151
|
Saito T, Bokhove M, Croci R, Zamora-Caballero S, Han L, Letarte M, de Sanctis D, Jovine L. Structural Basis of the Human Endoglin-BMP9 Interaction: Insights into BMP Signaling and HHT1. Cell Rep 2018; 19:1917-1928. [PMID: 28564608 PMCID: PMC5464963 DOI: 10.1016/j.celrep.2017.05.011] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/30/2017] [Accepted: 05/02/2017] [Indexed: 11/25/2022] Open
Abstract
Endoglin (ENG)/CD105 is an essential endothelial cell co-receptor of the transforming growth factor β (TGF-β) superfamily, mutated in hereditary hemorrhagic telangiectasia type 1 (HHT1) and involved in tumor angiogenesis and preeclampsia. Here, we present crystal structures of the ectodomain of human ENG and its complex with the ligand bone morphogenetic protein 9 (BMP9). BMP9 interacts with a hydrophobic surface of the N-terminal orphan domain of ENG, which adopts a new duplicated fold generated by circular permutation. The interface involves residues mutated in HHT1 and overlaps with the epitope of tumor-suppressing anti-ENG monoclonal TRC105. The structure of the C-terminal zona pellucida module suggests how two copies of ENG embrace homodimeric BMP9, whose binding is compatible with ligand recognition by type I but not type II receptors. These findings shed light on the molecular basis of the BMP signaling cascade, with implications for future therapeutic interventions in this fundamental pathway. Crystal structures of human ENG and its complex with BMP9 were determined The orphan domain of ENG adopts a fold that explains the effect of HHT1 mutations ZP module-mediated dimerization of ENG creates a clamp that secures homodimeric BMP9 ENG-bound BMP9 can interact with the ALK1 receptor but not the ActRIIB receptor
Collapse
Affiliation(s)
- Takako Saito
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Karolinska Institutet, Huddinge 14183, Sweden
| | - Marcel Bokhove
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Karolinska Institutet, Huddinge 14183, Sweden
| | - Romina Croci
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Karolinska Institutet, Huddinge 14183, Sweden
| | - Sara Zamora-Caballero
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Karolinska Institutet, Huddinge 14183, Sweden
| | - Ling Han
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Karolinska Institutet, Huddinge 14183, Sweden
| | - Michelle Letarte
- Molecular Medicine, Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, University of Toronto, Toronto, ON M5G 0A4, Canada; Department of Immunology, University of Toronto, Toronto, ON M5G 0A4, Canada
| | | | - Luca Jovine
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Karolinska Institutet, Huddinge 14183, Sweden.
| |
Collapse
|
152
|
Endoglin haploinsufficiency is associated with differential regulation of extracellular matrix production during skin fibrosis and cartilage repair in mice. J Cell Commun Signal 2018; 12:379-388. [PMID: 29488175 DOI: 10.1007/s12079-018-0461-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 02/07/2018] [Indexed: 01/17/2023] Open
Abstract
Transforming growth factor (TGF)-β is a multifunctional growth factor with potent pro-fibrotic effects. Endoglin is a TGF-β co-receptor that strongly regulates TGF-β signaling in a variety of cell types. Although aberrant regulation of TGF-β signaling is known to play a key role in fibrotic diseases such as scleroderma and impaired cartilage repair, the significance of endoglin function in regulating these processes is poorly understood. Here we examined whether endoglin haploinsufficiency regulates extracellular (ECM) protein expression and fibrotic responses during bleomycin induced skin fibrosis and surgically induced osteoarthritis, using endoglin-heterozygous (Eng+/-) mice and wild-type (Eng+/+) littermates. Skin fibrosis was induced by injecting mice intradermally with bleomycin or vehicle. Osteoarthritis was induced surgically by destabilization of medial meniscus. Dermal thickness, cartilage integrity and ECM protein expression were then determined. Eng+/- mice subjected to bleomycin challenge show a marked decrease in dermal thickness (P < 0.005) and reduced collagen content and decreased collagen I, fibronectin, alpha-smooth muscle actin levels as compared to Eng+/+ mice, both under basal and bleomycin treated conditions. Eng+/- mice undergoing surgically induced osteoarthritis show no differences in the degree of cartilage degradation, as compared to Eng+/+ mice, although chondrocytes isolated from Eng+/- display markedly enhanced collagen II levels. Our findings suggest that endoglin haploinsufficiency in mice ameliorates bleomycin-induced skin fibrosis suggesting that endoglin represents a pro-fibrotic factor in the mouse skin. However, endoglin haploinsufficiency does not protect these mice from surgically indiced cartilage degradation, demonstrating differential regulation of endoglin action during skin and cartilage repair.
