s)
- Nora S Sánchez
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232 USA.
| | - Joey V Barnett
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232 USA.
| |
Collapse
|
74
|
Stratman AN, Davis GE. Endothelial cell-pericyte interactions stimulate basement membrane matrix assembly: influence on vascular tube remodeling, maturation, and stabilization. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2012; 18:68-80. [PMID: 22166617 PMCID: PMC3919655 DOI: 10.1017/s1431927611012402] [Citation(s) in RCA: 165] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Extracellular matrix synthesis and deposition surrounding the developing vasculature are critical for vessel remodeling and maturation events. Although the basement membrane is an integral structure underlying endothelial cells (ECs), few studies, until recently, have been performed to understand its formation in this context. In this review article, we highlight new data demonstrating a corequirement for ECs and pericytes to properly deposit and assemble vascular basement membranes during morphogenic events. In EC only cultures or under conditions whereby pericyte recruitment is blocked, there is a lack of basement membrane assembly, decreased vessel stability (with increased susceptibility to pro-regressive stimuli), and increased EC tube widths (a marker of dysfunctional EC-pericyte interactions). ECs and pericytes both contribute basement membrane components and, furthermore, both cells induce the expression of particular components as well as integrins that recognize them. The EC-derived factors--platelet derived growth factor-BB and heparin binding-epidermal growth factor--are both critical for pericyte recruitment to EC tubes and concomitant vascular basement membrane formation in vitro and in vivo. Thus, heterotypic EC-pericyte interactions play a fundamental role in vascular basement membrane matrix deposition, a critical tube maturation event that is altered in key disease states such as diabetes and cancer.
Collapse
Affiliation(s)
- Amber N. Stratman
- Department of Medical Pharmacology and Physiology, University of Missouri- Columbia, 65212
| | - George E. Davis
- Department of Medical Pharmacology and Physiology, University of Missouri- Columbia, 65212
- Department of Pathology and Anatomical Sciences, University of Missouri- Columbia, 65212
| |
Collapse
|
75
|
Yuan L, Le Bras A, Sacharidou A, Itagaki K, Zhan Y, Kondo M, Carman CV, Davis GE, Aird WC, Oettgen P. ETS-related gene (ERG) controls endothelial cell permeability via transcriptional regulation of the claudin 5 (CLDN5) gene. J Biol Chem 2012; 287:6582-91. [PMID: 22235125 DOI: 10.1074/jbc.m111.300236] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
ETS-related gene (ERG) is a member of the ETS transcription factor family. Our previous studies have shown that ERG expression is highly enriched in endothelial cells (EC) both in vitro and in vivo. ERG expression is markedly repressed in response to inflammatory stimuli. It has been shown that ERG is a positive regulator of several EC-restricted genes including VE-cadherin, endoglin, and von Willebrand factor, and a negative regulator of other genes such as interleukin (IL)-8 and intercellular adhesion molecule (ICAM)-1. In this study we have identified a novel role for ERG in the regulation of EC barrier function. ERG knockdown results in marked increases in EC permeability. This is associated with a significant increase of stress fiber and gap formation in EC. Furthermore, we identify CLDN5 as a downstream target of ERG in EC. Thus, our results suggest that ERG plays a pivotal role in regulating EC barrier function and that this effect is mediated in part through its regulation of CLDN5 gene expression.
Collapse
Affiliation(s)
- Lei Yuan
- Division of Cardiology, Harvard Medical School, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Johnson JA, Hemnes AR, Perrien DS, Schuster M, Robinson LJ, Gladson S, Loibner H, Bai S, Blackwell TR, Tada Y, Harral JW, Talati M, Lane KB, Fagan KA, West J. Cytoskeletal defects in Bmpr2-associated pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2011; 302:L474-84. [PMID: 22180660 DOI: 10.1152/ajplung.00202.2011] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The heritable form of pulmonary arterial hypertension (PAH) is typically caused by a mutation in bone morphogenic protein receptor type 2 (BMPR2), and mice expressing Bmpr2 mutations develop PAH with features similar to human disease. BMPR2 is known to interact with the cytoskeleton, and human array studies in PAH patients confirm alterations in cytoskeletal pathways. The goal of this study was to evaluate cytoskeletal defects in BMPR2-associated PAH. Expression arrays on our Bmpr2 mutant mouse lungs revealed cytoskeletal defects as a prominent molecular consequence of universal expression of a Bmpr2 mutation (Rosa26-Bmpr2(R899X)). Pulmonary microvascular endothelial cells cultured from these mice have histological and functional cytoskeletal defects. Stable transfection of different BMPR2 mutations into pulmonary microvascular endothelial cells revealed that cytoskeletal defects are common to multiple BMPR2 mutations and are associated with activation of the Rho GTPase, Rac1. Rac1 defects are corrected in cell culture and in vivo through administration of exogenous recombinant human angiotensin-converting enzyme 2 (rhACE2). rhACE2 reverses 77% of gene expression changes in Rosa26-Bmpr2(R899X) transgenic mice, in particular, correcting defects in cytoskeletal function. Administration of rhACE2 to Rosa26-Bmpr2(R899X) mice with established PAH normalizes pulmonary pressures. Together, these findings suggest that cytoskeletal function is central to the development of BMPR2-associated PAH and that intervention against cytoskeletal defects may reverse established disease.
