1
|
Anzell AR, Kunz AB, Donovan JP, Tran TG, Lu X, Young S, Roman BL. Blood flow regulates acvrl1 transcription via ligand-dependent Alk1 activity. Angiogenesis 2024; 27:501-522. [PMID: 38727966 DOI: 10.1007/s10456-024-09924-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/19/2024] [Indexed: 05/21/2024]
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant disease characterized by the development of arteriovenous malformations (AVMs) that can result in significant morbidity and mortality. HHT is caused primarily by mutations in bone morphogenetic protein receptors ACVRL1/ALK1, a signaling receptor, or endoglin (ENG), an accessory receptor. Because overexpression of Acvrl1 prevents AVM development in both Acvrl1 and Eng null mice, enhancing ACVRL1 expression may be a promising approach to development of targeted therapies for HHT. Therefore, we sought to understand the molecular mechanism of ACVRL1 regulation. We previously demonstrated in zebrafish embryos that acvrl1 is predominantly expressed in arterial endothelial cells and that expression requires blood flow. Here, we document that flow dependence exhibits regional heterogeneity and that acvrl1 expression is rapidly restored after reinitiation of flow. Furthermore, we find that acvrl1 expression is significantly decreased in mutants that lack the circulating Alk1 ligand, Bmp10, and that, in the absence of flow, intravascular injection of BMP10 or the related ligand, BMP9, restores acvrl1 expression in an Alk1-dependent manner. Using a transgenic acvrl1:egfp reporter line, we find that flow and Bmp10 regulate acvrl1 at the level of transcription. Finally, we observe similar ALK1 ligand-dependent increases in ACVRL1 in human endothelial cells subjected to shear stress. These data suggest that ligand-dependent Alk1 activity acts downstream of blood flow to maintain or enhance acvrl1 expression via a positive feedback mechanism, and that ALK1 activating therapeutics may have dual functionality by increasing both ALK1 signaling flux and ACVRL1 expression.
Collapse
Affiliation(s)
- Anthony R Anzell
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amy B Kunz
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
- Allegheny Health Network, Pittsburgh, PA, USA
| | - James P Donovan
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thanhlong G Tran
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xinyan Lu
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Sarah Young
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Carnegie Mellon University, University Libraries, Pittsburgh, PA, USA
| | - Beth L Roman
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA.
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Anzell AR, Kunz AB, Donovan JP, Tran TG, Lu X, Young S, Roman BL. Blood flow regulates acvrl1 transcription via ligand-dependent Alk1 activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.576046. [PMID: 38328175 PMCID: PMC10849739 DOI: 10.1101/2024.01.25.576046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant disease characterized by the development of arteriovenous malformations (AVMs) that can result in significant morbidity and mortality. HHT is caused primarily by mutations in bone morphogenetic protein receptors ACVRL1/ALK1, a signaling receptor, or endoglin (ENG), an accessory receptor. Because overexpression of Acvrl1 prevents AVM development in both Acvrl1 and Eng null mice, enhancing ACVRL1 expression may be a promising approach to development of targeted therapies for HHT. Therefore, we sought to understand the molecular mechanism of ACVRL1 regulation. We previously demonstrated in zebrafish embryos that acvrl1 is predominantly expressed in arterial endothelial cells and that expression requires blood flow. Here, we document that flow dependence exhibits regional heterogeneity and that acvrl1 expression is rapidly restored after reinitiation of flow. Furthermore, we find that acvrl1 expression is significantly decreased in mutants that lack the circulating Alk1 ligand, Bmp10, and that BMP10 microinjection into the vasculature in the absence of flow enhances acvrl1 expression in an Alk1-dependent manner. Using a transgenic acvrl1:egfp reporter line, we find that flow and Bmp10 regulate acvrl1 at the level of transcription. Finally, we observe similar ALK1 ligand-dependent increases in ACVRL1 in human endothelial cells subjected to shear stress. These data suggest that Bmp10 acts downstream of blood flow to maintain or enhance acvrl1 expression via a positive feedback mechanism, and that ALK1 activating therapeutics may have dual functionality by increasing both ALK1 signaling flux and ACVRL1 expression.
Collapse
Affiliation(s)
- Anthony R. Anzell
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amy Biery Kunz
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
- Current affiliation: Allegheny Health Network, Pittsburgh, PA, USA
| | - James P. Donovan
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thanhlong G. Tran
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
- Current affiliation: National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xinyan Lu
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Sarah Young
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
- Current affiliation: Carnegie Mellon University, University Libraries, Pittsburgh, PA, USA
| | - Beth L. Roman
- Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Ahmed T, Ramonett A, Kwak EA, Kumar S, Flores PC, Ortiz HR, Langlais PR, Hund TJ, Mythreye K, Lee NY. Endothelial tip/stalk cell selection requires BMP9-induced β IV-spectrin expression during sprouting angiogenesis. Mol Biol Cell 2023; 34:ar72. [PMID: 37126382 PMCID: PMC10295478 DOI: 10.1091/mbc.e23-02-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 05/02/2023] Open
Abstract
βIV-Spectrin is a membrane cytoskeletal protein with specialized roles in the nervous system and heart. Recent evidence also indicates a fundamental role for βIV-spectrin in angiogenesis as its endothelial-specific gene deletion in mice enhances embryonic lethality due to hypervascularization and hemorrhagic defects. During early vascular sprouting, βIV-spectrin is believed to inhibit tip cell sprouting in favor of the stalk cell phenotype by mediating VEGFR2 internalization and degradation. Despite these essential roles, mechanisms governing βIV-spectrin expression remain unknown. Here we identify bone morphogenetic protein 9 (BMP9) as a major inducer of βIV-spectrin gene expression in the vascular system. We show that BMP9 signals through the ALK1/Smad1 pathway to induce βIV-spectrin expression, which then recruits CaMKII to the cell membrane to induce phosphorylation-dependent VEGFR2 turnover. Although BMP9 signaling promotes stalk cell behavior through activation of hallmark stalk cell genes ID-1/3 and Hes-1 and Notch signaling cross-talk, we find that βIV-spectrin acts upstream of these pathways as loss of βIV-spectrin in neonate mice leads to retinal hypervascularization due to excessive VEGFR2 levels, increased tip cell populations, and strong Notch inhibition irrespective of BMP9 treatment. These findings demonstrate βIV-spectrin as a BMP9 gene target critical for tip/stalk cell selection during nascent vessel sprouting.
Collapse
Affiliation(s)
- Tasmia Ahmed
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, AZ 85724
| | - Aaron Ramonett
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724
| | - Eun-A Kwak
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724
| | - Sanjay Kumar
- Division of Biology, Indian Institute of Science Education and Research, Tirupati 517507, India
| | - Paola Cruz Flores
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, AZ 85724
| | - Hannah R. Ortiz
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724
| | | | - Thomas J. Hund
- Department of Biomedical Engineering, Ohio State University, Columbus, OH 43210
| | - Karthikeyan Mythreye
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Nam Y. Lee
- Department of Chemistry & Biochemistry, University of Arizona, Tucson, AZ 85724
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724
- Comprehensive Cancer Center, University of Arizona, Tucson, AZ 85724
| |
Collapse
|
4
|
Activity of ROCKII not ROCKI promotes pulmonary metastasis of melanoma cells via modulating Smad2/3-MMP9 and FAK-Src-VEGF signalling. Cell Signal 2022; 97:110389. [PMID: 35718242 DOI: 10.1016/j.cellsig.2022.110389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/03/2022] [Accepted: 06/13/2022] [Indexed: 11/20/2022]
Abstract
Rho-associated coiled-coil kinase (ROCK) inhibition decreases tumourogenic growth, proliferation and angiogenesis. Multifaceted evidences are there about the role of ROCK in cancer progression, but isoform specific analysis in secondary pulmonary melanoma is still unaddressed. This study explored the operating function of ROCK in the metastasis of B16F10 mice melanoma cell line. Inhibition by KD-025 indicated dual wielding role of ROCKII as it is associated with the regulation of MMP9 activity responsible for extra-cellular matrix (ECM) degradation as well as angiogenic invasion as an effect of Src-FAK-STAT3 interaction dependent VEGF switching. We found the assisting role of ROCKII, not ROCKI in nuclear localization of Smads that effectively increased MMP9 expression and activity (p < 0.01). This cleaved the protein components of ECM thereby played a crucial role in tissue remodeling at secondary site during establishment of metastatic tumour. ROCKII phosphorylation at Ser1366 as an activation of the same was imprinted essential for oncogenic molecular bagatelle leading to histo-architectural change of pulmonary tissue with extracellular matrix degradation as a consequence of invasion. Direct correlation of pROCKIISer1366 with MMP9 as well as VEGF expression in vivo studies cue to demonstrate the importance of pROCKIISer1366 inhibition in the context of angiogenesis, and metastasis suggesting ROCKII signaling as a possible target for the treatment of secondary lung cancer specially in metastatic melanoma.
Collapse
|
5
|
Kulikauskas MR, X S, Bautch VL. The versatility and paradox of BMP signaling in endothelial cell behaviors and blood vessel function. Cell Mol Life Sci 2022; 79:77. [PMID: 35044529 PMCID: PMC8770421 DOI: 10.1007/s00018-021-04033-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/20/2021] [Accepted: 11/09/2021] [Indexed: 12/15/2022]
Abstract
Blood vessels expand via sprouting angiogenesis, and this process involves numerous endothelial cell behaviors, such as collective migration, proliferation, cell–cell junction rearrangements, and anastomosis and lumen formation. Subsequently, blood vessels remodel to form a hierarchical network that circulates blood and delivers oxygen and nutrients to tissue. During this time, endothelial cells become quiescent and form a barrier between blood and tissues that regulates transport of liquids and solutes. Bone morphogenetic protein (BMP) signaling regulates both proangiogenic and homeostatic endothelial cell behaviors as blood vessels form and mature. Almost 30 years ago, human pedigrees linked BMP signaling to diseases associated with blood vessel hemorrhage and shunts, and recent work greatly expanded our knowledge of the players and the effects of vascular BMP signaling. Despite these gains, there remain paradoxes and questions, especially with respect to how and where the different and opposing BMP signaling outputs are regulated. This review examines endothelial cell BMP signaling in vitro and in vivo and discusses the paradox of BMP signals that both destabilize and stabilize endothelial cell behaviors.