Collapse
|
153
|
Redgrave RE, Tual-Chalot S, Davison BJ, Singh E, Hall D, Amirrasouli MM, Gilchrist D, Medvinsky A, Arthur HM. Cardiosphere-Derived Cells Require Endoglin for Paracrine-Mediated Angiogenesis. Stem Cell Reports 2018; 8:1287-1298. [PMID: 28494939 PMCID: PMC5425789 DOI: 10.1016/j.stemcr.2017.04.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/12/2017] [Accepted: 04/13/2017] [Indexed: 12/12/2022] Open
Abstract
Clinical trials of stem cell therapy to treat ischemic heart disease primarily use heterogeneous stem cell populations. Small benefits occur via paracrine mechanisms that include stimulating angiogenesis, and increased understanding of these mechanisms would help to improve patient outcomes. Cardiosphere-derived-cells (CDCs) are an example of these heterogeneous stem cell populations, cultured from cardiac tissue. CDCs express endoglin, a co-receptor that binds specific transforming growth factor β (TGFβ) family ligands, including bone morphogenetic protein 9 (BMP9). In endothelial cells endoglin regulates angiogenic responses, and we therefore hypothesized that endoglin is required to promote the paracrine pro-angiogenic properties of CDCs. Cre/LoxP technology was used to genetically manipulate endoglin expression in CDCs, and we found that the pro-angiogenic properties of the CDC secretome are endoglin dependent both in vitro and in vivo. Importantly, BMP9 pre-treatment of endoglin-depleted CDCs restores their pro-angiogenic paracrine properties. As BMP9 signaling is normally required to maintain endoglin expression, we propose that media containing BMP9 could be critical for therapeutic CDC preparation. It is essential to understand how stem cell populations generate paracrine benefit Endoglin is necessary for the pro-angiogenic properties of the CDC secretome Pro-angiogenic defects of endoglin-depleted CDCs can be rescued by BMP9
Collapse
Affiliation(s)
- Rachael E Redgrave
- Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle NE1 3BZ, UK
| | - Simon Tual-Chalot
- Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle NE1 3BZ, UK
| | - Benjamin J Davison
- Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle NE1 3BZ, UK
| | - Esha Singh
- Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle NE1 3BZ, UK
| | - Darroch Hall
- Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle NE1 3BZ, UK
| | - Muhammad M Amirrasouli
- Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle NE1 3BZ, UK
| | - Derek Gilchrist
- Institute for Stem Cell Research, MRC Centre for Regenerative Medicine, Scottish Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Alexander Medvinsky
- Institute for Stem Cell Research, MRC Centre for Regenerative Medicine, Scottish Centre for Regenerative Medicine, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Helen M Arthur
- Institute of Genetic Medicine, Centre for Life, Newcastle University, Newcastle NE1 3BZ, UK.
| |
Collapse
|
154
|
Yu X, Chen X, Zheng XD, Zhang J, Zhao X, Liu Y, Zhang H, Zhang L, Yu H, Zhang M, Ma C, Hao X, Zhu D. Growth Differentiation Factor 11 Promotes Abnormal Proliferation and Angiogenesis of Pulmonary Artery Endothelial Cells. Hypertension 2018; 71:729-741. [PMID: 29463625 DOI: 10.1161/hypertensionaha.117.10350] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 10/09/2017] [Accepted: 01/24/2018] [Indexed: 12/12/2022]
Abstract
Disordered proliferation and angiogenesis of pulmonary artery endothelial cells is an important stage in the development of pulmonary arterial hypertension. Recent studies revealed that GDF11 (growth differentiation factor 11) induces endothelial cells proliferation and migration; however, whether GDF11 is directly involved in the pathogenesis of pulmonary arterial hypertension remains unknown. Here, we found that GDF11 was significantly upregulated and activated in 2 experimental pulmonary arterial hypertension models and cultured pulmonary artery endothelial cells. Genetic ablation of gdf11 in endothelial cells rescued pulmonary arterial hypertension features, as demonstrated by right ventricle hypertrophy, right ventricular systolic pressure, hemodynamics, cardiac function, and vascular remodeling. Moreover, we found that hypoxia significantly increased cell cycle progression, proliferation, migration, adhesion, and tube formation, which were significantly inhibited by GDF11 small interfering RNA. These events could be reproduced using cultured pulmonary artery endothelial cells and were dependent on Smad signaling. Moreover, hypoxia-induced GDF11 expression was regulated by the transcription factor zinc finger protein 740, which assisted RNA polymerase in recognizing and binding to the GDF11 promoter sequence located at a site (-753/-744; CCCCCCCCAC) upstream of the gene. This study identified a novel growth and differentiation factor signaling pathway involved in the zinc finger protein 740/GDF11/transforming growth factor-β receptor I/Smad signaling axis and involved in pulmonary artery endothelial cells proliferation and angiogenesis. These results provide critical insights for the development of novel therapeutic strategies for pulmonary arterial hypertension involving components of the GDF11 signaling system.