Collapse
Affiliation(s)
- Jennifer A Johnson
- Division of Allergy, Pulmonary, and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2650, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Lee CY, Bautch VL. Ups and Downs of Guided Vessel Sprouting: The Role of Polarity. Physiology (Bethesda) 2011; 26:326-33. [DOI: 10.1152/physiol.00018.2011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Blood vessel networks expand to meet oxygen demands via sprouting angiogenesis. This process is heterogeneous but not random; as sprouts form and extend, neighboring endothelial cells do not sprout but divide. Sprouting is regulated by local sprout guidance cues produced by the vessels themselves, as well as extrinsic cues. Endothelial cells in developing vessels orient in several axes to establish migratory polarity, apical-basolateral polarity, and planar cell polarity. Although little is known about how polarity axes are set up or maintained, they are important for vessel formation and function. This review focuses on the current knowledge of how blood vessel sprouting is regulated and guided, the role of endothelial cell polarity in forming vessels, and how these processes affect vessel function and are potentially perturbed in pathologies with vascular components.
Collapse
Affiliation(s)
| | - Victoria L. Bautch
- Department of Biology,
- McAllister Heart Institute,
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
78
|
Luo Y, Chen W, Zhou H, Liu L, Shen T, Alexander JS, Zheng S, Lu Y, Huang S. Cryptotanshinone inhibits lymphatic endothelial cell tube formation by suppressing VEGFR-3/ERK and small GTPase pathways. Cancer Prev Res (Phila) 2011; 4:2083-91. [PMID: 21881029 DOI: 10.1158/1940-6207.capr-11-0319] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cryptotanshinone (CPT), isolated from the plant Salvia miltiorrhiza Bunge, is a potential anticancer agent. However, the underlying mechanism remains to be defined. Here, we show that CPT inhibited lymphangiogenesis in an in vitro model (tube formation). This effect was partly attributed to inhibiting expression of VEGF receptor 3 (VEGFR-3) in murine lymphatic endothelial cells (LEC), as overexpression of VEGFR-3 conferred resistance to CPT inhibition of the tube formation, whereas downregulation of VEGFR-3 mimicked the effect of CPT, blocking the tube formation. Furthermore, CPT inhibited phosphorylation of the extracellular signal-related kinase 1/2 (ERK1/2). Overexpression of VEGFR-3 attenuated CPT inhibition of ERK1/2 phosphorylation, whereas downregulation of VEGFR-3 inhibited ERK1/2 phosphorylation in LECs. Expression of constitutively active MKK1 resulted in activation of ERK1/2 and partially prevented CPT inhibition of LEC tube formation. In addition, CPT also inhibited protein expression and activities of Rac1 and Cdc42 but not RhoA. Expression of constitutively active Rac1 and Cdc42 concurrently, but not Rac1 or Cdc42 alone, conferred resistance to CPT inhibition of LEC tube formation. Taken together, the results suggest that CPT inhibits LEC tube formation, in part, by inhibiting VEGFR-3-mediated ERK1/2 phosphorylation and, in part, by inhibiting expression of the small GTPases.
Collapse
Affiliation(s)
- Yan Luo
- Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Tetraspanin CD151 maintains vascular stability by balancing the forces of cell adhesion and cytoskeletal tension. Blood 2011; 118:4274-84. [PMID: 21832275 DOI: 10.1182/blood-2011-03-339531] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Tetraspanin CD151 is highly expressed in endothelial cells and regulates pathologic angiogenesis. However, the mechanism by which CD151 promotes vascular morphogenesis and whether CD151 engages other vascular functions are unclear. Here we report that CD151 is required for maintaining endothelial capillary-like structures formed in vitro and the integrity of endothelial cell-cell and cell-matrix contacts in vivo. In addition, vascular permeability is markedly enhanced in the absence of CD151. As a global regulator of endothelial cell-cell and cell-matrix adhesions, CD151 is needed for the optimal functions of various cell adhesion proteins. The loss of CD151 elevates actin cytoskeletal traction by up-regulating RhoA signaling and diminishes actin cortical meshwork by down-regulating Rac1 activity. The inhibition of RhoA or activation of cAMP signaling stabilizes CD151-silenced or -null endothelial structure in vascular morphogenesis. Together, our data demonstrate that CD151 maintains vascular stability by promoting endothelial cell adhesions, especially cell-cell adhesion, and confining cytoskeletal tension.
Collapse
|
80
|
Cerebral cavernous malformations: from molecular pathogenesis to genetic counselling and clinical management. Eur J Hum Genet 2011; 20:134-40. [PMID: 21829231 DOI: 10.1038/ejhg.2011.155] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Cerebral cavernous (or capillary-venous) malformations (CCM) have a prevalence of about 0.1-0.5% in the general population. Genes mutated in CCM encode proteins that modulate junction formation between vascular endothelial cells. Mutations lead to the development of abnormal vascular structures.In this article, we review the clinical features, molecular and genetic basis of the disease, and management.
