101
|
Neuropilin 1 sequestration by neuropathogenic mutant glycyl-tRNA synthetase is permissive to vascular homeostasis. Sci Rep 2017; 7:9216. [PMID: 28835631 PMCID: PMC5569042 DOI: 10.1038/s41598-017-10005-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 07/27/2017] [Indexed: 12/11/2022] Open
Abstract
The mechanism by which dominantly inherited mutations in the housekeeping gene GARS, which encodes glycyl-tRNA synthetase (GlyRS), mediate selective peripheral nerve toxicity resulting in Charcot-Marie-Tooth disease type 2D (CMT2D) is still largely unresolved. The transmembrane receptor protein neuropilin 1 (Nrp1) was recently identified as an aberrant extracellular binding partner of mutant GlyRS. Formation of the Nrp1/mutant GlyRS complex antagonises Nrp1 interaction with one of its main natural ligands, vascular endothelial growth factor-A (VEGF-A), contributing to neurodegeneration. However, reduced extracellular binding of VEGF-A to Nrp1 is known to disrupt post-natal blood vessel development and growth. We therefore analysed the vascular system at early and late symptomatic time points in CMT2D mouse muscles, retina, and sciatic nerve, as well as in embryonic hindbrain. Mutant tissues show no difference in blood vessel diameter, density/growth, and branching from embryonic development to three months, spanning the duration over which numerous sensory and neuromuscular phenotypes manifest. Our findings indicate that mutant GlyRS-mediated disruption of Nrp1/VEGF-A signalling is permissive to maturation and maintenance of the vasculature in CMT2D mice.
Collapse
|
102
|
Zhao M, Hu Y, Jin J, Yu Y, Zhang S, Cao J, Zhai Y, Wei R, Shou J, Cai W, Liu S, Yang X, Xu GT, Yang J, Corry DB, Su SB, Liu X, Yang T. Interleukin 37 promotes angiogenesis through TGF-β signaling. Sci Rep 2017; 7:6113. [PMID: 28733640 PMCID: PMC5522482 DOI: 10.1038/s41598-017-06124-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 05/30/2017] [Indexed: 01/10/2023] Open
Abstract
IL-37 is a novel pro-angiogenic cytokine that potently promotes endothelial cell activation and pathological angiogenesis in our previous study, but the mechanisms behind the pro-angiogenic effect of IL-37 are less well understood. Extending our observations, we found that TGF-β interacts with IL-37, and potently enhances the binding affinity of IL-37 to the ALK1 receptor complex, thus allowing IL-37 to signal through ALK1 to activate pro-angiogenic responses. We further show that TGF-β and ALK1 are required in IL-37 induced pro-angiogenic response in ECs and in the mouse model of Matrigel plug and oxygen-induced retinopathy. The result suggests that IL-37 induces pro-angiogenic responses through TGF-β, which may act as the bridging molecule that mediates IL-37 binding to the TGF-β receptor complex.
Collapse
Affiliation(s)
- Mengmeng Zhao
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yongguang Hu
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jiayi Jin
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ying Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Shanshan Zhang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingjing Cao
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yuanfen Zhai
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Rongbin Wei
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Juanjuan Shou
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenping Cai
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shangfeng Liu
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoping Yang
- Johns Hopkins University School of Medicine, Baltimore, United States
| | - Guo-Tong Xu
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianhua Yang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - David B Corry
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, United States
| | - Shao Bo Su
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.,State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xialin Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Tianshu Yang
- Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
103
|
Benn A, Hiepen C, Osterland M, Schütte C, Zwijsen A, Knaus P. Role of bone morphogenetic proteins in sprouting angiogenesis: differential BMP receptor-dependent signaling pathways balance stalk vs. tip cell competence. FASEB J 2017; 31:4720-4733. [PMID: 28733457 PMCID: PMC5636702 DOI: 10.1096/fj.201700193rr] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/27/2017] [Indexed: 01/04/2023]
Abstract
Before the onset of sprouting angiogenesis, the endothelium is prepatterned for the positioning of tip and stalk cells. Both cell identities are not static, as endothelial cells (ECs) constantly compete for the tip cell position in a dynamic fashion. Here, we show that both bone morphogenetic protein 2 (BMP2) and BMP6 are proangiogenic in vitro and ex vivo and that the BMP type I receptors, activin receptor-like kinase 3 (ALK3) and ALK2, play crucial and distinct roles in this process. BMP2 activates the expression of tip cell-associated genes, such as delta-like ligand 4 (DLL4) and kinase insert domain receptor (KDR), and p38-heat shock protein 27 (HSP27)-dependent cell migration, thereby generating tip cell competence. Whereas BMP6 also triggers collective cell migration via the p38-HSP27 signaling axis, BMP6 induces in addition SMAD1/5 signaling, thereby promoting the expression of stalk cell-associated genes, such as hairy and enhancer of split 1 (HES1) and fms-like tyrosine kinase 1 (FLT1). Specifically, ALK3 is required for sprouting from HUVEC spheroids, whereas ALK2 represses sprout formation. We demonstrate that expression levels and respective complex formation of BMP type I receptors in ECs determine stalk vs. tip cell identity, thus contributing to endothelial plasticity during sprouting angiogenesis. As antiangiogenic monotherapies that target the VEGF or ALK1 pathways have not fulfilled efficacy objectives in clinical trials, the selective targeting of the ALK2/3 pathways may be an attractive new approach.-Benn, A., Hiepen, C., Osterland, M., Schütte, C., Zwijsen, A., Knaus, P. Role of bone morphogenetic proteins in sprouting angiogenesis: differential BMP receptor-dependent signaling pathways balance stalk vs. tip cell competence.
Collapse
Affiliation(s)
- Andreas Benn
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Deutsche Forschungsgemeinschaft (DFG) Graduate School 1093, Berlin School of Integrative Oncology, Berlin, Germany.,DFG Graduate School 203, Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany.,Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, KU Leuven, Leuven, Belgium.,Department of Human Genetics, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Christian Hiepen
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,DFG Graduate School 203, Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| | - Marc Osterland
- Zuse Institute Berlin, Berlin, Germany.,Institute for Mathematics, Freie Universität Berlin, Berlin, Germany
| | - Christof Schütte
- Zuse Institute Berlin, Berlin, Germany.,Institute for Mathematics, Freie Universität Berlin, Berlin, Germany
| | - An Zwijsen
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, KU Leuven, Leuven, Belgium.,Department of Human Genetics, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; .,Deutsche Forschungsgemeinschaft (DFG) Graduate School 1093, Berlin School of Integrative Oncology, Berlin, Germany.,DFG Graduate School 203, Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
104
|
Targeting tumour vasculature by inhibiting activin receptor-like kinase (ALK)1 function. Biochem Soc Trans 2017; 44:1142-9. [PMID: 27528762 DOI: 10.1042/bst20160093] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Indexed: 12/23/2022]
Abstract
Angiogenesis is a hallmark of cancer and is now a validated therapeutic target in the clinical setting. Despite the initial success, anti-angiogenic compounds impinging on the vascular endothelial growth factor (VEGF) pathway display limited survival benefits in patients and resistance often develops due to activation of alternative pathways. Thus, finding and validating new targets is highly warranted. Activin receptor-like kinase (ALK)1 is a transforming growth factor beta (TGF-β) type I receptor predominantly expressed in actively proliferating endothelial cells (ECs). ALK1 has been shown to play a pivotal role in regulating angiogenesis by binding to bone morphogenetic protein (BMP)9 and 10. Two main pharmacological inhibitors, an ALK1-Fc fusion protein (Dalantercept/ACE-041) and a fully human antibody against the extracellular domain of ALK1 (PF-03446962) are currently under clinical development. Herein, we briefly recapitulate the role of ALK1 in blood vessel formation and the current status of the preclinical and clinical studies on inhibition of ALK1 signalling as an anti-angiogenic strategy. Future directions in terms of new combination regimens will also be presented.