Collapse
Affiliation(s)
- Xiufeng Yu
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Xinxin Chen
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Xiao Dong Zheng
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Junting Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Xijuan Zhao
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Ying Liu
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Hongyue Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Lixin Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Hao Yu
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Min Zhang
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Cui Ma
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Xuewei Hao
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.)
| | - Daling Zhu
- From the College of Medical Laboratory Science and Technology (X.Y., X.Z., L.Z., C.M., X.H.) and Department of Pharmacology (X.D.Z., H.Y.), Harbin Medical University (Daqing), P.R. China; Central Laboratory of Harbin Medical University (Daqing), P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.); and Department of Biopharmaceutical Sciences, College of Pharmacy, Harbin Medical University, P.R. China (X.Y., X.C., X.D.Z., J.Z., X.Z., Y.L., H.Z., L.Z., H.Y., M.Z., C.M., X.H., D.Z.).
| |
Collapse
|
155
|
Vascular deficiency of Smad4 causes arteriovenous malformations: a mouse model of Hereditary Hemorrhagic Telangiectasia. Angiogenesis 2018; 21:363-380. [PMID: 29460088 PMCID: PMC5878194 DOI: 10.1007/s10456-018-9602-0] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/28/2018] [Indexed: 12/18/2022]
Abstract
Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant vascular disorder that leads to abnormal connections between arteries and veins termed arteriovenous malformations (AVM). Mutations in TGFβ pathway members ALK1, ENG and SMAD4 lead to HHT. However, a Smad4 mouse model of HHT does not currently exist. We aimed to create and characterize a Smad4 endothelial cell (EC)-specific, inducible knockout mouse (Smad4f/f;Cdh5-CreERT2) that could be used to study AVM development in HHT. We found that postnatal ablation of Smad4 caused various vascular defects, including the formation of distinct AVMs in the neonate retina. Our analyses demonstrated that increased EC proliferation and size, altered mural cell coverage and distorted artery-vein gene expression are associated with Smad4 deficiency in the vasculature. Furthermore, we show that depletion of Smad4 leads to decreased Vegfr2 expression, and concurrent loss of endothelial Smad4 and Vegfr2 in vivo leads to AVM enlargement. Our work provides a new model in which to study HHT-associated phenotypes and links the TGFβ and VEGF signaling pathways in AVM pathogenesis.
Collapse
|
156
|
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: 251] [Impact Index Per Article: 35.9] [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
|
157
|
Goumans MJ, Zwijsen A, Ten Dijke P, Bailly S. Bone Morphogenetic Proteins in Vascular Homeostasis and Disease. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a031989. [PMID: 28348038 DOI: 10.1101/cshperspect.a031989] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is well established that control of vascular morphogenesis and homeostasis is regulated by vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), Delta-like 4 (Dll4), angiopoietin, and ephrin signaling. It has become clear that signaling by bone morphogenetic proteins (BMPs), which have a long history of studies in bone and early heart development, are also essential for regulating vascular function. Indeed, mutations that cause deregulated BMP signaling are linked to two human vascular diseases, hereditary hemorrhagic telangiectasia and pulmonary arterial hypertension. These observations are corroborated by data obtained with vascular cells in cell culture and in mouse models. BMPs are required for normal endothelial cell differentiation and for venous/arterial and lymphatic specification. In adult life, BMP signaling orchestrates neo-angiogenesis as well as vascular inflammation, remodeling, and calcification responses to shear and oxidative stress. This review emphasizes the pivotal role of BMPs in the vascular system, based on studies of mouse models and human vascular disorders.