Collapse
|
81
|
Examining the role of Rac1 in tumor angiogenesis and growth: a clinically relevant RNAi-mediated approach. Angiogenesis 2011; 14:457-66. [PMID: 21789714 DOI: 10.1007/s10456-011-9229-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 07/15/2011] [Indexed: 12/30/2022]
Abstract
Angiogenesis, the sprouting of new blood vessels from the pre-existing vasculature, is a well established target in anti-cancer therapy. It is thought that the Rho GTPase Rac1 is required during vascular endothelial growth factor (VEGF)-mediated angiogenesis. In the present study, we have used a clinically relevant RNA interference approach to silence Rac1 expression. Human umbilical vein endothelial cells were transiently transfected with non-specific control siRNA (siNS) or Rac1 siRNA (siRac1) using electroporation or Lipofectamine 2000. Functional assays with transfected endothelial cells were performed to determine the effect of Rac1 knockdown on angiogenesis in vitro. Silencing of Rac1 inhibited VEGF-mediated tube formation, cell migration, invasion and proliferation. In addition, treatment with Rac1 siRNA inhibited angiogenesis in an in vivo Matrigel plug assay. Intratumoral injections of siRac1 almost completely inhibited the growth of grafted Neuro2a tumors and reduced tumor angiogenesis. Together, these data indicate that Rac1 is an important regulator of VEGF-mediated angiogenesis. Knockdown of Rac1 may represent an attractive approach to inhibit tumor angiogenesis and growth.
Collapse
|
82
|
Connolly M, Veale DJ, Fearon U. Acute serum amyloid A regulates cytoskeletal rearrangement, cell matrix interactions and promotes cell migration in rheumatoid arthritis. Ann Rheum Dis 2011; 70:1296-303. [PMID: 21482536 DOI: 10.1136/ard.2010.142240] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Serum amyloid A (A-SAA) is an acute-phase protein with cytokine-like properties implicated in the pathogenesis of rheumatoid arthritis (RA), atherosclerosis, diabetes and Alzheimer's disease. This study characterises the mechanism of A-SAA-induced cytoskeletal rearrangement and migration in synovial fibroblasts and microvascular endothelial cells (human dermal endothelial cells; HDEC). METHODS Immunohistology and immunofluorescence were used to examine αvβ3 and β1-integrins, filamentous actin (F-actin) and focal adhesion expression in rheumatoid arthritis synovial tissue (RAST) and rheumatoid arthritis synovial fibroblast cells (RASFC). A-SAA-induced αvβ3 and β1-integrin binding was measured by adhesion assay. Cytoskeletal rearrangement and ρ-GTPase activation following A-SAA stimulation was examined using dual immunofluorescent staining for F-actin/vinculin staining, pull down assays and immunoblotting for Cdc42 and RhoA. Cell growth, invasion/migration, angiogenesis and actin formation were examined in the presence or absence of specific Rac1 and Cdc42 inhibitors (NSC23766 and 187-1). RESULTS αvβ3, β1-integrin and F-actin predominantly localised to vascular endothelium and lining layer cells in RAST, compared with osteoarthritis and normal control synovial tissue. A-SAA significantly increased αvβ3 and β1 binding in RASFC. A-SAA induced cytoskeletal disassembly, loss of focal adhesions and filopodia formation in RASFC and HDEC. A-SAA significantly induced Cdc42 activation but failed to promote RhoA activation in HDEC and synovial fibroblast cells. Blockade of Rac-1 and Cdc42 inhibited A-SAA-induced cell growth, invasion/migration, actin cytoskeletal rearrangement and angiogenesis. CONCLUSIONS These data show a novel mechanism for A-SAA-induced cell migrational events in RA mediated via cytoskeletal signalling pathways.
Collapse
Affiliation(s)
- M Connolly
- Department of Rheumatology, Dublin Academic Medical Centre, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Ireland
| | | | | |
Collapse
|
83
|
Qi Y, Liu J, Wu X, Brakebusch C, Leitges M, Han Y, Corbett SA, Lowry SF, Graham AM, Li S. Cdc42 controls vascular network assembly through protein kinase Cι during embryonic vasculogenesis. Arterioscler Thromb Vasc Biol 2011; 31:1861-70. [PMID: 21659643 DOI: 10.1161/atvbaha.111.230144] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE The goal of this study was to determine the role of Cdc42 in embryonic vasculogenesis and the underlying mechanisms. METHODS AND RESULTS By using genetically modified mouse embryonic stem (ES) cells, we demonstrate that ablation of the Rho GTPase Cdc42 blocks vascular network assembly during embryoid body (EB) vasculogenesis without affecting endothelial lineage differentiation. Reexpression of Cdc42 in mutant EBs rescues the mutant phenotype, establishing an essential role for Cdc42 in vasculogenesis. Chimeric analysis revealed that the vascular phenotype is caused by inactivation of Cdc42 in endothelial cells rather than surrounding cells. Endothelial cells isolated from Cdc42-null EBs are defective in directional migration and network assembly. In addition, activation of atypical protein kinase Cι (PKCι) is abolished in Cdc42-null endothelial cells, and PKCι ablation phenocopies the vascular abnormalities of the Cdc42-null EBs. Moreover, the inhibitory phosphorylation of glycogen synthase kinase-3β (GSK-3β) at Ser9 depends on Cdc42 and PKCι, and expression of kinase-dead GSK-3β in Cdc42-null EBs promotes the formation of linear endothelial segments without branches. These results suggest that PKCι and GSK-3β are downstream effectors of Cdc42 during vascular morphogenesis. CONCLUSIONS Cdc42 controls vascular network assembly but not endothelial lineage differentiation by activating PKCι during embryonic vasculogenesis.