Collapse
Affiliation(s)
- Molly R Kulikauskas
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Shaka X
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Victoria L Bautch
- Curriculum in Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
6
|
Pan P, Weinsheimer S, Cooke D, Winkler E, Abla A, Kim H, Su H. Review of treatment and therapeutic targets in brain arteriovenous malformation. J Cereb Blood Flow Metab 2021; 41:3141-3156. [PMID: 34162280 PMCID: PMC8669284 DOI: 10.1177/0271678x211026771] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/23/2022]
Abstract
Brain arteriovenous malformations (bAVM) are an important cause of intracranial hemorrhage (ICH), especially in younger patients. The pathogenesis of bAVM are largely unknown. Current understanding of bAVM etiology is based on studying genetic syndromes, animal models, and surgically resected specimens from patients. The identification of activating somatic mutations in the Kirsten rat sarcoma viral oncogene homologue (KRAS) gene and other mitogen-activated protein kinase (MAPK) pathway genes has opened up new avenues for bAVM study, leading to a paradigm shift to search for somatic, de novo mutations in sporadic bAVMs instead of focusing on inherited genetic mutations. Through the development of new models and understanding of pathways involved in maintaining normal vascular structure and functions, promising therapeutic targets have been identified and safety and efficacy studies are underway in animal models and in patients. The goal of this paper is to provide a thorough review or current diagnostic and treatment tools, known genes and key pathways involved in bAVM pathogenesis to summarize current treatment options and potential therapeutic targets uncovered by recent discoveries.
Collapse
Affiliation(s)
- Peipei Pan
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, USA
| | - Shantel Weinsheimer
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, USA
| | - Daniel Cooke
- Department of Radiology, University of California, San Francisco, USA
| | - Ethan Winkler
- Department of Neurosurgery, University of California, San Francisco, USA
| | - Adib Abla
- Department of Neurosurgery, University of California, San Francisco, USA
| | - Helen Kim
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, USA
| | - Hua Su
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, USA
| |
Collapse
|
7
|
Lyons O, Walker J, Seet C, Ikram M, Kuchta A, Arnold A, Hernández-Vásquez M, Frye M, Vizcay-Barrena G, Fleck RA, Patel AS, Padayachee S, Mortimer P, Jeffery S, Berland S, Mansour S, Ostergaard P, Makinen T, Modarai B, Saha P, Smith A. Mutations in EPHB4 cause human venous valve aplasia. JCI Insight 2021; 6:e140952. [PMID: 34403370 PMCID: PMC8492339 DOI: 10.1172/jci.insight.140952] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 08/11/2021] [Indexed: 11/25/2022] Open
Abstract
Venous valve (VV) failure causes chronic venous insufficiency, but the molecular regulation of valve development is poorly understood. A primary lymphatic anomaly, caused by mutations in the receptor tyrosine kinase EPHB4, was recently described, with these patients also presenting with venous insufficiency. Whether the venous anomalies are the result of an effect on VVs is not known. VV formation requires complex "organization" of valve-forming endothelial cells, including their reorientation perpendicular to the direction of blood flow. Using quantitative ultrasound, we identified substantial VV aplasia and deep venous reflux in patients with mutations in EPHB4. We used a GFP reporter in mice to study expression of its ligand, ephrinB2, and analyzed developmental phenotypes after conditional deletion of floxed Ephb4 and Efnb2 alleles. EphB4 and ephrinB2 expression patterns were dynamically regulated around organizing valve-forming cells. Efnb2 deletion disrupted the normal endothelial expression patterns of the gap junction proteins connexin37 and connexin43 (both required for normal valve development) around reorientating valve-forming cells and produced deficient valve-forming cell elongation, reorientation, polarity, and proliferation. Ephb4 was also required for valve-forming cell organization and subsequent growth of the valve leaflets. These results uncover a potentially novel cause of primary human VV aplasia.
Collapse
Affiliation(s)
- Oliver Lyons
- Academic Department of Vascular Surgery, Section of Vascular Risk and Surgery, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King’s College London, St. Thomas’ Hospital, London, United Kingdom
| | - James Walker
- Academic Department of Vascular Surgery, Section of Vascular Risk and Surgery, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King’s College London, St. Thomas’ Hospital, London, United Kingdom
| | - Christopher Seet
- Academic Department of Vascular Surgery, Section of Vascular Risk and Surgery, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King’s College London, St. Thomas’ Hospital, London, United Kingdom
| | - Mohammed Ikram
- Academic Department of Vascular Surgery, Section of Vascular Risk and Surgery, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King’s College London, St. Thomas’ Hospital, London, United Kingdom
| | - Adam Kuchta
- Department of Ultrasonic Angiology, Guy’s & St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Andrew Arnold
- Department of Ultrasonic Angiology, Guy’s & St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Magda Hernández-Vásquez
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Sweden
| | - Maike Frye
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Sweden
| | - Gema Vizcay-Barrena
- Centre for Ultrastructural Imaging, King’s College London, London, United Kingdom
| | - Roland A. Fleck
- Centre for Ultrastructural Imaging, King’s College London, London, United Kingdom
| | - Ashish S. Patel
- Academic Department of Vascular Surgery, Section of Vascular Risk and Surgery, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King’s College London, St. Thomas’ Hospital, London, United Kingdom
| | - Soundrie Padayachee
- Department of Ultrasonic Angiology, Guy’s & St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Peter Mortimer
- Molecular and Clinical Sciences Research Institute, St. George’s University of London, London, United Kingdom
| | - Steve Jeffery
- Molecular and Clinical Sciences Research Institute, St. George’s University of London, London, United Kingdom
| | - Siren Berland
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Sahar Mansour
- Molecular and Clinical Sciences Research Institute, St. George’s University of London, London, United Kingdom
- South West Thames Regional Genetics Service, St. George’s Hospital, London, United Kingdom
| | - Pia Ostergaard
- Molecular and Clinical Sciences Research Institute, St. George’s University of London, London, United Kingdom
| | - Taija Makinen
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Sweden
| | - Bijan Modarai
- Academic Department of Vascular Surgery, Section of Vascular Risk and Surgery, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King’s College London, St. Thomas’ Hospital, London, United Kingdom
| | - Prakash Saha
- Academic Department of Vascular Surgery, Section of Vascular Risk and Surgery, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King’s College London, St. Thomas’ Hospital, London, United Kingdom
| | - Alberto Smith
- Academic Department of Vascular Surgery, Section of Vascular Risk and Surgery, School of Cardiovascular Medicine and Sciences, BHF Centre of Research Excellence, King’s College London, St. Thomas’ Hospital, London, United Kingdom
| |
Collapse
|
8
|
Abstract
Brain arteriovenous malformation (bAVM) is the most common cause of intracranial hemorrhage (ICH), particularly in young patients. However, the exact cause of bAVM bleeding and rupture is not yet fully understood. In bAVMs, blood bypasses the entire capillary bed and directly flows from arteries to veins. The vessel walls in bAVMs have structural defects, which impair vascular integrity. Mural cells are essential structural and functional components of blood vessels and play a critical role in maintaining vascular integrity. Changes in mural cell number and coverage have been implicated in bAVMs. In this review, we discussed the roles of mural cells in bAVM pathogenesis. We focused on 1) the recent advances in human and animal studies of bAVMs; 2) the importance of mural cells in vascular integrity; 3) the regulatory signaling pathways that regulate mural cell function. More specifically, the platelet-derived growth factor-B (PDGF-B)/PDGF receptor-β (PDGFR-β), EphrinB2/EphB4, and angiopoietins/tie2 signaling pathways that regulate mural cell-recruitment during vascular remodeling were discussed in detail.
Collapse
|
9
|
Ruiz S, Zhao H, Chandakkar P, Papoin J, Choi H, Nomura-Kitabayashi A, Patel R, Gillen M, Diao L, Chatterjee PK, He M, Al-Abed Y, Wang P, Metz CN, Oh SP, Blanc L, Campagne F, Marambaud P. Correcting Smad1/5/8, mTOR, and VEGFR2 treats pathology in hereditary hemorrhagic telangiectasia models. J Clin Invest 2020; 130:942-957. [PMID: 31689244 DOI: 10.1172/jci127425] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 10/31/2019] [Indexed: 12/17/2022] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT), a genetic bleeding disorder leading to systemic arteriovenous malformations (AVMs), is caused by loss-of-function mutations in the ALK1/ENG/Smad1/5/8 pathway. Evidence suggests that HHT pathogenesis strongly relies on overactivated PI3K/Akt/mTOR and VEGFR2 pathways in endothelial cells (ECs). In the BMP9/10-immunoblocked (BMP9/10ib) neonatal mouse model of HHT, we report here that the mTOR inhibitor, sirolimus, and the receptor tyrosine kinase inhibitor, nintedanib, could synergistically fully block, but also reversed, retinal AVMs to avert retinal bleeding and anemia. Sirolimus plus nintedanib prevented vascular pathology in the oral mucosa, lungs, and liver of the BMP9/10ib mice, as well as significantly reduced gastrointestinal bleeding and anemia in inducible ALK1-deficient adult mice. Mechanistically, in vivo in BMP9/10ib mouse ECs, sirolimus and nintedanib blocked the overactivation of mTOR and VEGFR2, respectively. Furthermore, we found that sirolimus activated ALK2-mediated Smad1/5/8 signaling in primary ECs - including in HHT patient blood outgrowth ECs - and partially rescued Smad1/5/8 activity in vivo in BMP9/10ib mouse ECs. These data demonstrate that the combined correction of endothelial Smad1/5/8, mTOR, and VEGFR2 pathways opposes HHT pathogenesis. Repurposing of sirolimus plus nintedanib might provide therapeutic benefit in patients with HHT.
Collapse
Affiliation(s)
- Santiago Ruiz
- Litwin-Zucker Center for Alzheimer's Disease and Memory Disorders and
| | - Haitian Zhao
- Litwin-Zucker Center for Alzheimer's Disease and Memory Disorders and
| | | | - Julien Papoin
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Hyunwoo Choi
- Barrow Aneurysm and AVM Research Center, Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | | | - Radhika Patel
- Litwin-Zucker Center for Alzheimer's Disease and Memory Disorders and
| | - Matthew Gillen
- Litwin-Zucker Center for Alzheimer's Disease and Memory Disorders and
| | - Li Diao
- Center for Immunology and Inflammation
| | | | - Mingzhu He
- Center for Molecular Innovation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA
| | - Yousef Al-Abed
- Center for Molecular Innovation, The Feinstein Institutes for Medical Research, Manhasset, New York, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Ping Wang
- Center for Immunology and Inflammation.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Christine N Metz
- Institute of Molecular Medicine, and.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - S Paul Oh
- Barrow Aneurysm and AVM Research Center, Department of Neurobiology, Barrow Neurological Institute, Phoenix, Arizona, USA
| | - Lionel Blanc
- Center for Autoimmune, Musculoskeletal and Hematopoietic Diseases, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, New York, USA.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| | - Fabien Campagne
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and.,Department of Physiology and Biophysics, Weill Cornell Medical College, New York, New York, USA
| | - Philippe Marambaud
- Litwin-Zucker Center for Alzheimer's Disease and Memory Disorders and.,Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York, USA
| |
Collapse
|
10
|
Viallard C, Audiger C, Popovic N, Akla N, Lanthier K, Legault-Navarrete I, Melichar H, Costantino S, Lesage S, Larrivée B. BMP9 signaling promotes the normalization of tumor blood vessels. Oncogene 2020; 39:2996-3014. [PMID: 32042114 DOI: 10.1038/s41388-020-1200-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/10/2020] [Accepted: 01/29/2020] [Indexed: 01/09/2023]
Abstract
The presence of an immature tumor vascular network contributes to cancer dissemination and the development of resistance to therapies. Strategies to normalize the tumor vasculature are therefore of significant therapeutic interest for cancer treatments. VEGF inhibitors are used clinically to normalize tumor blood vessels. However, the time frame and dosage of these inhibitors required to achieve normalization is rather narrow, and there is a need to identify additional signaling targets to attain vascular normalization. In addition to VEGF, the endothelial-specific receptor Alk1 plays a critical role in vascular development and promotes vascular remodeling and maturation. Therefore, we sought to evaluate the effects of the Alk1 ligand BMP9 on tumor vascular formation. BMP9 overexpression in Lewis Lung Carcinoma (LLC) tumors significantly delayed tumor growth. Blood vessels in BMP9-overexpressing LLC tumors displayed markers of vascular maturation and were characterized by increased perivascular cell coverage. Tumor vasculature normalization was associated with decreased permeability and increased perfusion. These changes in vascular function in BMP9-overexpressing LLC tumors resulted in significant alterations of the tumor microenvironment, characterized by a decrease in hypoxia and an increase in immune infiltration. In conclusion, we show that BMP9 promotes vascular normalization in LLC tumors that leads to changes in the microenvironment.