Collapse
Affiliation(s)
- Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - An Zwijsen
- VIB Center for the Biology of Disease, 3000 Leuven, Belgium.,KU Leuven Department of Human Genetics, 3000 Leuven, Belgium
| | - Peter Ten Dijke
- Department of Molecular Cell Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,Cancer Genomics Centre Netherlands, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sabine Bailly
- Institut National de la Santé et de la Recherche Mécale (INSERM), U1036, 38000 Grenoble, France.,Laboratoire Biologie du Cancer et de l'Infection, Commissariat à l'Énergie Atomique et aux Energies Alternatives, Biosciences and Biotechnology Institute of Grenoble, 38000 Grenoble, France.,University of Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
158
|
Tian H, Huang JJ, Golzio C, Gao X, Hector-Greene M, Katsanis N, Blobe GC. Endoglin interacts with VEGFR2 to promote angiogenesis. FASEB J 2018; 32:2934-2949. [PMID: 29401587 DOI: 10.1096/fj.201700867rr] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Endoglin, a TGF-β coreceptor predominantly expressed in endothelial cells, plays an important role in vascular development and tumor-associated angiogenesis. However, the mechanism by which endoglin regulates angiogenesis, especially during tip cell formation, remains largely unknown. In this study, we report that endoglin promoted VEGF-induced tip cell formation. Mechanistically, endoglin interacted with VEGF receptor (VEGFR)-2 in a VEGF-dependent manner, which sustained VEGFR2 on the cell surface and prevented its degradation. Endoglin mutants deficient in the ability to interact with VEGFR2 failed to sustain VEGFR2 on the cell surface and to promote VEGF-induced tip cell formation. Further, an endoglin-targeting monoclonal antibody (mAb), TRC105, cooperated with a VEGF-A targeting mAb, bevacizumab, to inhibit VEGF signaling and tip cell formation in vitro and to inhibit tumor growth, metastasis, and tumor-associated angiogenesis in a murine tumor model. This study demonstrate a novel mechanism by which endoglin initiates and regulates VEGF-driven angiogenesis while providing a rationale for combining anti-VEGF and anti-endoglin therapy in patients with cancer.-Tian, H., Huang, J. J., Golzio, C., Gao, X., Hector-Greene, M., Katsanis, N., Blobe, G. C. Endoglin interacts with VEGFR2 to promote angiogenesis.
Collapse
Affiliation(s)
- Hongyu Tian
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Jennifer J Huang
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Christelle Golzio
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina, USA
| | - Xia Gao
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Melissa Hector-Greene
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| | - Nicholas Katsanis
- Center for Human Disease Modeling, Duke University Medical Center, Durham, North Carolina, USA
| | - Gerard C Blobe
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA.,Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
159
|
Nickel J, Ten Dijke P, Mueller TD. TGF-β family co-receptor function and signaling. Acta Biochim Biophys Sin (Shanghai) 2018; 50:12-36. [PMID: 29293886 DOI: 10.1093/abbs/gmx126] [Citation(s) in RCA: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 11/08/2017] [Indexed: 01/04/2023] Open
Abstract
Transforming growth factor-β (TGF-β) family members, which include TGF-βs, activins and bone morphogenetic proteins, are pleiotropic cytokines that elicit cell type-specific effects in a highly context-dependent manner in many different tissues. These secreted protein ligands signal via single-transmembrane Type I and Type II serine/threonine kinase receptors and intracellular SMAD transcription factors. Deregulation in signaling has been implicated in a broad array of diseases, and implicate the need for intricate fine tuning in cellular signaling responses. One important emerging mechanism by which TGF-β family receptor signaling intensity, duration, specificity and diversity are regulated and/or mediated is through cell surface co-receptors. Here, we provide an overview of the co-receptors that have been identified for TGF-β family members. While some appear to be specific to TGF-β family members, others are shared with other pathways and provide possible ways for signal integration. This review focuses on novel functions of TGF-β family co-receptors, which continue to be discovered.