Collapse
Affiliation(s)
- Yanmei Qi
- Department of Surgery, Robert Wood Johnson Medical School-University of Medicine and Dentistry of New Jersey, New Brunswick, 08903-0019, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
84
|
RhoJ is an endothelial cell-restricted Rho GTPase that mediates vascular morphogenesis and is regulated by the transcription factor ERG. Blood 2011; 118:1145-53. [PMID: 21628409 DOI: 10.1182/blood-2010-10-315275] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
ERG is a member of the ETS transcription factor family that is highly enriched in endothelial cells (ECs). To further define the role of ERG in regulating EC function, we evaluated the effect of ERG knock-down on EC lumen formation in 3D collagen matrices. Blockade of ERG using siRNA completely interferes with EC lumen formation. Quantitative PCR (QPCR) was used to identify potential downstream gene targets of ERG. In particular, we identified RhoJ as the Rho GTPase family member that is closely related to Cdc42 as a target of ERG. Knockdown of ERG expression in ECs led to a 75% reduction in the expression of RhoJ. Chromatin immunoprecipitation and transactivation studies demonstrated that ERG could bind to functional sites in the proximal promoter of the RhoJ gene. Knock-down of RhoJ similarly resulted in a marked reduction in the ability of ECs to form lumens. Suppression of either ERG or RhoJ during EC lumen formation was associated with a marked increase in RhoA activation and a decrease in Rac1 and Cdc42 activation and their downstream effectors. Finally, in contrast to other Rho GTPases, RhoJ exhibits a highly EC-restricted expression pattern in several different tissues, including the brain, heart, lung, and liver.
Collapse
|
85
|
Xu K, Cleaver O. Tubulogenesis during blood vessel formation. Semin Cell Dev Biol 2011; 22:993-1004. [PMID: 21624487 DOI: 10.1016/j.semcdb.2011.05.001] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2011] [Accepted: 05/16/2011] [Indexed: 12/13/2022]
Abstract
The ability to form and maintain a functional system of contiguous hollow tubes is a critical feature of vascular endothelial cells (ECs). Lumen formation, or tubulogenesis, occurs in blood vessels during both vasculogenesis and angiogenesis in the embryo. Formation of vascular lumens takes place prior to the establishment of blood flow and to vascular remodeling which results in a characteristic hierarchical vessel organization. While epithelial lumen formation has received intense attention in past decades, more recent work has only just begun to elucidate the mechanisms controlling the initiation and morphogenesis of endothelial lumens. Studies using in vitro and in vivo models, including zebrafish and mammals, are beginning to paint an emerging picture of how blood vessels establish their characteristic morphology and become patent. In this article, we review and discuss the molecular and cellular mechanisms driving the formation of vascular tubes, primarily in vivo, and we compare and contrast proposed models for blood vessel lumen formation.
Collapse
Affiliation(s)
- Ke Xu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | | |
Collapse
|
86
|
Min JK, Park H, Choi HJ, Kim Y, Pyun BJ, Agrawal V, Song BW, Jeon J, Maeng YS, Rho SS, Shim S, Chai JH, Koo BK, Hong HJ, Yun CO, Choi C, Kim YM, Hwang KC, Kwon YG. The WNT antagonist Dickkopf2 promotes angiogenesis in rodent and human endothelial cells. J Clin Invest 2011; 121:1882-1893. [PMID: 21540552 PMCID: PMC3083777 DOI: 10.1172/jci42556] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Accepted: 02/16/2011] [Indexed: 12/23/2022] Open
Abstract
Neovessel formation is a complex process governed by the orchestrated action of multiple factors that regulate EC specification and dynamics within a growing vascular tree. These factors have been widely exploited to develop therapies for angiogenesis-related diseases such as diabetic retinopathy and tumor growth and metastasis. WNT signaling has been implicated in the regulation and development of the vascular system, but the detailed mechanism of this process remains unclear. Here, we report that Dickkopf1 (DKK1) and Dickkopf2 (DKK2), originally known as WNT antagonists, play opposite functional roles in regulating angiogenesis. DKK2 induced during EC morphogenesis promoted angiogenesis in cultured human endothelial cells and in in vivo assays using mice. Its structural homolog, DKK1, suppressed angiogenesis and was repressed upon induction of morphogenesis. Importantly, local injection of DKK2 protein significantly improved tissue repair, with enhanced neovascularization in animal models of both hind limb ischemia and myocardial infarction. We further showed that DKK2 stimulated filopodial dynamics and angiogenic sprouting of ECs via a signaling cascade involving LRP6-mediated APC/Asef2/Cdc42 activation. Thus, our findings demonstrate the distinct functions of DKK1 and DKK2 in controlling angiogenesis and suggest that DKK2 may be a viable therapeutic target in the treatment of ischemic vascular diseases.
Collapse
Affiliation(s)
- Jeong-Ki Min
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Hongryeol Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Hyun-Jung Choi
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Yonghak Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Bo-Jeong Pyun
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Vijayendra Agrawal
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Byeong-Wook Song
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Jongwook Jeon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Yong-Sun Maeng
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Seung-Sik Rho
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Sungbo Shim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Jin-Ho Chai
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Bon-Kyoung Koo
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Hyo Jeong Hong
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Chae-Ok Yun
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Chulhee Choi
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Young-Myoung Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Ki-Chul Hwang
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea.