Collapse
|
105
|
Vascular heterogeneity and specialization in development and disease. Nat Rev Mol Cell Biol 2017; 18:477-494. [PMID: 28537573 DOI: 10.1038/nrm.2017.36] [Citation(s) in RCA: 425] [Impact Index Per Article: 53.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Blood and lymphatic vessels pervade almost all body tissues and have numerous essential roles in physiology and disease. The inner lining of these networks is formed by a single layer of endothelial cells, which is specialized according to the needs of the tissue that it supplies. Whereas the general mechanisms of blood and lymphatic vessel development are being defined with increasing molecular precision, studies of the processes of endothelial specialization remain mostly descriptive. Recent insights from genetic animal models illuminate how endothelial cells interact with each other and with their tissue environment, providing paradigms for vessel type- and organ-specific endothelial differentiation. Delineating these governing principles will be crucial for understanding how tissues develop and maintain, and how their function becomes abnormal in disease.
Collapse
|
106
|
Jin Y, Muhl L, Burmakin M, Wang Y, Duchez AC, Betsholtz C, Arthur HM, Jakobsson L. Endoglin prevents vascular malformation by regulating flow-induced cell migration and specification through VEGFR2 signalling. Nat Cell Biol 2017; 19:639-652. [PMID: 28530660 PMCID: PMC5467724 DOI: 10.1038/ncb3534] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 04/18/2017] [Indexed: 12/15/2022]
Abstract
Loss-of-function (LOF) mutations in the endothelial cell (EC) enriched gene endoglin (ENG) causes the human disease hereditary haemorrhagic telangiectasia-1, characterized by vascular malformations promoted by vascular endothelial growth factor A (VEGFA). How ENG deficiency alters EC behaviour to trigger these anomalies is not understood. Mosaic ENG deletion in the postnatal mouse rendered Eng LOF ECs insensitive to flow-mediated venous to arterial migration. Eng LOF ECs retained within arterioles acquired venous characteristics and secondary ENG-independent proliferation resulting in arterio-venous malformation (AVM). Analysis following simultaneous Eng LOF and overexpression (OE) revealed that ENG OE ECs dominate tip cell positions and home preferentially to arteries. ENG knock-down altered VEGFA-mediated VEGFR2 kinetics and promoted AKT signalling. Blockage of PI3K/AKT partly normalised flow-directed migration of ENG LOF ECs in vitro and reduced the severity of AVM in vivo. This demonstrates the requirement of ENG in flow-mediated migration and modulation of VEGFR2 signalling in vascular patterning.
Collapse
Affiliation(s)
- Yi Jin
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77 Stockholm, Sweden
| | - Lars Muhl
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77 Stockholm, Sweden
| | - Mikhail Burmakin
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77 Stockholm, Sweden
| | - Yixin Wang
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77 Stockholm, Sweden
| | - Anne-Claire Duchez
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77 Stockholm, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden.,Integrated Cardio Metabolic Centre (ICMC), Karolinska Institutet, Novum, Blickagången 6, SE14157 Huddinge, Sweden
| | - Helen M Arthur
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Lars Jakobsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77 Stockholm, Sweden
| |
Collapse
|
107
|
Siqueira M, Francis D, Gisbert D, Gomes FCA, Stipursky J. Radial Glia Cells Control Angiogenesis in the Developing Cerebral Cortex Through TGF-β1 Signaling. Mol Neurobiol 2017; 55:3660-3675. [PMID: 28523566 DOI: 10.1007/s12035-017-0557-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/12/2017] [Indexed: 10/19/2022]
Abstract
Neuroangiogenesis in the developing central nervous system is controlled by interactions between endothelial cells (ECs) and radial glia (RG) neural stem cells, although RG-derived molecules implicated in these events are not fully known. Here, we investigated the role of RG-secreted TGF-β1, in angiogenesis in the developing cerebral cortex. By isolation of murine microcapillary brain endothelial cells (MBECs), we demonstrate that conditioned medium from RG cultures (RG-CM) promoted MBEC migration and formation of vessel-like structures in vitro, in a TGF-β1-dependent manner. These events were followed by endothelial regulation of GPR124 and BAI-1 gene expression by RG-CM. Proteome profile of RG-CM identified angiogenesis-related molecules IGFBP2/3, osteopontin, endostatin, SDF1, fractalkine, TIMP1/4, Ang-1, pentraxin3, and Cyr61, some of them modulated by TGF-β1 induction. In vivo gain and loss of function assays targeting RG cells demonstrates a specific TGF-β1-dependent control of blood vessels branching in the cerebral cortex. Together, our results point to TGF-β1 signaling pathway as a potential mediator of the RG-EC interactions and shed light to the key role of RG in paving the brain vascular network.
Collapse
Affiliation(s)
- Michele Siqueira
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Daniel Francis
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Diego Gisbert
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Joice Stipursky
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil. .,Laboratório de Neurobiologia Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro - Centro de Ciências da Saúde, Bloco F, Sala F15, Ilha do Fundão, Rio de Janeiro, RJ, 21949-902, Brazil.
| |
Collapse
|
108
|
Fantin A, Lampropoulou A, Senatore V, Brash JT, Prahst C, Lange CA, Liyanage SE, Raimondi C, Bainbridge JW, Augustin HG, Ruhrberg C. VEGF165-induced vascular permeability requires NRP1 for ABL-mediated SRC family kinase activation. J Exp Med 2017; 214:1049-1064. [PMID: 28289053 PMCID: PMC5379968 DOI: 10.1084/jem.20160311] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 12/07/2016] [Accepted: 01/20/2017] [Indexed: 01/13/2023] Open
Abstract
Fantin et al. show that the VEGF isoform VEGF165 signals through a complex of VEGFR2 and NRP1, in which the NRP1 cytoplasmic domain promotes the ABL-mediated activation of SRC family kinases to evoke a hyperpermeability response, a known cause of pathological edema. The vascular endothelial growth factor (VEGF) isoform VEGF165 stimulates vascular growth and hyperpermeability. Whereas blood vessel growth is essential to sustain organ health, chronic hyperpermeability causes damaging tissue edema. By combining in vivo and tissue culture models, we show here that VEGF165-induced vascular leakage requires both VEGFR2 and NRP1, including the VEGF164-binding site of NRP1 and the NRP1 cytoplasmic domain (NCD), but not the known NCD interactor GIPC1. In the VEGF165-bound receptor complex, the NCD promotes ABL kinase activation, which in turn is required to activate VEGFR2-recruited SRC family kinases (SFKs). These results elucidate the receptor complex and signaling hierarchy of downstream kinases that transduce the permeability response to VEGF165. In a mouse model with choroidal neovascularisation akin to age-related macular degeneration, NCD loss attenuated vessel leakage without affecting neovascularisation. These findings raise the possibility that targeting NRP1 or its NCD interactors may be a useful therapeutic strategy in neovascular disease to reduce VEGF165-induced edema without compromising vessel growth.
Collapse
Affiliation(s)
- Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | | | - Valentina Senatore
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - James T Brash
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - Claudia Prahst
- Yale Cardiovascular Research Center, New Haven, CT 06511
| | - Clemens A Lange
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - Sidath E Liyanage
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - Claudio Raimondi
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - James W Bainbridge
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), 69120 Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis (CBTM), Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, London EC1V 9EL, England, UK
| |
Collapse
|
109
|
Lee HW, Chong DC, Ola R, Dunworth WP, Meadows S, Ka J, Kaartinen VM, Qyang Y, Cleaver O, Bautch VL, Eichmann A, Jin SW. Alk2/ACVR1 and Alk3/BMPR1A Provide Essential Function for Bone Morphogenetic Protein-Induced Retinal Angiogenesis. Arterioscler Thromb Vasc Biol 2017; 37:657-663. [PMID: 28232325 DOI: 10.1161/atvbaha.116.308422] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 02/09/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Increasing evidence suggests that bone morphogenetic protein (BMP) signaling regulates angiogenesis. Here, we aimed to define the function of BMP receptors in regulating early postnatal angiogenesis by analysis of inducible, endothelial-specific deletion of the BMP receptor components Bmpr2 (BMP type 2 receptor), Alk1 (activin receptor-like kinase 1), Alk2, and Alk3 in mouse retinal vessels. APPROACH AND RESULTS Expression analysis of several BMP ligands showed that proangiogenic BMP ligands are highly expressed in postnatal retinas. Consistently, BMP receptors are also strongly expressed in retina with a distinct pattern. To assess the function of BMP signaling in retinal angiogenesis, we first generated mice carrying an endothelial-specific inducible deletion of Bmpr2. Postnatal deletion of Bmpr2 in endothelial cells substantially decreased the number of angiogenic sprouts at the vascular front and branch points behind the front, leading to attenuated radial expansion. To identify critical BMPR1s (BMP type 1 receptors) associated with BMPR2 in retinal angiogenesis, we generated endothelial-specific inducible deletion of 3 BMPR1s abundantly expressed in endothelial cells and analyzed the respective phenotypes. Among these, endothelial-specific deletion of either Alk2/acvr1 or Alk3/Bmpr1a caused a delay in radial expansion, reminiscent of vascular defects associated with postnatal endothelial-specific deletion of BMPR2, suggesting that ALK2/ACVR1 and ALK3/BMPR1A are likely to be the critical BMPR1s necessary for proangiogenic BMP signaling in retinal vessels. CONCLUSIONS Our data identify BMP signaling mediated by coordination of ALK2/ACVR1, ALK3/BMPR1A, and BMPR2 as an essential proangiogenic cue for retinal vessels.