Collapse
Affiliation(s)
- Joachim Nickel
- Universitätsklinikum Würzburg, Lehrstuhl für Tissue Engineering und Regenerative Medizin und Fraunhofer Institut für Silicatforschung (ISC), Translationszentrum "Regenerative Therapien", Röntgenring 11, D-97070 Würzburg, Germany
| | - Peter Ten Dijke
- Department of Molecular and Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, The Netherlands
| | - Thomas D Mueller
- Lehrstuhl für molekulare Pflanzenphysiologie und Biophysik, Julius-von-Sachs Institut für Biowissenschaften, Universität Würzburg, Julius-von-Sachs-Platz 2, D-97082 Würzburg, Germany
| |
Collapse
|
160
|
Dingenouts CKE, Bakker W, Lodder K, Wiesmeijer KC, Moerkamp AT, Maring JA, Arthur HM, Smits AM, Goumans MJ. Inhibiting DPP4 in a mouse model of HHT1 results in a shift towards regenerative macrophages and reduces fibrosis after myocardial infarction. PLoS One 2017; 12:e0189805. [PMID: 29253907 PMCID: PMC5734765 DOI: 10.1371/journal.pone.0189805] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 12/02/2017] [Indexed: 12/11/2022] Open
Abstract
AIMS Hereditary Hemorrhagic Telangiectasia type-1 (HHT1) is a genetic vascular disorder caused by haploinsufficiency of the TGFβ co-receptor endoglin. Dysfunctional homing of HHT1 mononuclear cells (MNCs) towards the infarcted myocardium hampers cardiac recovery. HHT1-MNCs have elevated expression of dipeptidyl peptidase-4 (DPP4/CD26), which inhibits recruitment of CXCR4-expressing MNCs by inactivation of stromal cell-derived factor 1 (SDF1). We hypothesize that inhibiting DPP4 will restore homing of HHT1-MNCs to the infarcted heart and improve cardiac recovery. METHODS AND RESULTS After inducing myocardial infarction (MI), wild type (WT) and endoglin heterozygous (Eng+/-) mice were treated for 5 days with the DPP4 inhibitor Diprotin A (DipA). DipA increased the number of CXCR4+ MNCs residing in the infarcted Eng+/- hearts (Eng+/- 73.17±12.67 vs. Eng+/- treated 157.00±11.61, P = 0.0003) and significantly reduced infarct size (Eng+/- 46.60±9.33% vs. Eng+/- treated 27.02±3.04%, P = 0.03). Echocardiography demonstrated that DipA treatment slightly deteriorated heart function in Eng+/- mice. An increased number of capillaries (Eng+/- 61.63±1.43 vs. Eng+/- treated 74.30±1.74, P = 0.001) were detected in the infarct border zone whereas the number of arteries was reduced (Eng+/- 11.88±0.63 vs. Eng+/- treated 6.38±0.97, P = 0.003). Interestingly, while less M2 regenerative macrophages were present in Eng+/- hearts prior to DipA treatment, (WT 29.88±1.52% vs. Eng+/- 12.34±1.64%, P<0.0001), DPP4 inhibition restored the number of M2 macrophages to wild type levels. CONCLUSIONS In this study, we demonstrate that systemic DPP4 inhibition restores the impaired MNC homing in Eng+/- animals post-MI, and enhances cardiac repair, which might be explained by restoring the balance between the inflammatory and regenerative macrophages present in the heart.
Collapse
Affiliation(s)
| | - Wineke Bakker
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Kirsten Lodder
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Karien C. Wiesmeijer
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Asja T. Moerkamp
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Janita A. Maring
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Helen M. Arthur
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Newcastle upon Tyne, United Kingdom
| | - Anke M. Smits
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
161
|
Lempereur A, Canto PY, Richard C, Martin S, Thalgott J, Raymond K, Lebrin F, Drevon C, Jaffredo T. The TGFβ pathway is a key player for the endothelial-to-hematopoietic transition in the embryonic aorta. Dev Biol 2017; 434:292-303. [PMID: 29253505 DOI: 10.1016/j.ydbio.2017.12.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 12/07/2017] [Accepted: 12/08/2017] [Indexed: 12/30/2022]
Abstract
The embryonic aorta produces hematopoietic stem and progenitor cells from a hemogenic endothelium localized in the aortic floor through an endothelial to hematopoietic transition. It has been long proposed that the Bone Morphogenetic Protein (BMP)/Transforming Growth Factor ß (TGFß) signaling pathway was implicated in aortic hematopoiesis but the very nature of the signal was unknown. Here, using thorough expression analysis of the BMP/TGFß signaling pathway members in the endothelial and hematopoietic compartments of the aorta at pre-hematopoietic and hematopoietic stages, we show that the TGFß pathway is preferentially balanced with a prominent role of Alk1/TgfßR2/Smad1 and 5 on both chicken and mouse species. Functional analysis using embryonic stem cells mutated for Acvrl1 revealed an enhanced propensity to produce hematopoietic cells. Collectively, we reveal that TGFß through the Alk1/TgfßR2 receptor axis is acting on endothelial cells to produce hematopoiesis.