Therapeutic Antibody Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea.
College of Pharmacy and Division of Life Science and Pharmaceuticals, Ewha Womans University, Seoul, Republic of Korea.
Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
Cell Signaling and Bioimaging Laboratory, Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea.
Department of Biological Science, Sookmyung Women’s University, Yongsan-Ku, Seoul, Republic of Korea.
Department of Biological Sciences, Seoul National University, Silim-dong, Gwanak-gu, Seoul, Republic of Korea.
Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
Vascular System Research Center, Kangwon National University, Kangwon-Do, Republic of Korea
| |
Collapse
|
87
|
Combinatory action of VEGFR2 and MAP kinase pathways maintains endothelial-cell integrity. Cell Res 2011; 21:1080-7. [PMID: 21423276 DOI: 10.1038/cr.2011.41] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Blood vessels normally maintain stereotyped lumen diameters and their stable structures are crucial for vascular function. However, very little is known about the molecular mechanisms controlling the maintenance of vessel diameters and the integrity of endothelial cells. We investigated this issue in zebrafish embryos by a chemical genetics approach. Small molecule libraries were screened using live Tg(kdrl:GRCFP)(zn1) transgenic embryos in which endothelial cells are specifically labeled with GFP. By analyzing the effects of compounds on the morphology and function of embryonic blood vessels after lumen formation, PP1, a putative Src kinase inhibitor, was identified as capable of specifically reducing vascular lumen size by interrupting endothelial-cell integrity. The inhibitory effect is not due to Src or general VEGF signaling inhibition because another Src inhibitor and Src morpholino as well as several VEGFR inhibitors failed to produce a similar phenotype. After profiling a panel of 22 representative mammalian kinases and surveying published data, we selected a few possible new candidates. Combinational analysis of these candidate kinase inhibitors established that PP1 induced endothelial collapse by inhibiting both the VEGFR2 and MAP kinase pathways. More importantly, combinatory use of two clinically approved drugs Dasatinib and Sunitinib produced the same phenotype. This is the first study to elucidate the pathways controlling maintenance of endothelial integrity using a chemical genetics approach, indicating that endothelial integrity is controlled by the combined action of the VEGFR2 and MAP kinase pathways. Our results also suggest the possible side effect of the combination of two anticancer drugs on the circulatory system.
Collapse
|
88
|
Xu K, Sacharidou A, Fu S, Chong DC, Skaug B, Chen ZJ, Davis GE, Cleaver O. Blood vessel tubulogenesis requires Rasip1 regulation of GTPase signaling. Dev Cell 2011; 20:526-39. [PMID: 21396893 DOI: 10.1016/j.devcel.2011.02.010] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Revised: 01/06/2011] [Accepted: 02/23/2011] [Indexed: 01/30/2023]
Abstract
Cardiovascular function depends on patent blood vessel formation by endothelial cells (ECs). However, the mechanisms underlying vascular "tubulogenesis" are only beginning to be unraveled. We show that endothelial tubulogenesis requires the Ras interacting protein 1, Rasip1, and its binding partner, the RhoGAP Arhgap29. Mice lacking Rasip1 fail to form patent lumens in all blood vessels, including the early endocardial tube. Rasipl null angioblasts fail to properly localize the polarity determinant Par3 and display defective cell polarity, resulting in mislocalized junctional complexes and loss of adhesion to extracellular matrix (ECM). Similarly, depletion of either Rasip1 or Arhgap29 in cultured ECs blocks in vitro lumen formation, fundamentally alters the cytoskeleton, and reduces integrin-dependent adhesion to ECM. These defects result from increased RhoA/ROCK/myosin II activity and blockade of Cdc42 and Rac1 signaling. This study identifies Rasip1 as a unique, endothelial-specific regulator of Rho GTPase signaling, which is essential for blood vessel morphogenesis.
Collapse
Affiliation(s)
- Ke Xu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Hanjaya-Putra D, Yee J, Ceci D, Truitt R, Yee D, Gerecht S. Vascular endothelial growth factor and substrate mechanics regulate in vitro tubulogenesis of endothelial progenitor cells. J Cell Mol Med 2011; 14:2436-47. [PMID: 19968735 PMCID: PMC3823161 DOI: 10.1111/j.1582-4934.2009.00981.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Endothelial progenitor cells (EPCs) in the circulatory system have been suggested to maintain vascular homeostasis and contribute to adult vascular regeneration and repair. These processes require that EPCs break down the extracellular matrix (ECM), migrate, differentiate and undergo tube morphogenesis. Evidently, the ECM plays a critical role by providing biochemical and biophysical cues that regulate cellular behaviour. Using a chemically and mechanically tunable hydrogel to study tube morphogenesis in vitro, we show that vascular endothelial growth factor (VEGF) and substrate mechanics co-regulate tubulogenesis of EPCs. High levels of VEGF are required to initiate tube morphogenesis and activate matrix metalloproteinases (MMPs), which enable EPC migration. Under these conditions, the elasticity of the substrate affects the progression of tube morphogenesis. With decreases in substrate stiffness, we observe decreased MMP expression while increased cellular elongation, with intracellular vacuole extension and coalescence to open lumen compartments. RNAi studies demonstrate that membrane type 1-MMP (MT1-MMP) is required to enable the movement of EPCs on the matrix and that EPCs sense matrix stiffness through signalling cascades leading to the activation of the RhoGTPase Cdc42. Collectively, these results suggest that coupled responses for VEGF stimulation and modulation of substrate stiffness are required to regulate tube morphogenesis of EPCs.