Collapse
Affiliation(s)
- Heon-Woo Lee
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (H.-W.L., R.O., W.P.D., Y.Q., A.E., S.-W.J.); Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill (D.C.C., V.L.B.); Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX (S.M., O.C.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (J.K., S.-W.J.); and Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor (V.M.K.)
| | - Diana C Chong
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (H.-W.L., R.O., W.P.D., Y.Q., A.E., S.-W.J.); Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill (D.C.C., V.L.B.); Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX (S.M., O.C.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (J.K., S.-W.J.); and Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor (V.M.K.)
| | - Roxana Ola
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (H.-W.L., R.O., W.P.D., Y.Q., A.E., S.-W.J.); Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill (D.C.C., V.L.B.); Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX (S.M., O.C.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (J.K., S.-W.J.); and Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor (V.M.K.)
| | - William P Dunworth
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (H.-W.L., R.O., W.P.D., Y.Q., A.E., S.-W.J.); Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill (D.C.C., V.L.B.); Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX (S.M., O.C.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (J.K., S.-W.J.); and Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor (V.M.K.)
| | - Stryder Meadows
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (H.-W.L., R.O., W.P.D., Y.Q., A.E., S.-W.J.); Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill (D.C.C., V.L.B.); Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX (S.M., O.C.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (J.K., S.-W.J.); and Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor (V.M.K.)
| | - Jun Ka
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (H.-W.L., R.O., W.P.D., Y.Q., A.E., S.-W.J.); Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill (D.C.C., V.L.B.); Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX (S.M., O.C.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (J.K., S.-W.J.); and Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor (V.M.K.)
| | - Vesa M Kaartinen
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (H.-W.L., R.O., W.P.D., Y.Q., A.E., S.-W.J.); Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill (D.C.C., V.L.B.); Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX (S.M., O.C.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (J.K., S.-W.J.); and Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor (V.M.K.)
| | - Yibing Qyang
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (H.-W.L., R.O., W.P.D., Y.Q., A.E., S.-W.J.); Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill (D.C.C., V.L.B.); Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX (S.M., O.C.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (J.K., S.-W.J.); and Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor (V.M.K.)
| | - Ondine Cleaver
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (H.-W.L., R.O., W.P.D., Y.Q., A.E., S.-W.J.); Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill (D.C.C., V.L.B.); Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX (S.M., O.C.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (J.K., S.-W.J.); and Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor (V.M.K.)
| | - Victoria L Bautch
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (H.-W.L., R.O., W.P.D., Y.Q., A.E., S.-W.J.); Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill (D.C.C., V.L.B.); Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX (S.M., O.C.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (J.K., S.-W.J.); and Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor (V.M.K.).
| | - Anne Eichmann
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (H.-W.L., R.O., W.P.D., Y.Q., A.E., S.-W.J.); Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill (D.C.C., V.L.B.); Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX (S.M., O.C.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (J.K., S.-W.J.); and Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor (V.M.K.)
| | - Suk-Won Jin
- From the Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT (H.-W.L., R.O., W.P.D., Y.Q., A.E., S.-W.J.); Department of Biology and McAllister Heart Institute, University of North Carolina, Chapel Hill (D.C.C., V.L.B.); Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX (S.M., O.C.); School of Life Sciences and Cell Logistics Research Center, Gwangju Institute of Science and Technology, Korea (J.K., S.-W.J.); and Department of Biologic & Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor (V.M.K.)
| |
Collapse
|
110
|
Cho H, Sengupta S, Jeon SSH, Hur W, Choi HG, Seo HS, Lee BJ, Kim JH, Chung M, Jeon NL, Kim ND, Sim T. Identification of the First Selective Activin Receptor-Like Kinase 1 Inhibitor, a Reversible Version of L-783277. J Med Chem 2017; 60:1495-1508. [PMID: 28103025 DOI: 10.1021/acs.jmedchem.6b01679] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We synthesized 1 (San78-130), a reversible version of L-783277, as a selective and potent ALK1 inhibitor. Our study showed that 1 possesses great kinase selectivity against a panel of 342 kinases and more potent activity against ALK1 than L-783277. Among the six ALK isotypes (ALK1-6), ALK1 is most significantly inhibited by compound 1. Compound 1 suppresses the BMP9-induced Smad1/5 pathway by mainly inhibiting ALK1 in C2C12 cells. Our molecular dynamics simulations suggest that H-bonding interaction between the C-4' hydroxyl group of 1 and Arg334 of ALK1 substantially contributes to the ALK1 inhibition. To the best of our knowledge, 1 is the first selective ALK1 inhibitor. Furthermore, compound 1 promoted angiogenesis in both endothelial tube formation and microfluidic chip based 3D angiogenesis assays, suggesting that 1 could be a lead compound for therapeutic angiogenesis agents. Our study may provide an insight into designing selective and potent inhibitors against ALK1.
Collapse
Affiliation(s)
- Hanna Cho
- KU-KIST Graduate School of Converging Science and Technology, Korea University , 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Sandip Sengupta
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Sean S H Jeon
- KU-KIST Graduate School of Converging Science and Technology, Korea University , 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Wooyoung Hur
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hwan Geun Choi
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Hong-Seog Seo
- KU-KIST Graduate School of Converging Science and Technology, Korea University , 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea.,Cardiovascular Center, Korea University Guro Hospital , 80 Guro-dong, Guro-gu, Seoul 152-703, Republic of Korea
| | - Byung Joo Lee
- Fight Against Angiogenesis-related Blindness Laboratory, Clinical Research Institute, Seoul National University Hospital , 101, Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University , 103, Daehakro, Jongro-gu, Seoul 03080, Republic of Korea
| | - Jeong Hun Kim
- Fight Against Angiogenesis-related Blindness Laboratory, Clinical Research Institute, Seoul National University Hospital , 101, Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea.,Department of Biomedical Sciences, College of Medicine, Seoul National University , 103, Daehakro, Jongro-gu, Seoul 03080, Republic of Korea.,Department of Ophthalmology, College of Medicine, Seoul National University , 101, Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Minhwan Chung
- Mechanical Engineering, Seoul National University , 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Noo Li Jeon
- Mechanical Engineering, Seoul National University , 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Republic of Korea
| | - Nam Doo Kim
- Daegu-Gyeongbuk Medical Innovation Foundation , 2387 dalgubeol-daero, Suseong-gu, Daegu 42019, Republic of Korea
| | - Taebo Sim
- KU-KIST Graduate School of Converging Science and Technology, Korea University , 145 Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea.,Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST) , 5 Hwarangro 14-gil, Seongbuk-gu, Seoul 02792, Republic of Korea
| |
Collapse
|
111
|
Fernández-Robredo P, Selvam S, Powner MB, Sim DA, Fruttiger M. Neuropilin 1 Involvement in Choroidal and Retinal Neovascularisation. PLoS One 2017; 12:e0169865. [PMID: 28107458 PMCID: PMC5249189 DOI: 10.1371/journal.pone.0169865] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 12/23/2016] [Indexed: 11/30/2022] Open
Abstract
PURPOSE Inhibiting VEGF is the gold standard treatment for neovascular age-related macular degeneration (AMD). It is also effective in preventing retinal oedema and neovascularisation (NV) in diabetic retinopathy (DR) and retinal vein occlusions (RVO). Neuropilin 1 (Nrp1) is a co-receptor for VEGF and many other growth factors, and therefore a possible alternative drug target in intra ocular neovascular disease. Here we assessed choroidal and retinal NV in an inducible, endothelial specific knock out model for Nrp1. METHODS Crossing Nrp1 floxed mice with Pdgfb-CreERT2 mice produced tamoxifen-inducible, endothelial specific Nrp1 knock out mice (Nrp1ΔEC) and Cre-negative, control littermates. Cre-recombinase activity was confirmed in the Ai3(RCL-EYFP) reporter strain. Animals were subjected to laser-induced CNV (532 nm) and spectral domain-optical coherence tomography (SD-OCT) was performed immediately after laser and at day 7. Fluorescein angiography (FA) evaluated leakage and postmortem lectin staining in flat mounted RPE/choroid complexes was also used to measure CNV. Furthermore, retinal neovascularisation in the oxygen induced retinopathy (OIR) model was assessed by immunohistochemistry in retinal flatmounts. RESULTS In vivo FA, OCT and post-mortem lectin staining showed a statistically significant reduction in leakage (p<0.05), CNV volume (p<0.05) and CNV area (p<0.05) in the Nrp1ΔEC mice compared to their Cre-negative littermates. Also the OIR model showed reduced retinal NV in the mutant animals compared to wild types (p<0.001). CONCLUSION We have demonstrated reduced choroidal and retinal NV in animals that lack endothelial Nrp1, confirming a role of Nrp1 in those processes. Therefore, Nrp1 may be a promising drug target for neovascular diseases in the eye.