Collapse
Affiliation(s)
- Claire Viallard
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Biologie Moléculaire, Université de Montréal, Montréal, QC, Canada
| | - Cindy Audiger
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, Canada
| | - Natalija Popovic
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Biologie Moléculaire, Université de Montréal, Montréal, QC, Canada
| | - Naoufal Akla
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Biochimie, Université de Montréal, Montréal, QC, Canada
| | - Kevin Lanthier
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Biologie Moléculaire, Université de Montréal, Montréal, QC, Canada
| | | | - Heather Melichar
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Santiago Costantino
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département d'Ophtalmologie, Université de Montréal, Montréal, QC, Canada
| | - Sylvie Lesage
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada.,Département de Microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, Canada
| | - Bruno Larrivée
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada. .,Département de Biologie Moléculaire, Université de Montréal, Montréal, QC, Canada. .,Département d'Ophtalmologie, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
11
|
Logan SM, Storey KB. Angiogenic signaling in the lungs of a metabolically suppressed hibernating mammal ( Ictidomys tridecemlineatus). PeerJ 2019; 7:e8116. [PMID: 31763078 PMCID: PMC6870509 DOI: 10.7717/peerj.8116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/29/2019] [Indexed: 01/18/2023] Open
Abstract
To conserve energy in times of limited resource availability, particularly during cold winters, hibernators suppress even the most basic of physiologic processes. Breathing rates decrease from 40 breaths/minute to less than 1 breath/min as they decrease body temperature from 37 °C to ambient. Nevertheless, after months of hibernation, these incredible mammals emerge from torpor unscathed. This study was conducted to better understand the protective and possibly anti-inflammatory adaptations that hibernator lungs may use to prevent damage associated with entering and emerging from natural torpor. We postulated that the differential protein expression of soluble protein receptors (decoy receptors that sequester soluble ligands to inhibit signal transduction) would help identify inhibited inflammatory signaling pathways in metabolically suppressed lungs. Instead, the only two soluble receptors that responded to torpor were sVEGFR1 and sVEGFR2, two receptors whose full-length forms are bound by VEGF-A to regulate endothelial cell function and angiogenesis. Decreased sVEGFR1/2 correlated with increased total VEGFR2 protein levels. Maintained or increased levels of key γ-secretase subunits suggested that decreased sVEGFR1/2 protein levels were not due to decreased levels of intramembrane cleavage complex subunits. VEGF-A protein levels did not change, suggesting that hibernators may regulate VEGFR1/2 signaling at the level of the receptor instead of increasing relative ligand abundance. A panel of angiogenic factors used to identify biomarkers of angiogenesis showed a decrease in FGF-1 and an increase in BMP-9. Torpid lungs may use VEGF and BMP-9 signaling to balance angiogenesis and vascular stability, possibly through the activation of SMAD signaling for adaptive tissue remodeling.
Collapse
Affiliation(s)
- Samantha M. Logan
- Institute of Biochemistry, Departments of Biology and Chemistry, Carleton University, Ottawa, ON, Canada
| | - Kenneth B. Storey
- Institute of Biochemistry, Departments of Biology and Chemistry, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
12
|
Perron JC, Rodrigues AA, Surubholta N, Dodd J. Chemotropic signaling by BMP7 requires selective interaction at a key residue in ActRIIA. Biol Open 2019; 8:bio.042283. [PMID: 31208997 PMCID: PMC6679395 DOI: 10.1242/bio.042283] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BMP7 evokes acute chemotropic PI3K-dependent responses, such as growth cone collapse and monocyte chemotaxis, as well as classical Smad-dependent gene transcription. That these divergent responses can be activated in the same cell raises the question of how the BMP-dependent signaling apparatus is manipulated to produce chemotropic and transcriptional signals. RNA interference and site-directed mutagenesis were used to explore functional and structural BMP receptor requirements for BMP7-evoked chemotropic activity. We show that specific type II BMP receptor subunits, ActRIIA and BMPR2, are required for BMP7-induced growth cone collapse in developing spinal neurons and for chemotaxis of monocytes. Reintroduction of wild-type ActRIIA into monocytic cells lacking endogenous ActRIIA restores BMP7-evoked chemotaxis, whereas expression of an ActRIIA K76A receptor variant fails to rescue. BMP7-evoked Smad-dependent signaling is unaffected by either ActRIIA knockdown or expression of the ActRIIA K76A variant. In contrast, BMP7-evoked PI3K-dependent signaling is significantly disturbed in the presence of ActRIIA K76A. These results support a model for selective engagement of chemotropic BMPs with type II BMP receptors, through specific residues, that results in strict regulation of PI3K-dependent signal transduction. This article has an associated First Person interview with the first author of the paper. Summary: Chemotropic BMPs, typified by BMP7, mediate selective receptor recruitment and transduction of PI3K-dependent intracellular signals through interaction with a key residue in the ActRIIA type II BMP receptor.
Collapse
Affiliation(s)
- Jeanette C Perron
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY 11439, USA
| | - Alcina A Rodrigues
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY 11439, USA
| | - Nirupama Surubholta
- Department of Pharmaceutical Sciences, St. John's University, Queens, NY 11439, USA
| | - Jane Dodd
- Departments of Physiology & Cellular Biophysics and Neuroscience, Columbia University, New York, NY 10032, USA
| |
Collapse
|
13
|
Avecilla V. Effect of Transcriptional Regulator ID3 on Pulmonary Arterial Hypertension and Hereditary Hemorrhagic Telangiectasia. Int J Vasc Med 2019; 2019:2123906. [PMID: 31380118 PMCID: PMC6657613 DOI: 10.1155/2019/2123906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/26/2019] [Indexed: 11/17/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) can be discovered in patients who have a loss of function mutation of activin A receptor-like type 1 (ACVRL1) gene, a bone morphogenetic protein (BMP) type 1 receptor. Additionally, ACVRL1 mutations can lead to hereditary hemorrhagic telangiectasia (HHT), also known as Rendu-Osler-Weber disease, an autosomal dominant inherited disease that results in mucocutaneous telangiectasia and arteriovenous malformations (AVMs). Transcriptional regulator Inhibitor of DNA-Binding/Differentiation-3 (ID3) has been demonstrated to be involved in both PAH and HTT; however, the role of its overlapping molecular mechanistic effects has yet to be seen. This review will focus on the existing understanding of how ID3 may contribute to molecular involvement and perturbations thus altering both PAH and HHT outcomes. Improved understanding of how ID3 mediates these pathways will likely provide knowledge in the inhibition and regulation of these diseases through targeted therapies.
Collapse
Affiliation(s)
- Vincent Avecilla
- Department of Environmental Health Sciences, Robert Stempel College of Public Health & Social Work, Florida International University, Miami, FL 33199, USA
- Celgene Corporation, Summit, NJ 07901, USA
| |
Collapse
|
14
|
Shang T, Li S, Zhang Y, Lu L, Cui L, Guo FF. Hypoxia promotes differentiation of adipose-derived stem cells into endothelial cells through demethylation of ephrinB2. Stem Cell Res Ther 2019; 10:133. [PMID: 31109374 PMCID: PMC6528245 DOI: 10.1186/s13287-019-1233-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/06/2019] [Accepted: 04/10/2019] [Indexed: 01/22/2023] Open
Abstract
Background Delivery of endothelial cells into the ischemic tissue is emerging as an alternative approach in revascularization of injured tissues by means of angiogenesis to restore organ function. Adipose-derived stem cells (ASCs) are a readily accessible source of the mesenchymal stem cell with rapid expansion and multidifferentiation potential. The view has emerged that endothelial cells (ECs) differentiated from ASCs is a step forward for adult vascular repair in regenerative medicine and construction of the blood vessel by tissue engineering approach. Methods In this study, differentiation of human ASCs (hASCs) into vascular EC lineage was induced by combined treatment of vascular endothelial growth factor (VEGF) and bone morphogenetic protein-4 (BMP4) under hypoxia condition. The expression of CD31, VEGF-R2, and VE-cadherin was determined by immunofluorescent staining, real-time PCR, and western blot analysis. These differentiated cells acquired functional characteristics of mature ECs as determined by their tube formation ability, DiI-ac-LDL uptake, and nitric oxide secretion in vitro. The methylation status in the proximal promoter CpGs was determined by bisulfite sequencing. Results hASCs expressed endothelial cell markers including CD31, VEGF-R2, and VE-cadherin by combined treatment of VEGF and BMP4 under hypoxia condition. These differentiated cells exhibited the angiogenesis potential in vitro, and injection of these differentiated cells enhanced angiogenesis in the ischemic hindlimb of diabetic mice. Furthermore, it was found that hypoxia increased significantly EphrinB2 expression EC differentiation, which is greatly downregulated with EphrinB2 blockage. The methylation status in the proximal promoter CpG results showed that methylation of EphrinB2 promoter decreased in hASCs with exposure to hypoxia. Conclusion Our data demonstrate that hASCs can be efficiently induced to differentiate into vascular EC lineages which are mediated by demethylation of ephrinB2 under hypoxia condition.