Collapse
Affiliation(s)
- A Lempereur
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - P Y Canto
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - C Richard
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - S Martin
- CNRS UMR 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris CEDEX 05, France; MEMOLIFE Laboratory of Excellence and Paris Sciences et Lettres Research University, France
| | - J Thalgott
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - K Raymond
- Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands
| | - F Lebrin
- CNRS UMR 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, 11 Place Marcelin Berthelot, 75231 Paris CEDEX 05, France; Einthoven Laboratory for Experimental Vascular Medicine, Department of Internal Medicine (Nephrology), Leiden University Medical Center, Leiden, The Netherlands; MEMOLIFE Laboratory of Excellence and Paris Sciences et Lettres Research University, France
| | - C Drevon
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France
| | - T Jaffredo
- Sorbonne Universités, UPMC Univ Paris 06, IBPS, CNRS UMR7622, Inserm U 1156, Laboratoire de Biologie du Développement, 75005 Paris, France.
| |
Collapse
|
162
|
van de Pol V, Kurakula K, DeRuiter MC, Goumans MJ. Thoracic Aortic Aneurysm Development in Patients with Bicuspid Aortic Valve: What Is the Role of Endothelial Cells? Front Physiol 2017; 8:938. [PMID: 29249976 PMCID: PMC5714935 DOI: 10.3389/fphys.2017.00938] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/06/2017] [Indexed: 12/28/2022] Open
Abstract
Bicuspid aortic valve (BAV) is the most common type of congenital cardiac malformation. Patients with a BAV have a predisposition for the development of thoracic aortic aneurysm (TAA). This pathological aortic dilation may result in aortic rupture, which is fatal in most cases. The abnormal aortic morphology of TAAs results from a complex series of events that alter the cellular structure and extracellular matrix (ECM) composition of the aortic wall. Because the major degeneration is located in the media of the aorta, most studies aim to unravel impaired smooth muscle cell (SMC) function in BAV TAA. However, recent studies suggest that endothelial cells play a key role in both the initiation and progression of TAAs by influencing the medial layer. Aortic endothelial cells are activated in BAV mediated TAAs and have a substantial influence on ECM composition and SMC phenotype, by secreting several key growth factors and matrix modulating enzymes. In recent years there have been significant advances in the genetic and molecular understanding of endothelial cells in BAV associated TAAs. In this review, the involvement of the endothelial cells in BAV TAA pathogenesis is discussed. Endothelial cell functioning in vessel homeostasis, flow response and signaling will be highlighted to give an overview of the importance and the under investigated potential of endothelial cells in BAV-associated TAA.
Collapse
Affiliation(s)
- Vera van de Pol
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Kondababu Kurakula
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Marco C. DeRuiter
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
163
|
Hisamatsu D, Ohno-Oishi M, Nakamura S, Mabuchi Y, Naka-Kaneda H. Growth differentiation factor 6 derived from mesenchymal stem/stromal cells reduces age-related functional deterioration in multiple tissues. Aging (Albany NY) 2017; 8:1259-75. [PMID: 27311402 PMCID: PMC4931831 DOI: 10.18632/aging.100982] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 06/06/2016] [Indexed: 12/18/2022]
Abstract
The senescence-associated secretory phenotype (SASP) has attracted attention as a mechanism that connects cellular senescence to tissue dysfunction, and specific SASP factors have been identified as systemic pro-aging factors. However, little is known about the age-dependent changes in the secretory properties of stem cells. Young, but not old, mesenchymal stem/stromal cells (MSCs) are a well-known source of critical regenerative factors, but the identity of these factors remains elusive. In this study, we identified growth differentiation factor 6 (Gdf6; also known as Bmp13 and CDMP-2) as a regenerative factor secreted from young MSCs. The expression of specific secretory factors, including Gdf6, was regulated by the microRNA (miRNA) miR-17, whose expression declined with age. Upregulation of Gdf6 restored the osteogenic capacity of old MSCs in vitro and exerted positive effects in vivo on aging-associated pathologies such as reduced lymphopoiesis, insufficient muscle repair, reduced numbers of neural progenitors in the brain, and chronic inflammation. Our results suggest that manipulation of miRNA could enable control of the SASP, and that regenerative factors derived from certain types of young cells could be used to treat geriatric diseases.