Collapse
Affiliation(s)
- Patricia Fernández-Robredo
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
- Experimental Ophthalmology Laboratory, School of Medicine, University of Navarra, IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - Senthil Selvam
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Michael B. Powner
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
- Division of Optometry and Visual Science, School of Health Sciences, City University London, London, United Kingdom
| | - Dawn A. Sim
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
- NIHR Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
| | - Marcus Fruttiger
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
| |
Collapse
|
112
|
Muhl L, Folestad EB, Gladh H, Wang Y, Moessinger C, Jakobsson L, Eriksson U. Neuropilin 1 binds platelet-derived growth factor (PDGF)-D and is a co-receptor in PDGF-D/PDGF receptor β signaling. J Cell Sci 2017; 130:1365-1378. [DOI: 10.1242/jcs.200493] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 02/19/2017] [Indexed: 01/09/2023] Open
Abstract
Platelet-derived growth factor (PDGF)-D is a PDGF receptor β (PDGFRβ) specific ligand implicated in a number of pathological conditions, such as cardiovascular disease and cancer, but its biological function remains incompletely understood.
In this study, we demonstrate that PDGF-D binds directly to NRP1, with the requirement of the C-terminal Arg residue of PDGF-D. Stimulation with PDGF-D, but not PDGF-B, induced PDGFRβ/NRP1 complex formation in fibroblasts. Additionally, PDGF-D induced translocation of NRP1 to cell-cell junctions in endothelial cells, independent of PDGFRβ, altering the availability of NRP1 for VEGF-A/VEGF receptor 2 signaling. PDGF-D showed differential effects on pericyte behavior in ex vivo sprouting assays, compared to PDGF-B. Furthermore, PDGF-D induced PDGFRβ/NRP1 interaction in the trans-configuration between endothelial cells and pericytes.
In summary, we show that NRP1 can act as a co-receptor for PDGF-D in PDGFRβ signaling, possibly implicated in intercellular communication in the vascular wall.
Collapse
Affiliation(s)
- Lars Muhl
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Scheeles väg 2, A3:P4, S-17177 Stockholm, Sweden
| | - Erika Bergsten Folestad
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Scheeles väg 2, A3:P4, S-17177 Stockholm, Sweden
| | - Hanna Gladh
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Scheeles väg 2, A3:P4, S-17177 Stockholm, Sweden
| | - Yixin Wang
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Scheeles väg 2, A3:P4, S-17177 Stockholm, Sweden
| | - Christine Moessinger
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Scheeles väg 2, A3:P4, S-17177 Stockholm, Sweden
| | - Lars Jakobsson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Scheeles väg 2, A3:P4, S-17177 Stockholm, Sweden
| | - Ulf Eriksson
- Department of Medical Biochemistry and Biophysics, Division of Vascular Biology, Karolinska Institutet, Scheeles väg 2, A3:P4, S-17177 Stockholm, Sweden
| |
Collapse
|
113
|
The expanding role of neuropilin: regulation of transforming growth factor-β and platelet-derived growth factor signaling in the vasculature. Curr Opin Hematol 2016; 23:260-7. [PMID: 26849476 DOI: 10.1097/moh.0000000000000233] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Long recognized for its role in regulation of vascular endothelial growth factor signaling, neuropilin (Nrp)1 has emerged as a modulator of additional signaling pathways critical for vascular development and function. Here we review two novel functions of Nrp1 in blood vessels: regulation of transforming growth factor-β (TGFβ) signaling in endothelial cells and regulation of platelet-derived growth factor (PDGF) signaling in vascular smooth muscle cells. RECENT FINDINGS Novel mouse models demonstrate that Nrp1 fulfills vascular functions independent of vascular endothelial growth factor signaling. These include modulation of TGFβ-dependent inhibition of endothelial sprouting during developmental angiogenesis and PDGF signaling in vascular smooth muscle cells during development and disease. SUMMARY Broadening our understanding of how and where Nrp1 functions in the vasculature is critical for the development of targeted therapeutics for cancer and vascular diseases such as atherosclerosis and retinopathies.
Collapse
|
114
|
Ola R, Dubrac A, Han J, Zhang F, Fang JS, Larrivée B, Lee M, Urarte AA, Kraehling JR, Genet G, Hirschi KK, Sessa WC, Canals FV, Graupera M, Yan M, Young LH, Oh PS, Eichmann A. PI3 kinase inhibition improves vascular malformations in mouse models of hereditary haemorrhagic telangiectasia. Nat Commun 2016; 7:13650. [PMID: 27897192 PMCID: PMC5141347 DOI: 10.1038/ncomms13650] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 10/20/2016] [Indexed: 12/26/2022] Open
Abstract
Activin receptor-like kinase 1 (ALK1) is an endothelial serine-threonine kinase receptor for bone morphogenetic proteins (BMPs) 9 and 10. Inactivating mutations in the ALK1 gene cause hereditary haemorrhagic telangiectasia type 2 (HHT2), a disabling disease characterized by excessive angiogenesis with arteriovenous malformations (AVMs). Here we show that inducible, endothelial-specific homozygous Alk1 inactivation and BMP9/10 ligand blockade both lead to AVM formation in postnatal retinal vessels and internal organs including the gastrointestinal (GI) tract in mice. VEGF and PI3K/AKT signalling are increased on Alk1 deletion and BMP9/10 ligand blockade. Genetic deletion of the signal-transducing Vegfr2 receptor prevents excessive angiogenesis but does not fully revert AVM formation. In contrast, pharmacological PI3K inhibition efficiently prevents AVM formation and reverts established AVMs. Thus, Alk1 deletion leads to increased endothelial PI3K pathway activation that may be a novel target for the treatment of vascular lesions in HHT2.
Collapse
Affiliation(s)
- Roxana Ola
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Alexandre Dubrac
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Jinah Han
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Feng Zhang
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Jennifer S. Fang
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Bruno Larrivée
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Monica Lee
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Ana A. Urarte
- Vascular Signalling Laboratory, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Jan R. Kraehling
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Gael Genet
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Karen K. Hirschi
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - William C. Sessa
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Francesc V. Canals
- Translation Research Laboratory, Catalan Institute of Oncology, Idibell, Barcelona 08908, Spain
| | - Mariona Graupera
- Vascular Signalling Laboratory, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Minhong Yan
- Molecular Oncology, Genentech, Inc., South San Francisco, California 94080-4990, USA
| | - Lawrence H. Young
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Paul S. Oh
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, PO Box 100274, Gainesville, Florida 32610, USA
| | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
- Inserm U970, Paris Cardiovascular Research Center, Paris 75015, France
| |
Collapse
|
115
|
A mouse model of hereditary hemorrhagic telangiectasia generated by transmammary-delivered immunoblocking of BMP9 and BMP10. Sci Rep 2016; 5:37366. [PMID: 27874028 PMCID: PMC5118799 DOI: 10.1038/srep37366] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 10/27/2016] [Indexed: 01/03/2023] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a potentially life-threatening genetic vascular disorder caused by loss-of-function mutations in the genes encoding activin receptor-like kinase 1 (ALK1), endoglin, Smad4, and bone morphogenetic protein 9 (BMP9). Injections of mouse neonates with BMP9/10 blocking antibodies lead to HHT-like vascular defects in the postnatal retinal angiogenesis model. Mothers and their newborns share the same immunity through the transfer of maternal antibodies during lactation. Here, we investigated whether the transmammary delivery route could improve the ease and consistency of administering anti-BMP9/10 antibodies in the postnatal retinal angiogenesis model. We found that anti-BMP9/10 antibodies, when intraperitoneally injected into lactating dams, are efficiently transferred into the blood circulation of lactationally-exposed neonatal pups. Strikingly, pups receiving anti-BMP9/10 antibodies via lactation displayed consistent and robust vascular pathology in the retina, which included hypervascularization and defects in arteriovenous specification, as well as the presence of multiple and massive arteriovenous malformations. Furthermore, RNA-Seq analyses of neonatal retinas identified an increase in the key pro-angiogenic factor, angiopoietin-2, as the most significant change in gene expression triggered by the transmammary delivery of anti-BMP9/10 antibodies. Transmammary-delivered BMP9/10 immunoblocking in the mouse neonatal retina is therefore a practical, noninvasive, reliable, and robust model of HHT vascular pathology.