Collapse
Affiliation(s)
- Donny Hanjaya-Putra
- Department of Chemical and Biomolecular Engineering, Johns Hopkins Physical Science Oncology Center and Institute for NanoBio Technology, Johns Hopkins University, Baltimore, MD, USA
| | | | | | | | | | | |
Collapse
|
90
|
Davis GE, Stratman AN, Sacharidou A. Molecular Control of Vascular Tube Morphogenesis and Stabilization: Regulation by Extracellular Matrix, Matrix Metalloproteinases, and Endothelial Cell–Pericyte Interactions. BIOPHYSICAL REGULATION OF VASCULAR DIFFERENTIATION AND ASSEMBLY 2011. [DOI: 10.1007/978-1-4419-7835-6_2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
91
|
Davis GE, Stratman AN, Sacharidou A, Koh W. Molecular basis for endothelial lumen formation and tubulogenesis during vasculogenesis and angiogenic sprouting. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 288:101-65. [PMID: 21482411 DOI: 10.1016/b978-0-12-386041-5.00003-0] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Many studies reveal a fundamental role for extracellular matrix-mediated signaling through integrins and Rho GTPases as well as matrix metalloproteinases (MMPs) in the molecular control of vascular tube morphogenesis in three-dimensional (3D) tissue environments. Recent work has defined an endothelial cell (EC) lumen signaling complex of proteins that controls these vascular morphogenic events. These findings reveal a signaling interdependence between Cdc42 and MT1-MMP to control the 3D matrix-specific process of EC tubulogenesis. The EC tube formation process results in the creation of a network of proteolytically generated vascular guidance tunnels in 3D matrices that are utilized to remodel EC-lined tubes through EC motility and could facilitate processes such as flow-induced remodeling and arteriovenous EC sorting and differentiation. Within vascular guidance tunnels, key dynamic interactions occur between ECs and pericytes to affect vessel remodeling, diameter, and vascular basement membrane matrix assembly, a fundamental process necessary for endothelial tube maturation and stabilization. Thus, the EC lumen and tube formation mechanism coordinates the concomitant establishment of a network of vascular tubes within tunnel spaces to allow for flow responsiveness, EC-mural cell interactions, and vascular extracellular matrix assembly to control the development of the functional microcirculation.
Collapse
Affiliation(s)
- George E Davis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | | | | | | |
Collapse
|
92
|
Jain S, Gabunia K, Kelemen SE, Panetti TS, Autieri MV. The anti-inflammatory cytokine interleukin 19 is expressed by and angiogenic for human endothelial cells. Arterioscler Thromb Vasc Biol 2010; 31:167-75. [PMID: 20966397 DOI: 10.1161/atvbaha.110.214916] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To characterize the expression and function of interleukin (IL) 19, a recently described T-helper 2 anti-inflammatory IL, on endothelial cell (EC) pathophysiological features. METHODS AND RESULTS The expression and effects of anti-inflammatory ILs on EC activation and development of angiogenesis are uncharacterized. We demonstrate by immunohistochemistry and immunoblot that IL-19 is expressed in inflamed, but not normal, human coronary endothelium and can be induced in cultured human ECs by serum and basic fibroblast growth factor. IL-19 is mitogenic and chemotactic, and it promotes EC spreading. IL-19 activates the signaling proteins STAT3, p44/42, and Rac1. In functional ex vivo studies, IL-19 promotes cordlike structure formation of cultured ECs and enhances microvessel sprouting in the mouse aortic ring assay. IL-19 induces tube formation in gelatinous protein (Matrigel) plugs in vivo. CONCLUSIONS To our knowledge, these data are the first to report expression of the anti-inflammatory agent, IL-19, in ECs; and the first to indicate that IL-19 is mitogenic and chemotactic for ECs and can induce the angiogenic potential of ECs.
Collapse
Affiliation(s)
- Surbhi Jain
- Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | |
Collapse
|
93
|
Su SC, Maxwell SA, Bayless KJ. Annexin 2 regulates endothelial morphogenesis by controlling AKT activation and junctional integrity. J Biol Chem 2010; 285:40624-34. [PMID: 20947498 DOI: 10.1074/jbc.m110.157271] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Sprouting angiogenesis is a multistep process that involves endothelial cell activation, basement membrane degradation, proliferation, lumen formation, and stabilization. In this study, we identified annexin 2 as a regulator of endothelial morphogenesis using a three-dimensional in vitro model where sprouting angiogenesis was driven by sphingosine 1-phosphate and angiogenic growth factors. We observed that sphingosine 1-phosphate triggered annexin 2 translocation from the cytosol to the plasma membrane and its association with vascular endothelial (VE)-cadherin. In addition, annexin 2 depletion attenuated Akt activation, which was associated with increased phosphorylation of VE-cadherin and endothelial barrier leakage. Disrupting homotypic VE-cadherin interactions with EGTA, antibodies to the extracellular domain of VE-cadherin, or gene silencing all resulted in decreased Akt (but not Erk1/2) activation. Furthermore, expression of constitutively active Akt restored reduced endothelial sprouting responses observed with annexin 2 and VE-cadherin knockdown. Collectively, we report that annexin 2 regulates endothelial morphogenesis through an adherens junction-mediated pathway upstream of Akt.