Collapse
Affiliation(s)
- Ting Shang
- Department of Plastic Surgery, Beijing Shijitan Hospital affiliated to Capital Medical University, 10 Tieyi Road, Beijing, China
| | - Shuaijun Li
- Department of Plastic Surgery, Beijing Shijitan Hospital affiliated to Capital Medical University, 10 Tieyi Road, Beijing, China
| | - Yun Zhang
- Department of Plastic Surgery, Beijing Shijitan Hospital affiliated to Capital Medical University, 10 Tieyi Road, Beijing, China
| | - Laiya Lu
- Department of Orthopedics, Shanghai Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, China
| | - Lei Cui
- Department of Plastic Surgery, Beijing Shijitan Hospital affiliated to Capital Medical University, 10 Tieyi Road, Beijing, China. .,Department of Orthopedics, Shanghai Tongji Hospital, Tongji University School of Medicine, 389 Xincun Road, Shanghai, China.
| | - Fang Fang Guo
- Department of Plastic and Reconstructive surgery, Zhongda Hospital, Southeast University, 87 Dingjiaqiao street, Nanjing, Jiangsu Province, China.
| |
Collapse
|
15
|
Duran D, Zeng X, Jin SC, Choi J, Nelson-Williams C, Yatsula B, Gaillard J, Furey CG, Lu Q, Timberlake AT, Dong W, Sorscher MA, Loring E, Klein J, Allocco A, Hunt A, Conine S, Karimy JK, Youngblood MW, Zhang J, DiLuna ML, Matouk CC, Mane S, Tikhonova IR, Castaldi C, López-Giráldez F, Knight J, Haider S, Soban M, Alper SL, Komiyama M, Ducruet AF, Zabramski JM, Dardik A, Walcott BP, Stapleton CJ, Aagaard-Kienitz B, Rodesch G, Jackson E, Smith ER, Orbach DB, Berenstein A, Bilguvar K, Vikkula M, Gunel M, Lifton RP, Kahle KT. Mutations in Chromatin Modifier and Ephrin Signaling Genes in Vein of Galen Malformation. Neuron 2019; 101:429-443.e4. [PMID: 30578106 DOI: 10.1016/j.neuron.2018.11.041] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/12/2018] [Accepted: 11/20/2018] [Indexed: 02/05/2023]
Abstract
Normal vascular development includes the formation and specification of arteries, veins, and intervening capillaries. Vein of Galen malformations (VOGMs) are among the most common and severe neonatal brain arterio-venous malformations, shunting arterial blood into the brain's deep venous system through aberrant direct connections. Exome sequencing of 55 VOGM probands, including 52 parent-offspring trios, revealed enrichment of rare damaging de novo mutations in chromatin modifier genes that play essential roles in brain and vascular development. Other VOGM probands harbored rare inherited damaging mutations in Ephrin signaling genes, including a genome-wide significant mutation burden in EPHB4. Inherited mutations showed incomplete penetrance and variable expressivity, with mutation carriers often exhibiting cutaneous vascular abnormalities, suggesting a two-hit mechanism. The identified mutations collectively account for ∼30% of studied VOGM cases. These findings provide insight into disease biology and may have clinical implications for risk assessment.
Collapse
Affiliation(s)
- Daniel Duran
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Xue Zeng
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Sheng Chih Jin
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA; Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Jungmin Choi
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA; Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | | | - Bogdan Yatsula
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Jonathan Gaillard
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | | | - Qiongshi Lu
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Weilai Dong
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Michelle A Sorscher
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erin Loring
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Jennifer Klein
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA
| | - August Allocco
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Ava Hunt
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Sierra Conine
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Jason K Karimy
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Mark W Youngblood
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA; Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Jinwei Zhang
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Hatherly Laboratory, Exeter, UK
| | - Michael L DiLuna
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Charles C Matouk
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA
| | - Shrikant Mane
- Yale Center for Genome Analysis, West Haven, CT, USA
| | | | | | | | - James Knight
- Yale Center for Genome Analysis, West Haven, CT, USA
| | - Shozeb Haider
- University College London, School of Pharmacy, London, UK
| | - Mariya Soban
- University College London, School of Pharmacy, London, UK; Department of Biochemistry, Aligarh Muslim University, Aligarh, India
| | - Seth L Alper
- Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Masaki Komiyama
- Department of Neurointervention, Osaka City General Hospital, Osaka, Japan
| | - Andrew F Ducruet
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Joseph M Zabramski
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Alan Dardik
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Brian P Walcott
- Department of Neurological Surgery, University of Southern California, Los Angeles, CA, USA
| | - Christopher J Stapleton
- Department of Neurological Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | | | - Georges Rodesch
- Service de Neuroradiologie Diagnostique et Thérapeutique, Hôpital Foch, Suresnes, France
| | - Eric Jackson
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Edward R Smith
- Department of Neurointerventional Radiology, Boston Children's Hospital, Boston, MA, USA
| | - Darren B Orbach
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, USA; Department of Neurointerventional Radiology, Boston Children's Hospital, Boston, MA, USA
| | - Alejandro Berenstein
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kaya Bilguvar
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA; Yale Center for Genome Analysis, West Haven, CT, USA
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Murat Gunel
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA; Department of Genetics, Yale School of Medicine, New Haven, CT, USA
| | - Richard P Lifton
- Department of Genetics, Yale School of Medicine, New Haven, CT, USA; Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY, USA
| | - Kristopher T Kahle
- Department of Neurosurgery, Yale School of Medicine, New Haven, CT, USA; Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
16
|
Goumans MJ, Ten Dijke P. TGF-β Signaling in Control of Cardiovascular Function. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a022210. [PMID: 28348036 DOI: 10.1101/cshperspect.a022210] [Citation(s) in RCA: 192] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Genetic studies in animals and humans indicate that gene mutations that functionally perturb transforming growth factor β (TGF-β) signaling are linked to specific hereditary vascular syndromes, including Osler-Rendu-Weber disease or hereditary hemorrhagic telangiectasia and Marfan syndrome. Disturbed TGF-β signaling can also cause nonhereditary disorders like atherosclerosis and cardiac fibrosis. Accordingly, cell culture studies using endothelial cells or smooth muscle cells (SMCs), cultured alone or together in two- or three-dimensional cell culture assays, on plastic or embedded in matrix, have shown that TGF-β has a pivotal effect on endothelial and SMC proliferation, differentiation, migration, tube formation, and sprouting. Moreover, TGF-β can stimulate endothelial-to-mesenchymal transition, a process shown to be of key importance in heart valve cushion formation and in various pathological vascular processes. Here, we discuss the roles of TGF-β in vasculogenesis, angiogenesis, and lymphangiogenesis and the deregulation of TGF-β signaling in cardiovascular diseases.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Molecular Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
17
|
Cardiovascular Disease: An Introduction. BIOMATHEMATICAL AND BIOMECHANICAL MODELING OF THE CIRCULATORY AND VENTILATORY SYSTEMS 2018. [PMCID: PMC7123129 DOI: 10.1007/978-3-319-89315-0_1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Cardiovascular disease (CVD) is a collective term designating all types of affliction affecting the blood circulatory system, including the heart and vasculature, which, respectively, displaces and conveys the blood.
Collapse
|
18
|
Ruiz S, Chandakkar P, Zhao H, Papoin J, Chatterjee PK, Christen E, Metz CN, Blanc L, Campagne F, Marambaud P. Tacrolimus rescues the signaling and gene expression signature of endothelial ALK1 loss-of-function and improves HHT vascular pathology. Hum Mol Genet 2017; 26:4786-4798. [PMID: 28973643 PMCID: PMC5886173 DOI: 10.1093/hmg/ddx358] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/09/2017] [Accepted: 09/11/2017] [Indexed: 01/02/2023] Open
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is a highly debilitating and life-threatening genetic vascular disorder arising from endothelial cell (EC) proliferation and hypervascularization, for which no cure exists. Because HHT is caused by loss-of-function mutations in bone morphogenetic protein 9 (BMP9)-ALK1-Smad1/5/8 signaling, interventions aimed at activating this pathway are of therapeutic value. We interrogated the whole-transcriptome in human umbilical vein ECs (HUVECs) and found that ALK1 signaling inhibition was associated with a specific pro-angiogenic gene expression signature, which included a significant elevation of DLL4 expression. By screening the NIH clinical collections of FDA-approved drugs, we identified tacrolimus (FK-506) as the most potent activator of ALK1 signaling in BMP9-challenged C2C12 reporter cells. In HUVECs, tacrolimus activated Smad1/5/8 and opposed the pro-angiogenic gene expression signature associated with ALK1 loss-of-function, by notably reducing Dll4 expression. In these cells, tacrolimus also inhibited Akt and p38 stimulation by vascular endothelial growth factor, a major driver of angiogenesis. In the BMP9/10-immunodepleted postnatal retina-a mouse model of HHT vascular pathology-tacrolimus activated endothelial Smad1/5/8 and prevented the Dll4 overexpression and hypervascularization associated with this model. Finally, tacrolimus stimulated Smad1/5/8 signaling in C2C12 cells expressing BMP9-unresponsive ALK1 HHT mutants and in HHT patient blood outgrowth ECs. Tacrolimus repurposing has therefore therapeutic potential in HHT.
Collapse
Affiliation(s)
- Santiago Ruiz
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease
| | | | - Haitian Zhao
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease
| | | | - Prodyot K Chatterjee
- Center for Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
| | - Erica Christen
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease
| | - Christine N Metz
- Center for Biomedical Science, The Feinstein Institute for Medical Research, Manhasset, NY 11030, USA
- Hofstra Northwell School of Medicine, Hempstead, NY 11549, USA
| | - Lionel Blanc
- Center for Autoimmune and Musculoskeletal Disorders
- Hofstra Northwell School of Medicine, Hempstead, NY 11549, USA
| | - Fabien Campagne
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine
- Department of Physiology and Biophysics, The Weill Cornell Medical College, New York, NY 10021, USA
| | - Philippe Marambaud
- Litwin-Zucker Research Center for the Study of Alzheimer's Disease
- Hofstra Northwell School of Medicine, Hempstead, NY 11549, USA
| |
Collapse
|
19
|
Roman BL, Hinck AP. ALK1 signaling in development and disease: new paradigms. Cell Mol Life Sci 2017; 74:4539-4560. [PMID: 28871312 PMCID: PMC5687069 DOI: 10.1007/s00018-017-2636-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 08/01/2017] [Accepted: 08/28/2017] [Indexed: 12/21/2022]
Abstract
Activin A receptor like type 1 (ALK1) is a transmembrane serine/threonine receptor kinase in the transforming growth factor-beta receptor family that is expressed on endothelial cells. Defects in ALK1 signaling cause the autosomal dominant vascular disorder, hereditary hemorrhagic telangiectasia (HHT), which is characterized by development of direct connections between arteries and veins, or arteriovenous malformations (AVMs). Although previous studies have implicated ALK1 in various aspects of sprouting angiogenesis, including tip/stalk cell selection, migration, and proliferation, recent work suggests an intriguing role for ALK1 in transducing a flow-based signal that governs directed endothelial cell migration within patent, perfused vessels. In this review, we present an updated view of the mechanism of ALK1 signaling, put forth a unified hypothesis to explain the cellular missteps that lead to AVMs associated with ALK1 deficiency, and discuss emerging roles for ALK1 signaling in diseases beyond HHT.
Collapse
Affiliation(s)
- Beth L Roman
- Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto St, Pittsburgh, PA, 15261, USA.