Collapse
Affiliation(s)
- Daisuke Hisamatsu
- Laboratory for Stem Cell Competency, RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.,Department of Physiology, Keio University School of Medicine, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Michiko Ohno-Oishi
- Laboratory for Stem Cell Competency, RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Shiho Nakamura
- Laboratory for Stem Cell Competency, RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Yo Mabuchi
- Department of Biochemistry and Biophysics, Graduate School of Health Care Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Hayato Naka-Kaneda
- Laboratory for Stem Cell Competency, RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| |
Collapse
|
164
|
Hepatitis C Virus Core Protein Modulates Endoglin (CD105) Signaling Pathway for Liver Pathogenesis. J Virol 2017; 91:JVI.01235-17. [PMID: 28794048 DOI: 10.1128/jvi.01235-17] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/06/2017] [Indexed: 12/11/2022] Open
Abstract
Endoglin is part of the TGF-β receptor complex and has a crucial role in fibrogenesis and angiogenesis. It is also an important protein for tumor growth, survival, and cancer cell metastasis. In a previous study, we have shown that hepatitis C virus (HCV) infection induces epithelial-mesenchymal transition (EMT) state and cancer stem-like cell (CSC) properties in human hepatocytes. Our array data suggested that endoglin (CD105) mRNA is significantly upregulated in HCV-associated CSCs. In this study, we have observed increased endoglin expression on the cell surface of an HCV core-expressing hepatocellular carcinoma (HepG2) cell line or immortalized human hepatocytes (IHH) and activation of its downstream signaling molecules. The status of phospho-SMAD1/5 and the expression of inhibitor of DNA binding protein 1 (ID1) were upregulated in HCV-infected cells or viral core gene-transfected cells. Additionally, we observed upregulation of endoglin/ID1 mRNA expression in chronic HCV patient liver biopsy samples. CSC generation by HCV core protein was dependent on the endoglin signaling pathway using activin receptor-like kinase 1 (ALK1) Fc blocking peptide and endoglin small interfering RNA (siRNA). Further, follow-up from in vitro analysis suggested that the antiapoptosis Bcl2 protein, proliferation-related cyclin D1 protein, and CSC-associated Hes1, Notch1, Nanog, and Sox2 proteins are enhanced during infection or ectopic expression of HCV core protein.IMPORTANCE Endoglin plays a crucial role in fibrogenesis and angiogenesis and is an important protein for tumor growth, survival, and cancer cell metastasis. Endoglin enhances ALK1-SMAD1/5 signaling in different cell types, leading to increased proliferation and migration responses. We have observed endoglin expression on the HCV core-expressing cell surface of human hepatocyte origin and activation of phospho-SMAD1/5 and ID1 downstream signaling molecules. ID1 protein plays a role in CSC properties, and we found that this pathway is important for antiapoptotic and cell proliferation signaling. Blocking of endoglin-ALK1-SMAD1/5 might be a good candidate for therapy for liver cancer stem cells together with liver cirrhosis.
Collapse
|
165
|
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
|
166
|
Deschênes M, Chabot B. The emerging role of alternative splicing in senescence and aging. Aging Cell 2017; 16:918-933. [PMID: 28703423 PMCID: PMC5595669 DOI: 10.1111/acel.12646] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2017] [Indexed: 12/22/2022] Open
Abstract
Deregulation of precursor mRNA splicing is associated with many illnesses and has been linked to age-related chronic diseases. Here we review recent progress documenting how defects in the machinery that performs intron removal and controls splice site selection contribute to cellular senescence and organismal aging. We discuss the functional association linking p53, IGF-1, SIRT1, and ING-1 splice variants with senescence and aging, and review a selection of splicing defects occurring in accelerated aging (progeria), vascular aging, and Alzheimer's disease. Overall, it is becoming increasingly clear that changes in the activity of splicing factors and in the production of key splice variants can impact cellular senescence and the aging phenotype.