Collapse
Affiliation(s)
- Shih-Chi Su
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, College Station, Texas 77843-1114, USA
| | | | | |
Collapse
|
94
|
Wang Y, Kaiser MS, Larson JD, Nasevicius A, Clark KJ, Wadman SA, Roberg-Perez SE, Ekker SC, Hackett PB, McGrail M, Essner JJ. Moesin1 and Ve-cadherin are required in endothelial cells during in vivo tubulogenesis. Development 2010; 137:3119-28. [PMID: 20736288 DOI: 10.1242/dev.048785] [Citation(s) in RCA: 156] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Endothelial tubulogenesis is a crucial step in the formation of functional blood vessels during angiogenesis and vasculogenesis. Here, we use in vivo imaging of living zebrafish embryos expressing fluorescent fusion proteins of beta-Actin, alpha-Catenin, and the ERM family member Moesin1 (Moesin a), to define a novel cord hollowing process that occurs during the initial stages of tubulogenesis in intersegmental vessels (ISVs) in the embryo. We show that the primary lumen elongates along cell junctions between at least two endothelial cells during embryonic angiogenesis. Moesin1-EGFP is enriched around structures that resemble intracellular vacuoles, which fuse with the luminal membrane during expansion of the primary lumen. Analysis of silent heart mutant embryos shows that initial lumen formation in the ISVs is not dependent on blood flow; however, stabilization of a newly formed lumen is dependent upon blood flow. Zebrafish moesin1 knockdown and cell transplantation experiments demonstrate that Moesin1 is required in the endothelial cells of the ISVs for in vivo lumen formation. Our analyses suggest that Moesin1 contributes to the maintenance of apical/basal cell polarity of the ISVs as defined by adherens junctions. Knockdown of the adherens junction protein Ve-cadherin disrupts formation of the apical membrane and lumen in a cell-autonomous manner. We suggest that Ve-cadherin and Moesin1 function to establish and maintain apical/basal polarity during multicellular lumen formation in the ISVs.
Collapse
Affiliation(s)
- Ying Wang
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA 50011, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Kaully T, Kaufman-Francis K, Lesman A, Levenberg S. Vascularization--the conduit to viable engineered tissues. TISSUE ENGINEERING PART B-REVIEWS 2010; 15:159-69. [PMID: 19309238 DOI: 10.1089/ten.teb.2008.0193] [Citation(s) in RCA: 210] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Long-term viability of thick three-dimensional engineered tissue constructs is a major challenge. Addressing it requires development of vessel-like network that will allow the survival of the construct in vitro and its integration in vivo owing to improved vascularization after implantation. Resulting from work of various research groups, several approaches were developed aiming engineered tissue vascularization: (1) embodiment of angiogenesis growth factors in the polymeric scaffolds for prolonged release, (2) coculture of endothelial cells with target tissue cells and angiogenesis signaling cells, (3) use of microfabrication methods for creating designed channels for allowing nutrients to flow and/or for directing endothelial cells attachment, and (4) decellularization of organs and blood vessels for creating extracellular matrix. A synergistic effect is expected by combining several of these approaches as already demonstrated in some of the latest studies. Current paper reviews the progress in each approach and recent achievements toward vascularization of engineered tissues.
Collapse
Affiliation(s)
- Tamar Kaully
- Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | | | | | | |
Collapse
|
96
|
Lampugnani MG, Orsenigo F, Rudini N, Maddaluno L, Boulday G, Chapon F, Dejana E. CCM1 regulates vascular-lumen organization by inducing endothelial polarity. J Cell Sci 2010; 123:1073-80. [PMID: 20332120 DOI: 10.1242/jcs.059329] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Little is known about the molecular mechanisms that regulate the organization of vascular lumen. In this paper we show that lumen formation correlates with endothelial polarization. Adherens junctions (AJs) and VE-cadherin (VEC, encoded by CDH5) are required for endothelial apicobasal polarity in vitro and during embryonic development. Silencing of CDH5 gene expression leads to abrogation of endothelial polarity accompanied by strong alterations in lumenal structure. VEC co-distributes with members of the Par polarity complex (Par3 and PKCzeta) and is needed for activation of PKCzeta. CCM1 is encoded by the CCM1 gene, which is mutated in 60% of patients affected by cerebral cavernous malformation (CCM). The protein interacts with VEC and directs AJ organization and AJ association with the polarity complex, both in cell-culture models and in human CCM1 lesions. Both VEC and CCM1 control Rap1 concentration at cell-cell junctions. We propose that VEC, CCM1 and Rap1 form a signaling complex. In the absence of any of these proteins, AJs are dismantled, cell polarity is lost and vascular lumenal structure is severely altered.