Collapse
|
116
|
Notch regulates BMP responsiveness and lateral branching in vessel networks via SMAD6. Nat Commun 2016; 7:13247. [PMID: 27834400 PMCID: PMC5114582 DOI: 10.1038/ncomms13247] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 09/15/2016] [Indexed: 12/13/2022] Open
Abstract
Functional blood vessel growth depends on generation of distinct but coordinated responses from endothelial cells. Bone morphogenetic proteins (BMP), part of the TGFβ superfamily, bind receptors to induce phosphorylation and nuclear translocation of SMAD transcription factors (R-SMAD1/5/8) and regulate vessel growth. However, SMAD1/5/8 signalling results in both pro- and anti-angiogenic outputs, highlighting a poor understanding of the complexities of BMP signalling in the vasculature. Here we show that BMP6 and BMP2 ligands are pro-angiogenic in vitro and in vivo, and that lateral vessel branching requires threshold levels of R-SMAD phosphorylation. Endothelial cell responsiveness to these pro-angiogenic BMP ligands is regulated by Notch status and Notch sets responsiveness by regulating a cell-intrinsic BMP inhibitor, SMAD6, which affects BMP responses upstream of target gene expression. Thus, we reveal a paradigm for Notch-dependent regulation of angiogenesis: Notch regulates SMAD6 expression to affect BMP responsiveness of endothelial cells and new vessel branch formation. The mechanism underlying endothelial cell responses to BMP signals is unknown. Here, the authors show that the endothelial response to pro-angiogenic BMP ligands is regulated by Notch via its effect on SMAD6, a known inhibitor of BMP intracellular signaling cascade.
Collapse
|
117
|
Baeyens N, Larrivée B, Ola R, Hayward-Piatkowskyi B, Dubrac A, Huang B, Ross TD, Coon BG, Min E, Tsarfati M, Tong H, Eichmann A, Schwartz MA. Defective fluid shear stress mechanotransduction mediates hereditary hemorrhagic telangiectasia. J Cell Biol 2016; 214:807-16. [PMID: 27646277 PMCID: PMC5037412 DOI: 10.1083/jcb.201603106] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/30/2016] [Indexed: 11/27/2022] Open
Abstract
Morphogenesis of the vascular system is strongly modulated by mechanical forces from blood flow. Hereditary hemorrhagic telangiectasia (HHT) is an inherited autosomal-dominant disease in which arteriovenous malformations and telangiectasias accumulate with age. Most cases are linked to heterozygous mutations in Alk1 or Endoglin, receptors for bone morphogenetic proteins (BMPs) 9 and 10. Evidence suggests that a second hit results in clonal expansion of endothelial cells to form lesions with poor mural cell coverage that spontaneously rupture and bleed. We now report that fluid shear stress potentiates BMPs to activate Alk1 signaling, which correlates with enhanced association of Alk1 and endoglin. Alk1 is required for BMP9 and flow responses, whereas endoglin is only required for enhancement by flow. This pathway mediates both inhibition of endothelial proliferation and recruitment of mural cells; thus, its loss blocks flow-induced vascular stabilization. Identification of Alk1 signaling as a convergence point for flow and soluble ligands provides a molecular mechanism for development of HHT lesions.
Collapse
Affiliation(s)
- Nicolas Baeyens
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06511
| | - Bruno Larrivée
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06511 Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - Roxana Ola
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06511
| | - Brielle Hayward-Piatkowskyi
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06511
| | - Alexandre Dubrac
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06511
| | - Billy Huang
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06511
| | - Tyler D Ross
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06511
| | - Brian G Coon
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06511
| | - Elizabeth Min
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06511
| | - Maya Tsarfati
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06511
| | - Haibin Tong
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Yale Cardiovascular Research Center, Yale University School of Medicine, New Haven, CT 06511 Jilin Provincial Key Laboratory of Molecular Geriatric Medicine, Life Science Research Center, Beihua University, Jilin 132013, China
| | - Anne Eichmann
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Institut National de la Santé et de la Recherche Médicale U970, Paris Center for Cardiovascular Research, 75015 Paris, France
| | - Martin A Schwartz
- Department of Medicine (Cardiology), Yale University School of Medicine, New Haven, CT 06511 Department of Cell Biology, Yale University, New Haven, CT 06510 Department of Biomedical Engineering, Yale University, New Haven, CT 06510
| |
Collapse
|
118
|
Chen P, Xu K, Tao S, Zhou T, Tong Y, Ding H, Zhang L, Chu W, Wu C, Xie Y. Phase-Transformation Engineering in Cobalt Diselenide Realizing Enhanced Catalytic Activity for Hydrogen Evolution in an Alkaline Medium. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:7527-7532. [PMID: 27309389 DOI: 10.1002/adma.201601663] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 05/02/2016] [Indexed: 06/06/2023]
Abstract
Phase-transformation engineering is successfully applied in designing an alkaline hydrogen evolution reaction (HER) electrocatalyst. Benefiting from phase-transformation engineering, which endows higher electrical conductivity, ideal water adsorption energy, and faster transformation efficiency of Hads into hydrogen, cubic-phase CoSe2 realizes an enhanced electrocatalytic activity for HER under alkaline conditions.
Collapse
Affiliation(s)
- Pengzuo Chen
- Hefei National Laboratory for Physical Science at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center (CAS) and CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Kun Xu
- Hefei National Laboratory for Physical Science at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center (CAS) and CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Shi Tao
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| | - Tianpei Zhou
- Hefei National Laboratory for Physical Science at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center (CAS) and CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Yun Tong
- Hefei National Laboratory for Physical Science at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center (CAS) and CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Hui Ding
- Hefei National Laboratory for Physical Science at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center (CAS) and CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Lidong Zhang
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| | - Wangsheng Chu
- National Synchrotron Radiation Laboratory, University of Science and Technology of China, Hefei, Anhui, 230029, P. R. China
| | - Changzheng Wu
- Hefei National Laboratory for Physical Science at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center (CAS) and CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China.
| | - Yi Xie
- Hefei National Laboratory for Physical Science at the Microscale, CAS Center for Excellence in Nanoscience, Hefei Science Center (CAS) and CAS Key Laboratory of Mechanical Behavior and Design of Materials, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| |
Collapse
|
119
|
Ntumba K, Akla N, Oh SP, Eichmann A, Larrivée B. BMP9/ALK1 inhibits neovascularization in mouse models of age-related macular degeneration. Oncotarget 2016; 7:55957-55969. [PMID: 27517154 PMCID: PMC5302889 DOI: 10.18632/oncotarget.11182] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 07/13/2016] [Indexed: 12/15/2022] Open
Abstract
Age-related macular degeneration (AMD) is the leading cause of blindness in aging populations of industrialized countries. The drawbacks of inhibitors of vascular endothelial growth factor (VEGFs) currently used for the treatment of AMD, which include resistance and potential serious side-effects, require the identification of new therapeutic targets to modulate angiogenesis. BMP9 signaling through the endothelial Alk1 serine-threonine kinase receptor modulates the response of endothelial cells to VEGF and promotes vessel quiescence and maturation during development. Here, we show that BMP9/Alk1 signaling inhibits neovessel formation in mouse models of pathological ocular angiogenesis relevant to AMD. Activating Alk1 signaling in laser-induced choroidal neovascularization (CNV) and oxygen-induced retinopathy (OIR) inhibited neovascularization and reduced the volume of vascular lesions. Alk1 signaling was also found to interfere with VEGF signaling in endothelial cells whereas BMP9 potentiated the inhibitory effects of VEGFR2 signaling blockade, both in OIR and laser-induced CNV. Together, our data show that targeting BMP9/Alk1 efficiently prevents the growth of neovessels in AMD models and introduce a new approach to improve conventional anti-VEGF therapies.