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Abstract
Correct organization of the vascular tree requires the balanced activities of several signaling pathways that regulate tubulogenesis and vascular branching, elongation, and pruning. When this balance is lost, the vessels can be malformed and fragile, and they can lose arteriovenous differentiation. In this review, we concentrate on the transforming growth factor (TGF)-β/bone morphogenetic protein (BMP) pathway, which is one of the most important and complex signaling systems in vascular development. Inactivation of these pathways can lead to altered vascular organization in the embryo. In addition, many vascular malformations are related to deregulation of TGF-β/BMP signaling. Here, we focus on two of the most studied vascular malformations that are induced by deregulation of TGF-β/BMP signaling: hereditary hemorrhagic telangiectasia (HHT) and cerebral cavernous malformation (CCM). The first of these is related to loss-of-function mutation of the TGF-β/BMP receptor complex and the second to increased signaling sensitivity to TGF-β/BMP. In this review, we discuss the potential therapeutic targets against these vascular malformations identified so far, as well as their basis in general mechanisms of vascular development and stability.
Collapse
Affiliation(s)
- Sara I Cunha
- From the Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden (S.I.C., P.U.M., E.D.); FIRC Institute of Molecular Oncology, Milan, Italy (E.D., M.G.L.); and Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy (M.G.L.)
| | - Peetra U Magnusson
- From the Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden (S.I.C., P.U.M., E.D.); FIRC Institute of Molecular Oncology, Milan, Italy (E.D., M.G.L.); and Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy (M.G.L.)
| | - Elisabetta Dejana
- From the Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden (S.I.C., P.U.M., E.D.); FIRC Institute of Molecular Oncology, Milan, Italy (E.D., M.G.L.); and Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy (M.G.L.).
| | - Maria Grazia Lampugnani
- From the Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden (S.I.C., P.U.M., E.D.); FIRC Institute of Molecular Oncology, Milan, Italy (E.D., M.G.L.); and Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy (M.G.L.)
| |
Collapse
|
21
|
Contribution of Inhibitor of DNA Binding/Differentiation-3 and Endocrine Disrupting Chemicals to Pathophysiological Aspects of Chronic Disease. BIOMED RESEARCH INTERNATIONAL 2017; 2017:6307109. [PMID: 28785583 PMCID: PMC5530454 DOI: 10.1155/2017/6307109] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/15/2017] [Accepted: 05/29/2017] [Indexed: 12/12/2022]
Abstract
The overwhelming increase in the global incidence of obesity and its associated complications such as insulin resistance, atherosclerosis, pulmonary disease, and degenerative disorders including dementia constitutes a serious public health problem. The Inhibitor of DNA Binding/Differentiation-3 (ID3), a member of the ID family of transcriptional regulators, has been shown to play a role in adipogenesis and therefore ID3 may influence obesity and metabolic health in response to environmental factors. This review will highlight the current understanding of how ID3 may contribute to complex chronic diseases via metabolic perturbations. Based on the increasing number of reports that suggest chronic exposure to and accumulation of endocrine disrupting chemicals (EDCs) within the human body are associated with metabolic disorders, we will also consider the impact of these chemicals on ID3. Improved understanding of the ID3 pathways by which exposure to EDCs can potentiate complex chronic diseases in populations with metabolic disorders (obesity, metabolic syndrome, and glucose intolerance) will likely provide useful knowledge in the prevention and control of complex chronic diseases associated with exposure to environmental pollutants.
Collapse
|
22
|
Zhang R, Zhu W, Su H. Vascular Integrity in the Pathogenesis of Brain Arteriovenous Malformation. ACTA NEUROCHIRURGICA. SUPPLEMENT 2016; 121:29-35. [PMID: 26463919 DOI: 10.1007/978-3-319-18497-5_6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Brain arteriovenous malformation (bAVM) is an important cause of intracranial hemorrhage (ICH), particularly in the young population. ICH is the first clinical symptom in about 50 % of bAVM patients. The vessels in bAVM are fragile and prone to rupture, causing bleeding into the brain. About 30 % of unruptured and non-hemorrhagic bAVMs demonstrate microscopic evidence of hemosiderin in the vascular wall. In bAVM mouse models, vascular mural cell coverage is reduced in the AVM lesion, accompanied by vascular leakage and microhemorrhage. In this review, we discuss possible signaling pathways involved in abnormal vascular development in bAVM.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, 1001 Potrero Avenue, 1363, San Francisco, CA, 94110, USA
| | - Wan Zhu
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, 1001 Potrero Avenue, 1363, San Francisco, CA, 94110, USA
| | - Hua Su
- Department of Anesthesia and Perioperative Care, Center for Cerebrovascular Research, University of California, San Francisco, 1001 Potrero Avenue, 1363, San Francisco, CA, 94110, USA.
| |
Collapse
|
23
|
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: 121] [Impact Index Per Article: 13.4] [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
|
24
|
Alam SK, Yadav VK, Bajaj S, Datta A, Dutta SK, Bhattacharyya M, Bhattacharya S, Debnath S, Roy S, Boardman LA, Smyrk TC, Molina JR, Chakrabarti S, Chowdhury S, Mukhopadhyay D, Roychoudhury S. DNA damage-induced ephrin-B2 reverse signaling promotes chemoresistance and drives EMT in colorectal carcinoma harboring mutant p53. Cell Death Differ 2015; 23:707-22. [PMID: 26494468 DOI: 10.1038/cdd.2015.133] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 07/31/2015] [Accepted: 08/25/2015] [Indexed: 01/13/2023] Open
Abstract
Mutation in the TP53 gene positively correlates with increased incidence of chemoresistance in different cancers. In this study, we investigated the mechanism of chemoresistance and epithelial-to-mesenchymal transition (EMT) in colorectal cancer involving the gain-of-function (GOF) mutant p53/ephrin-B2 signaling axis. Bioinformatic analysis of the NCI-60 data set and subsequent hub prediction identified EFNB2 as a possible GOF mutant p53 target gene, responsible for chemoresistance. We show that the mutant p53-NF-Y complex transcriptionally upregulates EFNB2 expression in response to DNA damage. Moreover, the acetylated form of mutant p53 protein is recruited on the EFNB2 promoter and positively regulates its expression in conjunction with coactivator p300. In vitro cell line and in vivo nude mice data show that EFNB2 silencing restores chemosensitivity in mutant p53-harboring tumors. In addition, we observed high expression of EFNB2 in patients having neoadjuvant non-responder colorectal carcinoma compared with those having responder version of the disease. In the course of deciphering the drug resistance mechanism, we also show that ephrin-B2 reverse signaling induces ABCG2 expression after drug treatment that involves JNK-c-Jun signaling in mutant p53 cells. Moreover, 5-fluorouracil-induced ephrin-B2 reverse signaling promotes tumorigenesis through the Src-ERK pathway, and drives EMT via the Src-FAK pathway. We thus conclude that targeting ephrin-B2 might enhance the therapeutic potential of DNA-damaging chemotherapeutic agents in mutant p53-bearing human tumors.
Collapse
Affiliation(s)
- S K Alam
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - V K Yadav
- G.N.R. Knowledge Centre for Genome Informatics, Proteomics and Structural Biology Unit, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, India
| | - S Bajaj
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Hospital/The Ontario Cancer Institute, Toronto, ON, Canada
| | - A Datta
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - S K Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - M Bhattacharyya
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - S Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - S Debnath
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - S Roy
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - L A Boardman
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - T C Smyrk
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - J R Molina
- Division of Medical Oncology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - S Chakrabarti
- Structural Biology and Bioinformatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - S Chowdhury
- G.N.R. Knowledge Centre for Genome Informatics, Proteomics and Structural Biology Unit, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, India.,Structural Biology Unit, CSIR-Institute of Genomics and Integrative Biology, Mall Road, Delhi, India
| | - D Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - S Roychoudhury
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| |
Collapse
|
25
|
Abstract
Formation of arterial vasculature, here termed arteriogenesis, is a central process in embryonic vascular development as well as in adult tissues. Although the process of capillary formation, angiogenesis, is relatively well understood, much remains to be learned about arteriogenesis. Recent discoveries point to the key role played by vascular endothelial growth factor receptor 2 in control of this process and to newly identified control circuits that dramatically influence its activity. The latter can present particularly attractive targets for a new class of therapeutic agents capable of activation of this signaling cascade in a ligand-independent manner, thereby promoting arteriogenesis in diseased tissues.
Collapse
Affiliation(s)
- Michael Simons
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (M.S., A.E.) and Departments of Cell Biology (M.S.) and Molecular Physiology (A.E.), Yale University School of Medicine, New Haven, CT.
| | - Anne Eichmann
- From the Department of Internal Medicine, Yale Cardiovascular Research Center, Section of Cardiovascular Medicine (M.S., A.E.) and Departments of Cell Biology (M.S.) and Molecular Physiology (A.E.), Yale University School of Medicine, New Haven, CT.
| |
Collapse
|
26
|
VEGF, Notch and TGFβ/BMPs in regulation of sprouting angiogenesis and vascular patterning. Biochem Soc Trans 2015; 42:1576-83. [PMID: 25399573 DOI: 10.1042/bst20140231] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The blood vasculature is constantly adapting to meet the demand from tissue. In so doing, branches may form, reorganize or regress. These complex processes employ integration of multiple signalling cascades, some of them being restricted to endothelial and mural cells and, hence, suitable for targeting of the vasculature. Both genetic and drug targeting experiments have demonstrated the requirement for the vascular endothelial growth factor (VEGF) system, the Delta-like-Notch system and the transforming growth factor β (TGFβ)/bone morphogenetic protein (BMP) cascades in vascular development. Although several of these signalling cascades in part converge into common downstream components, they differ in temporal and spatial regulation and expression. For example, the pro-angiogenic VEGFA is secreted by cells in need of oxygen, presented to the basal side of the endothelium, whereas BMP9 and BMP10 are supplied via the bloodstream in constant interaction with the apical side to suppress angiogenesis. Delta-like 4 (DLL4), on the other hand, is provided as an endothelial membrane bound ligand. In the present article, we discuss recent data on the integration of these pathways in the process of sprouting angiogenesis and vascular patterning and malformation.
Collapse
|
27
|
Rochon ER, Wright DS, Schubert MM, Roman BL. Context-specific interactions between Notch and ALK1 cannot explain ALK1-associated arteriovenous malformations. Cardiovasc Res 2015; 107:143-52. [PMID: 25969392 DOI: 10.1093/cvr/cvv148] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 05/07/2015] [Indexed: 01/17/2023] Open
Abstract
AIMS Notch and activin receptor-like kinase 1 (ALK1) have been implicated in arterial specification, angiogenic tip/stalk cell differentiation, and development of arteriovenous malformations (AVMs), and ALK1 can cooperate with Notch to up-regulate expression of Notch target genes in cultured endothelial cells. These findings suggest that Notch and ALK1 might collaboratively program arterial identity and prevent AVMs. We therefore sought to investigate the interaction between Notch and Alk1 signalling in the developing vertebrate vasculature. METHODS AND RESULTS We modulated Notch and Alk1 activities in zebrafish embryos and examined effects on Notch target gene expression and vascular morphology. Although Alk1 is not necessary for expression of Notch target genes in arterial endothelium, loss of Notch signalling unmasks a role for Alk1 in supporting hey2 and ephrinb2a expression in the dorsal aorta. In contrast, Notch and Alk1 play opposing roles in hey2 expression in cranial arteries and dll4 expression in all arterial endothelium, with Notch inducing and Alk1 repressing these genes. Although alk1 loss increases expression of dll4, AVMs in alk1 mutants could neither be phenocopied by Notch activation nor rescued by Dll4/Notch inhibition. CONCLUSION Control of Notch targets in arterial endothelium is context-dependent, with gene-specific and region-specific requirements for Notch and Alk1. Alk1 is not required for arterial identity, and perturbations in Notch signalling cannot account for alk1 mutant-associated AVMs. These data suggest that AVMs associated with ALK1 mutation are not caused by defective arterial specification or altered Notch signalling.