Collapse
|
97
|
|
98
|
Tawa H, Rikitake Y, Takahashi M, Amano H, Miyata M, Satomi-Kobayashi S, Kinugasa M, Nagamatsu Y, Majima T, Ogita H, Miyoshi J, Hirata KI, Takai Y. Role of Afadin in Vascular Endothelial Growth Factor– and Sphingosine 1-Phosphate–Induced Angiogenesis. Circ Res 2010; 106:1731-42. [DOI: 10.1161/circresaha.110.216747] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale
:
Angiogenesis contributes to physiological and pathological conditions, including atherosclerosis. The Rap1 small G protein regulates vascular integrity and angiogenesis. However, little is known about the effectors of Rap1 involved in angiogenesis. It is not known whether afadin, an adherens junction protein that connects immunoglobulin-like adhesion molecule nectins to the actin cytoskeleton and binds activated Rap1, plays a role in angiogenesis.
Objective
:
We investigated the role of endothelial afadin in angiogenesis and attempted to clarify the underlying molecular mechanism.
Methods and Results
:
Treatment of human umbilical vein endothelial cells (HUVECs) with vascular endothelial growth factor (VEGF) and sphingosine 1-phosphate (S1P) induced the activation of Rap1. Activated Rap1 regulated intracellular localization of afadin. Knockdown of Rap1 or afadin by small interfering RNA inhibited the VEGF- and S1P-induced capillary-like network formation, migration, and proliferation, and increased the serum deprivation-induced apoptosis of HUVECs. Knockdown of Rap1 or afadin decreased the accumulation of adherens and tight junction proteins to the cell–cell contact sites. Rap1 regulated the interaction between afadin and phosphatidylinositol 3-kinase (PI3K), recruitment of the afadin–PI3K complex to the leading edge, and the activation of Akt, indicating the involvement of Rap1 and afadin in the PI3K–Akt signaling pathway. Binding of afadin to Rap1 regulated the activity of Rap1 in a positive-feedback manner. In vivo, conditional deletion of afadin in mouse vascular endothelium using a Cre-loxP system impaired the VEGF- and S1P-induced angiogenesis.
Conclusions
:
These results demonstrate a novel molecular mechanism by which Rap1 and afadin regulate the VEGF- and S1P-induced angiogenesis.
Collapse
Affiliation(s)
- Hideto Tawa
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Yoshiyuki Rikitake
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Motonori Takahashi
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Hisayuki Amano
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Muneaki Miyata
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Seimi Satomi-Kobayashi
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Mitsuo Kinugasa
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Yuichi Nagamatsu
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Takashi Majima
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Hisakazu Ogita
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Jun Miyoshi
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Ken-ichi Hirata
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| | - Yoshimi Takai
- From the Divisions of Molecular and Cellular Biology (H.T., Y.R., M.T., H.A., M.M., S.S.-K., M.K., Y.N., T.M., H.O., Y.T.) and Signal Transduction (Y.R., M.M.), Department of Biochemistry and Molecular Biology; and Division of Cardiovascular Medicine (H.T., Y.R., M.T., S.S.-K., M.K., K.-i.H.), Department of Internal Medicine, Kobe University Graduate School of Medicine; and Department of Molecular Biology (T.M., J.M.), Osaka Medical Center for Cancer and Cardiovascular Diseases, Japan
| |
Collapse
|
99
|
Abstract
Angiogenesis, or the formation of new blood vessels from the preexisting vasculature, is a key component in numerous physiologic and pathologic responses and has broad impact in many medical and surgical specialties. In this review, we discuss the key cellular steps that lead to the neovascularization of tissues and highlight the main molecular mechanisms and mediators in this process. We include discussions on proteolytic enzymes, cell-matrix interactions, and pertinent cell signaling pathways and end with a survey of the mechanisms that lead to the stabilization and maturation of neovasculatures.
Collapse
|
100
|
Chen L, Chu W, Xu Y, Chen P, Lao F, Sun Q, Feng X, Han D. Time-series investigation of fused vesicles in microvessel endothelial cells with atomic force microscopy. Microsc Res Tech 2010; 73:152-9. [PMID: 19725060 DOI: 10.1002/jemt.20766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Vesicles or caveolae within endothelial cells, fusing together to form vacuolar organelles, are implicated in macromolecular transport and cellular element transmigration across the blood-brain barrier (BBB) during inflammation and ischemia. Vacuolar organelles have been described by transmission electron microscopy and immunofluorescence, but the details of their dynamics have not been well addressed yet. Herein, by using tapping mode atomic force microscopy (AFM), we observed the time-series changes of fused vesicles within the serum-free cultured rat cerebral microvessel endothelial cells. The fused vesicles were certainly proved by fluorescent staining of Fm4-64 combining simultaneous AFM imaging, as well as the field emission scanning electron microscopy technique. And energy dispersive spectrum results additionally implied that there may be specific structure and compositions around the vesicle region. These results indicate that increased vesicles in BBB may contribute to the formation of fused vesicles and a higher probability to construct the trans-endothelial channel across endothelium layer. Furthermore, the AFM application may open up a new approach to investigate the details of transcellular process by fused vesicles.
Collapse
Affiliation(s)
- Long Chen
- National Center for Nanoscience and Technology, Beijing 100190, China
| | | | | | | | | | | | | | | |
Collapse
|