Collapse
Affiliation(s)
- Kalonji Ntumba
- Department of Biomedical Sciences, Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - Naoufal Akla
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
| | - S. Paul Oh
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, USA
| | - Anne Eichmann
- Yale Cardiovascular Research Center, New Haven, CT, USA
- Inserm U970, Paris Cardiovascular Research Center, Paris, France
| | - Bruno Larrivée
- Department of Biomedical Sciences, Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Molecular Biology, Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, University of Montreal, Montreal, Quebec, Canada
- Department of Biological Sciences, Université du Québec à Montréal, Montréal, Quebec, Canada
| |
Collapse
|
120
|
Abstract
Transforming growth factor β (TGF-β) family members signal via heterotetrameric complexes of type I and type II dual specificity kinase receptors. The activation and stability of the receptors are controlled by posttranslational modifications, such as phosphorylation, ubiquitylation, sumoylation, and neddylation, as well as by interaction with other proteins at the cell surface and in the cytoplasm. Activation of TGF-β receptors induces signaling via formation of Smad complexes that are translocated to the nucleus where they act as transcription factors, as well as via non-Smad pathways, including the Erk1/2, JNK and p38 MAP kinase pathways, and the Src tyrosine kinase, phosphatidylinositol 3'-kinase, and Rho GTPases.
Collapse
Affiliation(s)
- Carl-Henrik Heldin
- Ludwig Institute for Cancer Research Ltd., Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Aristidis Moustakas
- Ludwig Institute for Cancer Research Ltd., Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23 Uppsala, Sweden
| |
Collapse
|
121
|
Abstract
Vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are uniquely required to balance the formation of new blood vessels with the maintenance and remodelling of existing ones, during development and in adult tissues. Recent advances have greatly expanded our understanding of the tight and multi-level regulation of VEGFR2 signalling, which is the primary focus of this Review. Important insights have been gained into the regulatory roles of VEGFR-interacting proteins (such as neuropilins, proteoglycans, integrins and protein tyrosine phosphatases); the dynamics of VEGFR2 endocytosis, trafficking and signalling; and the crosstalk between VEGF-induced signalling and other endothelial signalling cascades. A clear understanding of this multifaceted signalling web is key to successful therapeutic suppression or stimulation of vascular growth.
Collapse
|
122
|
Gordon EJ, Fukuhara D, Weström S, Padhan N, Sjöström EO, van Meeteren L, He L, Orsenigo F, Dejana E, Bentley K, Spurkland A, Claesson-Welsh L. The endothelial adaptor molecule TSAd is required for VEGF-induced angiogenic sprouting through junctional c-Src activation. Sci Signal 2016; 9:ra72. [PMID: 27436360 DOI: 10.1126/scisignal.aad9256] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Activation of vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2) by VEGF binding is critical for vascular morphogenesis. In addition, VEGF disrupts the endothelial barrier by triggering the phosphorylation and turnover of the junctional molecule VE-cadherin, a process mediated by the VEGFR2 downstream effectors T cell-specific adaptor (TSAd) and the tyrosine kinase c-Src. We investigated whether the VEGFR2-TSAd-c-Src pathway was required for angiogenic sprouting. Indeed, Tsad-deficient embryoid bodies failed to sprout in response to VEGF. Tsad-deficient mice displayed impaired angiogenesis specifically during tracheal vessel development, but not during retinal vasculogenesis, and in VEGF-loaded Matrigel plugs, but not in those loaded with FGF. The SH2 and proline-rich domains of TSAd bridged VEGFR2 and c-Src, and this bridging was critical for the localization of activated c-Src to endothelial junctions and elongation of the growing sprout, but not for selection of the tip cell. These results revealed that vascular sprouting and permeability are both controlled through the VEGFR2-TSAd-c-Src signaling pathway in a subset of tissues, which may be useful in developing strategies to control tissue-specific pathological angiogenesis.
Collapse
Affiliation(s)
- Emma J Gordon
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden.
| | - Daisuke Fukuhara
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden
| | - Simone Weström
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden
| | - Narendra Padhan
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden
| | - Elisabet O Sjöström
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden
| | - Laurens van Meeteren
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden
| | - Liqun He
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden
| | - Fabrizio Orsenigo
- FIRC Institute of Molecular Oncology Foundation, IFOM, Milan 20139, Italy
| | - Elisabetta Dejana
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden. FIRC Institute of Molecular Oncology Foundation, IFOM, Milan 20139, Italy
| | - Katie Bentley
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden. Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Anne Spurkland
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo 0317, Norway
| | - Lena Claesson-Welsh
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjöldsv 20, Uppsala 75185, Sweden.
| |
Collapse
|
123
|
Hashimoto T, Tsuneki M, Foster TR, Santana JM, Bai H, Wang M, Hu H, Hanisch JJ, Dardik A. Membrane-mediated regulation of vascular identity. BIRTH DEFECTS RESEARCH. PART C, EMBRYO TODAY : REVIEWS 2016; 108:65-84. [PMID: 26992081 PMCID: PMC5310768 DOI: 10.1002/bdrc.21123] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 02/22/2016] [Indexed: 02/06/2023]
Abstract
Vascular diseases span diverse pathology, but frequently arise from aberrant signaling attributed to specific membrane-associated molecules, particularly the Eph-ephrin family. Originally recognized as markers of embryonic vessel identity, Eph receptors and their membrane-associated ligands, ephrins, are now known to have a range of vital functions in vascular physiology. Interactions of Ephs with ephrins at cell-to-cell interfaces promote a variety of cellular responses such as repulsion, adhesion, attraction, and migration, and frequently occur during organ development, including vessel formation. Elaborate coordination of Eph- and ephrin-related signaling among different cell populations is required for proper formation of the embryonic vessel network. There is growing evidence supporting the idea that Eph and ephrin proteins also have postnatal interactions with a number of other membrane-associated signal transduction pathways, coordinating translation of environmental signals into cells. This article provides an overview of membrane-bound signaling mechanisms that define vascular identity in both the embryo and the adult, focusing on Eph- and ephrin-related signaling. We also discuss the role and clinical significance of this signaling system in normal organ development, neoplasms, and vascular pathologies.
Collapse
Affiliation(s)
- Takuya Hashimoto
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, Connecticut
- Department of Vascular Surgery, The University of Tokyo, Tokyo, Japan
| | - Masayuki Tsuneki
- Division of Cancer Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Trenton R. Foster
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Jeans M. Santana
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Hualong Bai
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
- Department of Vascular Surgery, The 1st Affiliated Hospital of Zhengzhou University, Henan, China
| | - Mo Wang
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Haidi Hu
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Jesse J. Hanisch
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
| | - Alan Dardik
- The Department of Surgery and the Vascular Biology and Therapeutics Program, Yale University, New Haven, Connecticut
- Department of Surgery, VA Connecticut Healthcare Systems, West Haven, Connecticut
| |
Collapse
|
124
|
Raimondi C, Brash JT, Fantin A, Ruhrberg C. NRP1 function and targeting in neurovascular development and eye disease. Prog Retin Eye Res 2016; 52:64-83. [PMID: 26923176 PMCID: PMC4854174 DOI: 10.1016/j.preteyeres.2016.02.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/08/2016] [Accepted: 02/10/2016] [Indexed: 12/19/2022]
Abstract
Neuropilin 1 (NRP1) is expressed by neurons, blood vessels, immune cells and many other cell types in the mammalian body and binds a range of structurally and functionally diverse extracellular ligands to modulate organ development and function. In recent years, several types of mouse knockout models have been developed that have provided useful tools for experimental investigation of NRP1 function, and a multitude of therapeutics targeting NRP1 have been designed, mostly with the view to explore them for cancer treatment. This review provides a general overview of current knowledge of the signalling pathways that are modulated by NRP1, with particular focus on neuronal and vascular roles in the brain and retina. This review will also discuss the potential of NRP1 inhibitors for the treatment for neovascular eye diseases.