Collapse
Affiliation(s)
- Elizabeth R Rochon
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Daniel S Wright
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Max M Schubert
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Beth L Roman
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA Department of Human Genetics, University of Pittsburgh Graduate School of Public Health, 130 DeSoto St, Pittsburgh, PA 15261, USA
| |
Collapse
|
28
|
Lauzon MA, Daviau A, Marcos B, Faucheux N. Growth factor treatment to overcome Alzheimer's dysfunctional signaling. Cell Signal 2015; 27:1025-38. [PMID: 25744541 DOI: 10.1016/j.cellsig.2015.02.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 02/16/2015] [Indexed: 10/23/2022]
Abstract
The number of people suffering from Alzheimer's disease (AD) will increase as the world population ages, creating a huge socio-economic burden. The three pathophysiological hallmarks of AD are the cholinergic system dysfunction, the β-amyloid peptide deposition and the Tau protein hyperphosphorylation. Current treatments have only transient effects and each tends to concentrate on a single pathophysiological aspect of AD. This review first provides an overall view of AD in terms of its pathophysiological symptoms and signaling dysfunction. We then examine the therapeutic potential of growth factors (GFs) by showing how they can overcome the dysfunctional cell signaling that occurs in AD. Finally, we discuss new alternatives to GFs that help overcome the problem of brain uptake, such as small peptides, with evidence from some of our unpublished data on human neuronal cell line.
Collapse
Affiliation(s)
- Marc-Antoine Lauzon
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Alex Daviau
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Bernard Marcos
- Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada
| | - Nathalie Faucheux
- Cell-Biomaterial Biohybrid Systems, Department of Chemical and Biotechnological Engineering, Université de Sherbrooke, 2500 boul. de l'Université, Sherbrooke, Québec J1K 2R1, Canada.
| |
Collapse
|
29
|
Thalgott J, Dos-Santos-Luis D, Lebrin F. Pericytes as targets in hereditary hemorrhagic telangiectasia. Front Genet 2015; 6:37. [PMID: 25763012 PMCID: PMC4327729 DOI: 10.3389/fgene.2015.00037] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/26/2015] [Indexed: 12/04/2022] Open
Abstract
Defective paracrine Transforming Growth Factor-β (TGF-β) signaling between endothelial cells and the neighboring mural cells have been thought to lead to the development of vascular lesions that are characteristic of Hereditary Hemorrhagic Telangiectasia (HHT). This review highlights recent progress in our understanding of TGF-β signaling in mural cell recruitment and vessel stabilization and how perturbed TGF-β signaling might contribute to defective endothelial-mural cell interaction affecting vessel functionalities. Our recent findings have provided exciting insights into the role of thalidomide, a drug that reduces both the frequency and the duration of epistaxis in individuals with HHT by targeting mural cells. These advances provide opportunities for the development of new therapies for vascular malformations.
Collapse
Affiliation(s)
- Jérémy Thalgott
- INSERM, Center for Interdisciplinary Research in Biology, UMR CNRS 7241/INSERM U1050, Group Pathological Angiogenesis and Vessel Normalization, Collège de France Paris, France
| | - Damien Dos-Santos-Luis
- INSERM, Center for Interdisciplinary Research in Biology, UMR CNRS 7241/INSERM U1050, Group Pathological Angiogenesis and Vessel Normalization, Collège de France Paris, France
| | - Franck Lebrin
- INSERM, Center for Interdisciplinary Research in Biology, UMR CNRS 7241/INSERM U1050, Group Pathological Angiogenesis and Vessel Normalization, Collège de France Paris, France
| |
Collapse
|
30
|
Roman BL, Finegold DN. Genetic and Molecular Basis for Hereditary Hemorrhagic Telangiectasia. CURRENT GENETIC MEDICINE REPORTS 2014. [DOI: 10.1007/s40142-014-0061-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
31
|
Arnold TD, Niaudet C, Pang MF, Siegenthaler J, Gaengel K, Jung B, Ferrero GM, Mukouyama YS, Fuxe J, Akhurst R, Betsholtz C, Sheppard D, Reichardt LF. Excessive vascular sprouting underlies cerebral hemorrhage in mice lacking αVβ8-TGFβ signaling in the brain. Development 2014; 141:4489-99. [PMID: 25406396 DOI: 10.1242/dev.107193] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Vascular development of the central nervous system and blood-brain barrier (BBB) induction are closely linked processes. The role of factors that promote endothelial sprouting and vascular leak, such as vascular endothelial growth factor A, are well described, but the factors that suppress angiogenic sprouting and their impact on the BBB are poorly understood. Here, we show that integrin αVβ8 activates angiosuppressive TGFβ gradients in the brain, which inhibit endothelial cell sprouting. Loss of αVβ8 in the brain or downstream TGFβ1-TGFBR2-ALK5-Smad3 signaling in endothelial cells increases vascular sprouting, branching and proliferation, leading to vascular dysplasia and hemorrhage. Importantly, BBB function in Itgb8 mutants is intact during early stages of vascular dysgenesis before hemorrhage. By contrast, Pdgfb(ret/ret) mice, which exhibit severe BBB disruption and vascular leak due to pericyte deficiency, have comparatively normal vascular morphogenesis and do not exhibit brain hemorrhage. Our data therefore suggest that abnormal vascular sprouting and patterning, not BBB dysfunction, underlie developmental cerebral hemorrhage.
Collapse
Affiliation(s)
- Thomas D Arnold
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94158, USA Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-177 77 Stockholm, Sweden
| | - Colin Niaudet
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-177 77 Stockholm, Sweden
| | - Mei-Fong Pang
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-177 77 Stockholm, Sweden
| | - Julie Siegenthaler
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Konstantin Gaengel
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-177 77 Stockholm, Sweden
| | - Bongnam Jung
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-177 77 Stockholm, Sweden
| | - Gina M Ferrero
- Department of Physiology and Neuroscience Program, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yoh-suke Mukouyama
- Laboratory of Stem Cell and Neuro-Vascular Biology, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD 20892, USA
| | - Jonas Fuxe
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-177 77 Stockholm, Sweden
| | - Rosemary Akhurst
- Helen Diller Cancer Center and Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Christer Betsholtz
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-177 77 Stockholm, Sweden
| | - Dean Sheppard
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Louis F Reichardt
- Department of Physiology and Neuroscience Program, University of California, San Francisco, San Francisco, CA 94158, USA
| |
Collapse
|
32
|
Rostama B, Peterson SM, Vary CPH, Liaw L. Notch signal integration in the vasculature during remodeling. Vascul Pharmacol 2014; 63:97-104. [PMID: 25464152 PMCID: PMC4304902 DOI: 10.1016/j.vph.2014.10.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 10/06/2014] [Accepted: 10/10/2014] [Indexed: 02/06/2023]
Abstract
Notch signaling plays many important roles in homeostasis and remodeling in the vessel wall, and serves a critical role in the communication between endothelial cells and smooth muscle cells. Within blood vessels, Notch signaling integrates with multiple pathways by mechanisms including direct protein–protein interaction, cooperative or synergistic regulation of signal cascades, and co-regulation of transcriptional targets. After establishment of the mature blood vessel, the spectrum and intensity of Notch signaling change during phases of active remodeling or disease progression. These changes can be mediated by regulation via microRNAs and protein stability or signaling, and corresponding changes in complementary signaling pathways. Notch also affects endothelial cells on a system level by regulating key metabolic components. This review will outline the most recent findings of Notch activity in blood vessels, with a focus on how Notch signals integrate with other molecular signaling pathways controlling vascular phenotype.
Collapse
Affiliation(s)
- Bahman Rostama
- Center for Molecular Medicine, Maine Medical Center Research Institute, USA
| | | | | | | |
Collapse
|
33
|
Nielsen CM, Cuervo H, Ding VW, Kong Y, Huang EJ, Wang RA. Deletion of Rbpj from postnatal endothelium leads to abnormal arteriovenous shunting in mice. Development 2014; 141:3782-92. [PMID: 25209249 DOI: 10.1242/dev.108951] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Arteriovenous malformations (AVMs) are tortuous vessels characterized by arteriovenous (AV) shunts, which displace capillaries and shunt blood directly from artery to vein. Notch signaling regulates embryonic AV specification by promoting arterial, as opposed to venous, endothelial cell (EC) fate. To understand the essential role of endothelial Notch signaling in postnatal AV organization, we used inducible Cre-loxP recombination to delete Rbpj, a mediator of canonical Notch signaling, from postnatal ECs in mice. Deletion of endothelial Rbpj from birth resulted in features of AVMs by P14, including abnormal AV shunting and tortuous vessels in the brain, intestine and heart. We further analyzed brain AVMs, as they pose particular health risks. Consistent with AVM pathology, we found cerebral hemorrhage, hypoxia and necrosis, and neurological deficits. AV shunts originated from capillaries (and possibly venules), with the earliest detectable morphological abnormalities in AV connections by P8. Prior to AV shunt formation, alterations in EC gene expression were detected, including decreased Efnb2 and increased Pai1, which encodes a downstream effector of TGFβ signaling. After AV shunts had formed, whole-mount immunostaining showed decreased Efnb2 and increased Ephb4 expression within AV shunts, suggesting that ECs were reprogrammed from arterial to venous identity. Deletion of Rbpj from adult ECs led to tortuosities in gastrointestinal, uterine and skin vascular beds, but had mild effects in the brain. Our results demonstrate a temporal requirement for Rbpj in postnatal ECs to maintain proper artery, capillary and vein organization and to prevent abnormal AV shunting and AVM pathogenesis.
Collapse
Affiliation(s)
- Corinne M Nielsen
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Henar Cuervo
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Vivianne W Ding
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Yupeng Kong
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| | - Eric J Huang
- Department of Pathology, University of California, San Francisco, CA 94143, USA
| | - Rong A Wang
- Laboratory for Accelerated Vascular Research, Department of Surgery, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
34
|
Han C, Choe SW, Kim YH, Acharya AP, Keselowsky BG, Sorg BS, Lee YJ, Oh SP. VEGF neutralization can prevent and normalize arteriovenous malformations in an animal model for hereditary hemorrhagic telangiectasia 2. Angiogenesis 2014; 17:823-830. [PMID: 24957885 DOI: 10.1007/s10456-014-9436-3] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 06/05/2014] [Indexed: 12/18/2022]
Abstract
Arteriovenous malformation (AVM) refers to a vascular anomaly where arteries and veins are directly connected through a complex, tangled web of abnormal AV fistulae without a normal capillary network. Hereditary hemorrhagic telangiectasia (HHT) types 1 and 2 arise from heterozygous mutations in endoglin (ENG) and activin receptor-like kinase 1 (ALK1), respectively. HHT patients possess AVMs in various organs, and telangiectases (small AVMs) along the mucocutaneous surface. Understanding why and how AVMs develop is crucial for developing therapies to inhibit the formation, growth, or maintenance of AVMs in HHT patients. Previously, we have shown that secondary factors such as wounding are required for Alk1-deficient vessels to develop skin AVMs. Here, we present evidences that AVMs establish from nascent arteries and veins rather than from remodeling of a preexistent capillary network in the wound-induced skin AVM model. We also show that VEGF can mimic the wound effect on skin AVM formation, and VEGF-neutralizing antibody can prevent skin AVM formation and ameliorate internal bleeding in Alk1-deficient adult mice. With topical applications at different stages of AVM development, we demonstrate that the VEGF blockade can prevent the formation of AVM and cease the progression of AVM development. Taken together, the presented experimental model is an invaluable system for precise molecular mechanism of action of VEGF blockades as well as for preclinical screening of drug candidates for epistaxis and gastrointestinal bleedings.
Collapse
Affiliation(s)
- Chul Han
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Se-Woon Choe
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32610.,Department of Biomedical Engineering, Tongmyong University, Busan, Republic of Korea
| | - Yong Hwan Kim
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32610
| | - Abhinav P Acharya
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL 32611
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL 32611
| | - Brian S Sorg
- J. Crayton Pruitt Family Department of Biomedical Engineering, College of Engineering, University of Florida, Gainesville, FL 32611
| | - Young-Jae Lee
- World Class University program, Lee Gil Ya Cancer and Diabetes Institute, Gachon University of Medicine and Science, Incheon, Republic of Korea
| | - S Paul Oh
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, FL 32610.,World Class University program, Lee Gil Ya Cancer and Diabetes Institute, Gachon University of Medicine and Science, Incheon, Republic of Korea
| |
Collapse
|
35
|
Genetic variants of Adam17 differentially regulate TGFβ signaling to modify vascular pathology in mice and humans. Proc Natl Acad Sci U S A 2014; 111:7723-8. [PMID: 24812125 DOI: 10.1073/pnas.1318761111] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Outcome of TGFβ1 signaling is context dependent and differs between individuals due to germ-line genetic variation. To explore innate genetic variants that determine differential outcome of reduced TGFβ1 signaling, we dissected the modifier locus Tgfbm3, on mouse chromosome 12. On a NIH/OlaHsd genetic background, the Tgfbm3b(C57) haplotype suppresses prenatal lethality of Tgfb1(-/-) embryos and enhances nuclear accumulation of mothers against decapentaplegic homolog 2 (Smad2) in embryonic cells. Amino acid polymorphisms within a disintegrin and metalloprotease 17 (Adam17) can account, at least in part, for this Tgfbm3b effect. ADAM17 is known to down-regulate Smad2 signaling by shedding the extracellular domain of TGFβRI, and we show that the C57 variant is hypomorphic for down-regulation of Smad2/3-driven transcription. Genetic variation at Tgfbm3 or pharmacological inhibition of ADAM17, modulates postnatal circulating endothelial progenitor cell (CEPC) numbers via effects on TGFβRI activity. Because CEPC numbers correlate with angiogenic potential, this suggests that variant Adam17 is an innate modifier of adult angiogenesis, acting through TGFβR1. To determine whether human ADAM17 is also polymorphic and interacts with TGFβ signaling in human vascular disease, we investigated hereditary hemorrhagic telangiectasia (HHT), which is caused by mutations in TGFβ/bone morphogenetic protein receptor genes, ENG, encoding endoglin (HHT1), or ACVRL1 encoding ALK1 (HHT2), and considered a disease of excessive abnormal angiogenesis. HHT manifests highly variable incidence and severity of clinical features, ranging from small mucocutaneous telangiectases to life-threatening visceral and cerebral arteriovenous malformations (AVMs). We show that ADAM17 SNPs associate with the presence of pulmonary AVM in HHT1 but not HHT2, indicating genetic variation in ADAM17 can potentiate a TGFβ-regulated vascular disease.
Collapse
|
36
|
Potential roles of bone morphogenetic protein (BMP)-9 in human liver diseases. Int J Mol Sci 2014; 15:5199-220. [PMID: 24670474 PMCID: PMC4013558 DOI: 10.3390/ijms15045199] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 03/07/2014] [Accepted: 03/17/2014] [Indexed: 01/01/2023] Open
Abstract
Bone morphogenetic proteins (BMP-2 to BMP-15) belong to the Transforming Growth Factor (TGF)-β superfamily and, besides their well-documented roles during embryogenesis and bone formation, some of them have recently been described to be involved in the pathogenesis of different organs, including the liver. The role of BMPs in liver damage responses including hepatocellular carcinoma (HCC) development has only begun to be addressed and strong evidence supports the concept of a pro-tumorigenic role of BMP signaling in HCC cells. BMP-9 (also termed Growth and Differentiation Factor (GDF)-2) represents the most recently discovered member of the BMP family. We have previously demonstrated that in HCC patient samples BMP-9 expression was positively associated with the tumor seize (“T stage”) and that it enhanced cell migration and induced epithelial to mesenchymal transition (EMT) in HCC cells in vitro. In another study we recently found that BMP-9 promotes growth in HCC cells, but not in non-transformed hepatocytes. Published as well as unpublished results obtained with primary hepatocytes support the concept of a dual function of BMP-9 in the liver: while in primary, non-malignant cells BMP-9 stabilizes the epithelial phenotype and inhibits proliferation, in HCC cells it induces cell growth and the acquisition of a migratory phenotype. In this review article we summarize current knowledge about BMPs in liver diseases, with special focus on the role of BMP-9 in HCC development and progression, that may provide new clues for a better understanding of the contribution of BMP-signaling to chronic liver diseases.
Collapse
|
37
|
Atri D, Larrivée B, Eichmann A, Simons M. Endothelial signaling and the molecular basis of arteriovenous malformation. Cell Mol Life Sci 2013; 71:10.1007/s00018-013-1475-1. [PMID: 24077895 PMCID: PMC3969452 DOI: 10.1007/s00018-013-1475-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 09/08/2013] [Accepted: 09/10/2013] [Indexed: 12/21/2022]
Abstract
Arteriovenous malformations occur when abnormalities of vascular patterning result in the flow of blood from arteries to veins without an intervening capillary bed. Recent work has revealed the importance of the Notch and TGF-β signaling pathways in vascular patterning. Specifically, Notch signaling has an increasingly apparent role in arterial specification and suppression of branching, whereas TGF-β is implicated in vascular smooth muscle development and remodeling under angiogenic stimuli. These physiologic roles, consequently, have implicated both pathways in the pathogenesis of arteriovenous malformation. In this review, we summarize the studies of endothelial signaling that contribute to arteriovenous malformation and the roles of genes implicated in their pathogenesis. We further discuss how endothelial signaling may contribute to vascular smooth muscle development and how knowledge of signaling pathways may provide us targets for medical therapy in these vascular lesions.
Collapse
Affiliation(s)
- Deepak Atri
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, United States
| | - Bruno Larrivée
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, United States
- Department of Ophthalmology, Hôpital Maisonneuve-Rosemont Research Centre, University of Montreal, Montreal, Canada
| | - Anne Eichmann
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, United States
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, Paris, France
| | - Michael Simons
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, United States
| |
Collapse
|
38
|
Hawinkels LJ, Garcia de Vinuesa A, Ten Dijke P. Activin receptor-like kinase 1 as a target for anti-angiogenesis therapy. Expert Opin Investig Drugs 2013; 22:1371-83. [PMID: 24053899 DOI: 10.1517/13543784.2013.837884] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Formation of blood vessels from pre-existing ones, also termed angiogenesis, is of crucial importance for the outgrowth of tumours beyond 1 - 2 mm³. Therefore, anti-angiogenic therapies, mainly focussing on inhibition of vascular endothelial growth factor (VEGF) are used in clinical therapy. However, although initially reducing tumour size, therapy resistance occurs frequently and new targets are needed. A possible target is activin receptor-like kinase (ALK)-1, a transforming growth factor (TGF)-β type-I receptor, which binds bone morphogenetic protein (BMP)-9 and -10 with high affinity and has an important role in regulating angiogenesis. AREAS COVERED Several approaches to interfere with ALK1 signalling have been developed, that is, ALK1 neutralising antibodies and a soluble ALK1 extracellular domain/Fc fusion protein (ALK1-Fc), acting as a ligand trap. In this review, we discuss the involvement of ALK1 in angiogenesis, in a variety of diseases and the current status of the development of ALK1 inhibitors for cancer therapy. EXPERT OPINION Based on current, mainly preclinical studies on inhibition of ALK1 signalling by ligand traps and neutralising antibodies, targeting ALK1 seems very promising. Both ALK1-Fc and neutralising antibodies strongly inhibit angiogenesis in vitro and in vivo. The results from the first Phase I clinical trials are to be reported soon and multiple Phase II studies are ongoing.
Collapse
Affiliation(s)
- Lukas Jac Hawinkels
- Leiden University Medical Centre, Cancer Genomics Centre Netherlands and Centre for BioMedical Genetics, Department of Molecular Cell Biology , Building-2, S1-P, PO box 9600, 2300 RC Leiden , The Netherlands +31 71 526 9272 ; +31 71 526 8270 ;
| | | | | |
Collapse
|
39
|
Liu X, Qin J, Luo Q, Bi Y, Zhu G, Jiang W, Kim SH, Li M, Su Y, Nan G, Cui J, Zhang W, Li R, Chen X, Kong Y, Zhang J, Wang J, Rogers MR, Zhang H, Shui W, Zhao C, Wang N, Liang X, Wu N, He Y, Luu HH, Haydon RC, Shi LL, Li T, He TC, Li M. Cross-talk between EGF and BMP9 signalling pathways regulates the osteogenic differentiation of mesenchymal stem cells. J Cell Mol Med 2013; 17:1160-72. [PMID: 23844832 PMCID: PMC4118175 DOI: 10.1111/jcmm.12097] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 05/13/2013] [Accepted: 05/20/2013] [Indexed: 01/13/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent progenitors, which give rise to several lineages, including bone, cartilage and fat. Epidermal growth factor (EGF) stimulates cell growth, proliferation and differentiation. EGF acts by binding with high affinity to epidermal growth factor receptor (EGFR) on the cell surface and stimulating the intrinsic protein tyrosine kinase activity of its receptor, which initiates a signal transduction cascade causing a variety of biochemical changes within the cell and regulating cell proliferation and differentiation. We have identified BMP9 as one of the most osteogenic BMPs in MSCs. In this study, we investigate if EGF signalling cross-talks with BMP9 and regulates BMP9-induced osteogenic differentiation. We find that EGF potentiates BMP9-induced early and late osteogenic markers of MSCs in vitro, which can be effectively blunted by EGFR inhibitors Gefitinib and Erlotinib or receptor tyrosine kinase inhibitors AG-1478 and AG-494 in a dose- and time-dependent manner. Furthermore, EGF significantly augments BMP9-induced bone formation in the cultured mouse foetal limb explants. In vivo stem cell implantation experiment reveals that exogenous expression of EGF in MSCs can effectively potentiate BMP9-induced ectopic bone formation, yielding larger and more mature bone masses. Interestingly, we find that, while EGF can induce BMP9 expression in MSCs, EGFR expression is directly up-regulated by BMP9 through Smad1/5/8 signalling pathway. Thus, the cross-talk between EGF and BMP9 signalling pathways in MSCs may underline their important roles in regulating osteogenic differentiation. Harnessing the synergy between BMP9 and EGF should be beneficial for enhancing osteogenesis in regenerative medicine.