Collapse
Affiliation(s)
- Claudio Raimondi
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - James T Brash
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Alessandro Fantin
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK
| | - Christiana Ruhrberg
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London EC1V 9EL, UK.
| |
Collapse
|
125
|
Yan XC, Cao J, Liang L, Wang L, Gao F, Yang ZY, Duan JL, Chang TF, Deng SM, Liu Y, Dou GR, Zhang J, Zheng QJ, Zhang P, Han H. miR-342-5p Is a Notch Downstream Molecule and Regulates Multiple Angiogenic Pathways Including Notch, Vascular Endothelial Growth Factor and Transforming Growth Factor β Signaling. J Am Heart Assoc 2016; 5:JAHA.115.003042. [PMID: 26857067 PMCID: PMC4802463 DOI: 10.1161/jaha.115.003042] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Endothelial cells (ECs) form blood vessels through angiogenesis that is regulated by coordination of vascular endothelial growth factor (VEGF), Notch, transforming growth factor β, and other signals, but the detailed molecular mechanisms remain unclear. Methods and Results Small RNA sequencing initially identified miR‐342‐5p as a novel downstream molecule of Notch signaling in ECs. Reporter assay, quantitative reverse transcription polymerase chain reaction and Western blot analysis indicated that miR‐342‐5p targeted endoglin and modulated transforming growth factor β signaling by repressing SMAD1/5 phosphorylation in ECs. Transfection of miR‐342‐5p inhibited EC proliferation and lumen formation and reduced angiogenesis in vitro and in vivo, as assayed by using a fibrin beads–based sprouting assay, mouse aortic ring culture, and intravitreal injection of miR‐342‐5p agomir in P3 pups. Moreover, miR‐342‐5p promoted the migration of ECs, accompanied by reduced endothelial markers and increased mesenchymal markers, indicative of increased endothelial–mesenchymal transition. Transfection of endoglin at least partially reversed endothelial–mesenchymal transition induced by miR‐342‐5p. The expression of miR‐342‐5p was upregulated by transforming growth factor β, and inhibition of miR‐342‐5p attenuated the inhibitory effects of transforming growth factor β on lumen formation and sprouting by ECs. In addition, VEGF repressed miR‐342‐5p expression, and transfection of miR‐342‐5p repressed VEGFR2 and VEGFR3 expression and VEGF‐triggered Akt phosphorylation in ECs. miR‐342‐5p repressed angiogenesis in a laser‐induced choroidal neovascularization model in mice, highlighting its clinical potential. Conclusions miR‐342‐5p acts as a multifunctional angiogenic repressor mediating the effects and interaction among angiogenic pathways.
Collapse
Affiliation(s)
- Xian-Chun Yan
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jing Cao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Liang Liang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Li Wang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fang Gao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zi-Yan Yang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Juan-Li Duan
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tian-Fang Chang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - San-Ming Deng
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuan Liu
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guo-Rui Dou
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jian Zhang
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qi-Jun Zheng
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ping Zhang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
126
|
García de Vinuesa A, Abdelilah-Seyfried S, Knaus P, Zwijsen A, Bailly S. BMP signaling in vascular biology and dysfunction. Cytokine Growth Factor Rev 2015; 27:65-79. [PMID: 26823333 DOI: 10.1016/j.cytogfr.2015.12.005] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The vascular system is critical for developmental growth, tissue homeostasis and repair but also for tumor development. Bone morphogenetic protein (BMP) signaling has recently emerged as a fundamental pathway of the endothelium by regulating cardiovascular and lymphatic development and by being causative for several vascular dysfunctions. Two vascular disorders have been directly linked to impaired BMP signaling: pulmonary arterial hypertension and hereditary hemorrhagic telangiectasia. Endothelial BMP signaling critically depends on the cellular context, which includes among others vascular heterogeneity, exposure to flow, and the intertwining with other signaling cascades (Notch, WNT, Hippo and hypoxia). The purpose of this review is to highlight the most recent findings illustrating the clear need for reconsidering the role of BMPs in vascular biology.
Collapse
Affiliation(s)
- Amaya García de Vinuesa
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Leiden, The Netherlands
| | - Salim Abdelilah-Seyfried
- Institute of Biochemistry and Biology, Potsdam University, Karl-Liebknecht-Straße 24-25, D-14476 Potsdam, Germany; Institute of Molecular Biology, Hannover Medical School, Carl-Neuberg Straße 1, D-30625 Hannover, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universitaet Berlin, Berlin, Germany
| | - An Zwijsen
- VIB Center for the Biology of Disease, Leuven, Belgium; KU Leuven, Department of Human Genetics, Leuven, Belgium
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Médicale (INSERM, U1036), Grenoble F-38000, France; Commissariat à l'Énergie Atomique et aux Energies Alternatives, Institut de Recherches en Technologies et Sciences pour le Vivant, Laboratoire Biologie du Cancer et de l'Infection, Grenoble F-38000, France; Université Grenoble-Alpes, Grenoble F-38000, France.
| |
Collapse
|
127
|
Ye L, Jiang WG. Bone morphogenetic proteins in tumour associated angiogenesis and implication in cancer therapies. Cancer Lett 2015; 380:586-597. [PMID: 26639195 DOI: 10.1016/j.canlet.2015.10.036] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/17/2015] [Accepted: 10/12/2015] [Indexed: 02/09/2023]
Abstract
Bone morphogenetic protein (BMP) belongs to transforming growth factor-β superfamily. To date, more than 20 BMPs have been identified in humans. BMPs play a critical role in embryonic and postnatal development, and also in maintaining homeostasis in different organs and tissues by regulating cell differentiation, proliferation, survival and motility. They play important roles in the development and progression of certain malignancies, including prostate cancer, breast cancer, lung cancer, etc. Recently, more evidence shows that BMPs are also involved in tumour associated angiogenesis. For example BMP can either directly regulate the functions of vascular endothelial cells or indirectly influence the angiogenesis via regulation of angiogenic factors, such as vascular endothelial growth factor (VEGF). Such crosstalk can also be reflected in the interaction with other angiogenic factors, like hepatocyte growth factor (HGF) and basic fibroblast growth factor (bFGF). All these factors are involved in the orchestration of the angiogenic process during tumour development and progression. Review of the relevant studies will provide a comprehensive prospective on current understanding and shed light on the corresponding therapeutic opportunity.
Collapse
Affiliation(s)
- Lin Ye
- Metastasis & Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| | - Wen G Jiang
- Metastasis & Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| |
Collapse
|
128
|
Borggrefe T, Lauth M, Zwijsen A, Huylebroeck D, Oswald F, Giaimo BD. The Notch intracellular domain integrates signals from Wnt, Hedgehog, TGFβ/BMP and hypoxia pathways. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:303-13. [PMID: 26592459 DOI: 10.1016/j.bbamcr.2015.11.020] [Citation(s) in RCA: 150] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 11/18/2015] [Accepted: 11/19/2015] [Indexed: 01/12/2023]
Abstract
Notch signaling is a highly conserved signal transduction pathway that regulates stem cell maintenance and differentiation in several organ systems. Upon activation, the Notch receptor is proteolytically processed, its intracellular domain (NICD) translocates into the nucleus and activates expression of target genes. Output, strength and duration of the signal are tightly regulated by post-translational modifications. Here we review the intracellular post-translational regulation of Notch that fine-tunes the outcome of the Notch response. We also describe how crosstalk with other conserved signaling pathways like the Wnt, Hedgehog, hypoxia and TGFβ/BMP pathways can affect Notch signaling output. This regulation can happen by regulation of ligand, receptor or transcription factor expression, regulation of protein stability of intracellular key components, usage of the same cofactors or coregulation of the same key target genes. Since carcinogenesis is often dependent on at least two of these pathways, a better understanding of their molecular crosstalk is pivotal.