Collapse
Affiliation(s)
- Xing Liu
- Stem Cell Biology and Therapy Laboratory of the Key Laboratory for Pediatrics designated by Chinese Ministry of Education and Chongqing Bureau of Education, Department of Orthopaedic Surgery, The Children's Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Yang J, Li X, Li Y, Southwood M, Ye L, Long L, Al-Lamki RS, Morrell NW. Id proteins are critical downstream effectors of BMP signaling in human pulmonary arterial smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2013; 305:L312-21. [PMID: 23771884 DOI: 10.1152/ajplung.00054.2013] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Bone morphogenetic protein type II receptor (BMPR-II) mutations are responsible for over 70% of cases of heritable pulmonary arterial hypertension (PAH). Loss of BMP signaling promotes pulmonary vascular remodeling via modulation of pulmonary artery smooth muscle cell (PASMC) proliferation. Id proteins (Id1-4) are major downstream transcriptional targets of BMP signaling. However, the impact of BMPR-II mutation on the expression of the range of Id proteins and the contribution of individual Id proteins to abnormal PASMC function remain unclear. Human PASMCs were used to determine the expression of Id proteins (Id1-4) by real-time PCR and immunoblotting. The BMP responses in control cells were compared with PASMCs harboring BMPR-II mutations and cells in which BMPR-II was knocked down by siRNA transfection. Id3 expression in pulmonary vessels was also investigated in BMPR-II mutant mice and in patients with heritable PAH. BMP4 and BMP6, but not BMP9, induced mRNA expression of Id1, Id2, and Id3. The BMP-stimulated induction of Id1 and Id3 was markedly reduced in BMPR-II mutant PASMCs and in control PASMCs following siRNA silencing of BMPR-II. Pulmonary arteries in BMPR-II mutant mice and patients with heritable PAH demonstrated reduced levels of Id3 compared with control subjects. Lentiviral overexpression of Id3 reduced cell cycle progression and inhibited proliferation of PASMCs. Lipopolysaccharide further reduced Id3 expression in mutant PASMCs. In conclusion, Id proteins, and particularly Id1 and Id3, are critical downstream effectors of BMP signaling in PASMCs. Loss of BMPR-II function reduces the induction of Id genes in PASMCs, Id1, and Id3 regulate the proliferation of PASMCs via cell cycle inhibition, an effect that may be exacerbated by inflammatory stimuli.
Collapse
Affiliation(s)
- Jun Yang
- Dept. of Medicine, Level 5, Box 157 Addenbrooke's Hospitals, Hills Rd., Cambridge, CB2 0QQ, UK..
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Bone morphogenetic protein 9 (BMP9) controls lymphatic vessel maturation and valve formation. Blood 2013; 122:598-607. [PMID: 23741013 DOI: 10.1182/blood-2012-12-472142] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Lymphatic vessels are critical for the maintenance of tissue fluid homeostasis and their dysfunction contributes to several human diseases. The activin receptor-like kinase 1 (ALK1) is a transforming growth factor-β family type 1 receptor that is expressed on both blood and lymphatic endothelial cells (LECs). Its high-affinity ligand, bone morphogenetic protein 9 (BMP9), has been shown to be critical for retinal angiogenesis. The aim of this work was to investigate whether BMP9 could play a role in lymphatic development. We found that Bmp9 deficiency in mice causes abnormal lymphatic development. Bmp9-knockout (KO) pups presented hyperplastic mesenteric collecting vessels that maintained LYVE-1 expression. In accordance with this result, we found that BMP9 inhibited LYVE-1 expression in LECs in an ALK1-dependent manner. Bmp9-KO pups also presented a significant reduction in the number and in the maturation of mesenteric lymphatic valves at embryonic day 18.5 and at postnatal days 0 and 4. Interestingly, the expression of several genes known to be involved in valve formation (Foxc2, Connexin37, EphrinB2, and Neuropilin1) was upregulated by BMP9 in LECS. Finally, we demonstrated that Bmp9-KO neonates and adult mice had decreased lymphatic draining efficiency. These data identify BMP9 as an important extracellular regulator in the maturation of the lymphatic vascular network affecting valve development and lymphatic vessel function.
Collapse
|
42
|
Choi EJ, Kim YH, Choe SW, Tak YG, Garrido-Martin EM, Chang M, Lee YJ, Oh SP. Enhanced responses to angiogenic cues underlie the pathogenesis of hereditary hemorrhagic telangiectasia 2. PLoS One 2013; 8:e63138. [PMID: 23675457 PMCID: PMC3651154 DOI: 10.1371/journal.pone.0063138] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 03/28/2013] [Indexed: 12/11/2022] Open
Abstract
Hereditary Hemorrhagic Telangiectasia (HHT) is a genetic vascular disease in which arteriovenous malformations (AVMs) manifest in skin and multiple visceral organs. HHT is caused by heterozygous mutations in endoglin (ENG), activin receptor-like kinase 1 (ALK1), or SMAD4. ALK1 regulates angiogenesis, but the precise function of ALK1 in endothelial cells (ECs) remains elusive. Since most blood vessels of HHT patients do not produce pathological vascular lesions, ALK1 heterozygous ECs may be normal unless additional genetic or environmental stresses are imposed. To investigate the cellular and biochemical phenotypes of Alk1-null versus Alk1-heterozygous ECs, we have generated pulmonary EC lines in which a genotype switch from the Alk1-conditional allele (Alk1 (2f)) to the Alk1-null allele (Alk1 (1f)) can be induced by tamoxifen treatment. Alk1-null (1 f/1 f) ECs displayed increased migratory properties in vitro in response to bFGF compared with Alk1-het (2 f/1 f) ECs. The 1 f/1 f-ECs formed a denser and more persistent tubular network as compared with their parental 2 f/1 f-ECs. Interestingly, the response to BMP-9 on SMAD1/5 phosphorylation was impaired in both 2 f/1 f- and 1 f/1 f-ECs at a comparable manner, suggesting that other factors in addition to SMADs may play a crucial role for enhanced angiogenic activity in 1 f/1 f-ECs. We also demonstrated in vivo that Alk1-deficient ECs exhibited high migratory and invasive properties. Taken together, these data suggest that enhanced responses to angiogenic cues in ALK1-deficient ECs underlie the pathogenesis of HHT2.
Collapse
Affiliation(s)
- Eun-Jung Choi
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Yong Hwan Kim
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Se-woon Choe
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Yu Gyoung Tak
- World Class University Program, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Eva M. Garrido-Martin
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, United States of America
| | - Myron Chang
- Department of Biostatistics, University of Florida, Gainesville, Florida, United States of America
| | - Young Jae Lee
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, United States of America
- World Class University Program, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - S. Paul Oh
- Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville, Florida, United States of America
- World Class University Program, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
- * E-mail:
| |
Collapse
|
43
|
Beets K, Huylebroeck D, Moya IM, Umans L, Zwijsen A. Robustness in angiogenesis: notch and BMP shaping waves. Trends Genet 2012; 29:140-9. [PMID: 23279848 DOI: 10.1016/j.tig.2012.11.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 10/31/2012] [Accepted: 11/16/2012] [Indexed: 12/20/2022]
Abstract
Vascular patterning involves sprouting of blood vessels, which is governed by orchestrated communication between cells in the surrounding tissue and endothelial cells (ECs) lining the blood vessels. Single ECs are selected for sprouting by hypoxia-induced stimuli and become the 'tip' or leader cell that guides new sprouts. The 'stalk' or trailing ECs proliferate for tube extension and lumenize the nascent vessel. Stalk and tip cells can dynamically switch their identities during this process in a Notch-dependent manner. Here, we review recent studies showing that bone morphogenetic protein (BMP) signaling coregulates Notch target genes in ECs. In particular, we focus on how Delta-like ligand 4 (DLL4)-Notch and BMP effector interplay may drive nonsynchronized oscillatory gene expression in ECs essential for setting sharp tip-stalk cell boundaries while sustaining a dynamic pool of nonsprouting ECs. Deeper knowledge about the coregulation of vessel plasticity in different vascular beds may result in refinement of anti-angiogenesis and vessel normalization therapies.
Collapse
Affiliation(s)
- Karen Beets
- Laboratory of Developmental Signaling, VIB Center for the Biology of Disease, VIB, 3000 Leuven, Belgium
| | | | | | | | | |
Collapse
|
44
|
Abstract
BMP9 signaling has been implicated in hereditary hemorrhagic telangiectasia (HHT) and vascular remodeling, acting via the HHT target genes, endoglin and ALK1. This study sought to identify endothelial BMP9-regulated proteins that could affect the HHT phenotype. Gene ontology analysis of cDNA microarray data obtained after BMP9 treatment of primary human endothelial cells indicated regulation of chemokine, adhesion, and inflammation pathways. These responses included the up-regulation of the chemokine CXCL12/SDF1 and down-regulation of its receptor CXCR4. Quantitative mass spectrometry identified additional secreted proteins, including the chemokine CXCL10/IP10. RNA knockdown of endoglin and ALK1 impaired SDF1/CXCR4 regulation by BMP9. Because of the association of SDF1 with ischemia, we analyzed its expression under hypoxia in response to BMP9 in vitro, and during the response to hindlimb ischemia, in endoglin-deficient mice. BMP9 and hypoxia were additive inducers of SDF1 expression. Moreover, the data suggest that endoglin deficiency impaired SDF1 expression in endothelial cells in vivo. Our data implicate BMP9 in regulation of the SDF1/CXCR4 chemokine axis in endothelial cells and point to a role for BMP9 signaling via endoglin in a switch from an SDF1-responsive autocrine phenotype to an SDF1 nonresponsive paracrine state that represses endothelial cell migration and may promote vessel maturation.
Collapse
|