Collapse
Affiliation(s)
| | - Matthias Lauth
- Institute of Molecular Biology and Tumor Research, Philipps University Marburg, Germany
| | - An Zwijsen
- VIB Center for the Biology of Disease and Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Danny Huylebroeck
- Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Franz Oswald
- University Medical Center Ulm, Department of Internal Medicine I, Ulm, Germany
| | | |
Collapse
|
129
|
Hirota S, Clements TP, Tang LK, Morales JE, Lee HS, Oh SP, Rivera GM, Wagner DS, McCarty JH. Neuropilin 1 balances β8 integrin-activated TGFβ signaling to control sprouting angiogenesis in the brain. Development 2015; 142:4363-73. [PMID: 26586223 PMCID: PMC4689212 DOI: 10.1242/dev.113746] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 11/06/2015] [Indexed: 12/31/2022]
Abstract
Angiogenesis in the developing central nervous system (CNS) is regulated by neuroepithelial cells, although the genes and pathways that couple these cells to blood vessels remain largely uncharacterized. Here, we have used biochemical, cell biological and molecular genetic approaches to demonstrate that β8 integrin (Itgb8) and neuropilin 1 (Nrp1) cooperatively promote CNS angiogenesis by mediating adhesion and signaling events between neuroepithelial cells and vascular endothelial cells. β8 integrin in the neuroepithelium promotes the activation of extracellular matrix (ECM)-bound latent transforming growth factor β (TGFβ) ligands and stimulates TGFβ receptor signaling in endothelial cells. Nrp1 in endothelial cells suppresses TGFβ activation and signaling by forming intercellular protein complexes with β8 integrin. Cell type-specific ablation of β8 integrin, Nrp1, or canonical TGFβ receptors results in pathological angiogenesis caused by defective neuroepithelial cell-endothelial cell adhesion and imbalances in canonical TGFβ signaling. Collectively, these data identify a paracrine signaling pathway that links the neuroepithelium to blood vessels and precisely balances TGFβ signaling during cerebral angiogenesis. Summary: Neuropilin 1 and β8 integrin cooperatively promote cerebral angiogenesis by mediating adhesion and signaling events between neuroepithelial cells and vascular endothelial cells in the mouse brain.
Collapse
Affiliation(s)
- Shinya Hirota
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Leung K Tang
- College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | - John E Morales
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hye Shin Lee
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - S Paul Oh
- Department of Physiology and Functional Genomics, University of Florida, Gainseville, FL 32610, USA
| | - Gonzalo M Rivera
- College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Daniel S Wagner
- Department of Biosciences, Rice University, Houston, TX 77005, USA
| | - Joseph H McCarty
- Department of Neurosurgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
130
|
Maes H, Olmeda D, Soengas MS, Agostinis P. Vesicular trafficking mechanisms in endothelial cells as modulators of the tumor vasculature and targets of antiangiogenic therapies. FEBS J 2015; 283:25-38. [PMID: 26443003 DOI: 10.1111/febs.13545] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 09/21/2015] [Accepted: 10/02/2015] [Indexed: 11/25/2022]
Abstract
A common feature of solid tumors is their ability to incite the formation of new blood and lymph vessels trough the processes of angiogenesis and lymphangiogenesis, respectively, to support tumor growth and favor metastatic dissemination. As a result of the lack of feedback regulatory control mechanisms or due to the exacerbated presence of pro-angiogenic signals within the tumor microenvironment, the tumor endothelium receives continuous signals to sprout and develop, generating vessels that are structurally and functionally abnormal. An emerging mechanism playing a central role in shaping the tumor vasculature is the endothelial-vesicular network that regulates trafficking/export and degradation of key signaling proteins and membrane receptors, including the vascular endothelial growth-factor receptor-2/3 and members of the Notch pathway. Here we will discuss recent evidence highlighting how vesicular trafficking mechanisms in endothelial cells contribute to pathological angiogenesis/lymphangiogenesis and can provide novel and exploitable targets in antiangiogenic therapies.
Collapse
Affiliation(s)
- Hannelore Maes
- Cell Death Research & Therapy (CDRT) Unit, Department of Cellular and Molecular Medicine, KU Leuven University of Leuven, Belgium
| | - David Olmeda
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - María S Soengas
- Melanoma Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Unit, Department of Cellular and Molecular Medicine, KU Leuven University of Leuven, Belgium
| |
Collapse
|
131
|
Rostama B, Turner JE, Seavey GT, Norton CR, Gridley T, Vary CPH, Liaw L. DLL4/Notch1 and BMP9 Interdependent Signaling Induces Human Endothelial Cell Quiescence via P27KIP1 and Thrombospondin-1. Arterioscler Thromb Vasc Biol 2015; 35:2626-37. [PMID: 26471266 DOI: 10.1161/atvbaha.115.306541] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/05/2015] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Bone morphogenetic protein-9 (BMP9)/activin-like kinase-1 and delta-like 4 (DLL4)/Notch promote endothelial quiescence, and we aim to understand mechanistic interactions between the 2 pathways. We identify new targets that contribute to endothelial quiescence and test whether loss of Dll4(+/-) in adult vasculature alters BMP signaling. APPROACH AND RESULTS Human endothelial cells respond synergistically to BMP9 and DLL4 stimulation, showing complete quiescence and induction of HEY1 and HEY2. Canonical BMP9 signaling via activin-like kinase-1-Smad1/5/9 was disrupted by inhibition of Notch signaling, even in the absence of exogenous DLL4. Similarly, DLL4 activity was suppressed when the basal activin-like kinase-1-Smad1/5/9 pathway was inhibited, showing that these pathways are interdependent. BMP9/DLL4 required induction of P27(KIP1) for quiescence, although multiple factors are involved. To understand these mechanisms, we used proteomics data to identify upregulation of thrombospondin-1, which contributes to the quiescence phenotype. To test whether Dll4 regulates BMP/Smad pathways and endothelial cell phenotype in vivo, we characterized the vasculature of Dll4(+/-) mice, analyzing endothelial cells in the lung, heart, and aorta. Together with changes in endothelial structure and vascular morphogenesis, we found that loss of Dll4 was associated with a significant upregulation of pSmad1/5/9 signaling in lung endothelial cells. Because steady-state endothelial cell proliferation rates were not different in the Dll4(+/-) mice, we propose that the upregulation of pSmad1/5/9 signaling compensates to maintain endothelial cell quiescence in these mice. CONCLUSIONS DLL4/Notch and BMP9/activin-like kinase-1 signaling rely on each other's pathways for full activity. This represents an important mechanism of cross talk that enhances endothelial quiescence and sensitively coordinates cellular responsiveness to soluble and cell-tethered ligands.
Collapse
Affiliation(s)
- Bahman Rostama
- From the Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough
| | - Jacqueline E Turner
- From the Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough
| | - Guy T Seavey
- From the Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough
| | - Christine R Norton
- From the Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough
| | - Thomas Gridley
- From the Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough
| | - Calvin P H Vary
- From the Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough
| | - Lucy Liaw
- From the Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough.
| |
Collapse
|
132
|
Guo HF, Vander Kooi CW. Neuropilin Functions as an Essential Cell Surface Receptor. J Biol Chem 2015; 290:29120-6. [PMID: 26451046 DOI: 10.1074/jbc.r115.687327] [Citation(s) in RCA: 162] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The Neuropilins (Nrps) are a family of essential cell surface receptors involved in multiple fundamental cellular signaling cascades. Nrp family members have key functions in VEGF-dependent angiogenesis and semaphorin-dependent axon guidance, controlling signaling and cross-talk between these fundamental physiological processes. More recently, Nrp function has been found in diverse signaling and adhesive functions, emphasizing their role as pleiotropic co-receptors. Pathological Nrp function has been shown to be important in aberrant activation of both canonical and alternative pathways. Here we review key recent insights into Nrp function in human health and disease.
Collapse
Affiliation(s)
- Hou-Fu Guo
- From the Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536
| | - Craig W Vander Kooi
- From the Department of Molecular and Cellular Biochemistry and Center for Structural Biology, University of Kentucky, Lexington, Kentucky 40536
| |
Collapse
|
133
|
Abstract
The developing central nervous system (CNS) is vascularised through the angiogenic invasion of blood vessels from a perineural vascular plexus, followed by continued sprouting and remodelling until a hierarchical vascular network is formed. Remarkably, vascularisation occurs without perturbing the intricate architecture of the neurogenic niches or the emerging neural networks. We discuss the mouse hindbrain, forebrain and retina as widely used models to study developmental angiogenesis in the mammalian CNS and provide an overview of key cellular and molecular mechanisms regulating the vascularisation of these organs. CNS vascularisation is initiated during embryonic development. CNS vascularisation is studied in the mouse forebrain, hindbrain and retina models. Neuroglial cells interact with endothelial cells to promote angiogenesis. Neuroglial cells produce growth factors and matrix cues to pattern vessels.
Collapse
|