Collapse
Affiliation(s)
- Mathieu Deschênes
- Department of Microbiology and Infectious DiseasesFaculty of Medicine and Health SciencesUniversité de SherbrookeSherbrookeQuebecJ1E 4K8Canada
| | - Benoit Chabot
- Department of Microbiology and Infectious DiseasesFaculty of Medicine and Health SciencesUniversité de SherbrookeSherbrookeQuebecJ1E 4K8Canada
| |
Collapse
|
167
|
de Jager SCA, Meeuwsen JAL, van Pijpen FM, Zoet GA, Barendrecht AD, Franx A, Pasterkamp G, van Rijn BB, Goumans MJ, den Ruijter HM. Preeclampsia and coronary plaque erosion: Manifestations of endothelial dysfunction resulting in cardiovascular events in women. Eur J Pharmacol 2017; 816:129-137. [PMID: 28899695 DOI: 10.1016/j.ejphar.2017.09.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/31/2017] [Accepted: 09/08/2017] [Indexed: 12/29/2022]
Abstract
Atherosclerosis is the major underlying pathology of cardiovascular disease (CVD). The risk for CVD is increased in women with a history of preeclampsia. Multiple studies have indicated that accelerated atherosclerosis underlies this increased CVD risk. Furthermore, it has been suggested that endothelial dysfunction and inflammation play an important role in the increased CVD risk of women with preeclampsia. Rupture or erosion of atherosclerotic plaques can induce the formation of thrombi that underlie the onset of acute clinical CVD such as myocardial infarction and stroke. In relatively young women, cardiovascular events are mainly due to plaque erosions. Eroded plaques have a distinct morphology compared to ruptured plaques, but have been understudied as a substrate for CVD. The currently available evidence points towards lesions with features of stability such as high collagen content and smooth muscle cells and with distinct mechanisms that further promote the pro-thrombotic environment such as Toll Like Receptor (TLR) signaling and endothelial apoptosis. These suggested mechanisms, that point to endothelial dysfunction and intimal thickening, may also play a role in preeclampsia. Pregnancy is considered a stress test for the cardiovascular system with preeclampsia as an additional pathological substrate for earlier manifestation of vascular disease. This review provides a summary of the possible common mechanisms involved in preeclampsia and accelerated atherosclerosis in young females and highlights plaque erosion as a likely substrate for CVD events in women with a history of preeclampsia.
Collapse
Affiliation(s)
- Saskia C A de Jager
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, The Netherlands.
| | - John A L Meeuwsen
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, The Netherlands
| | - Freeke M van Pijpen
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, The Netherlands
| | - Gerbrand A Zoet
- Wilhelmina Children's Hospital Birth Centre, Division of Woman and Baby, University Medical Center Utrecht, The Netherlands
| | - Arjan D Barendrecht
- Laboratory of Clinical Chemistry and Haematology, University Medical Center Utrecht, The Netherlands
| | - Arie Franx
- Wilhelmina Children's Hospital Birth Centre, Division of Woman and Baby, University Medical Center Utrecht, The Netherlands
| | - Gerard Pasterkamp
- Laboratory of Clinical Chemistry and Haematology, University Medical Center Utrecht, The Netherlands
| | - Bas B van Rijn
- Wilhelmina Children's Hospital Birth Centre, Division of Woman and Baby, University Medical Center Utrecht, The Netherlands; Academic Unit of Human Development and Health, Institute for Life Sciences, University of Southampton, United Kingdom
| | - Marie-José Goumans
- Department of Molecular Cell Biology, Leiden University Medical Center, The Netherlands
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, University Medical Center Utrecht, The Netherlands
| |
Collapse
|
168
|
Jin Y, Muhl L, Burmakin M, Wang Y, Duchez AC, Betsholtz C, Arthur HM, Jakobsson L. Endoglin prevents vascular malformation by regulating flow-induced cell migration and specification through VEGFR2 signalling. Nat Cell Biol 2017; 19:639-652. [PMID: 28530660 PMCID: PMC5467724 DOI: 10.1038/ncb3534] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 04/18/2017] [Indexed: 12/15/2022]
Abstract
Loss-of-function (LOF) mutations in the endothelial cell (EC) enriched gene endoglin (ENG) causes the human disease hereditary haemorrhagic telangiectasia-1, characterized by vascular malformations promoted by vascular endothelial growth factor A (VEGFA). How ENG deficiency alters EC behaviour to trigger these anomalies is not understood. Mosaic ENG deletion in the postnatal mouse rendered Eng LOF ECs insensitive to flow-mediated venous to arterial migration. Eng LOF ECs retained within arterioles acquired venous characteristics and secondary ENG-independent proliferation resulting in arterio-venous malformation (AVM). Analysis following simultaneous Eng LOF and overexpression (OE) revealed that ENG OE ECs dominate tip cell positions and home preferentially to arteries. ENG knock-down altered VEGFA-mediated VEGFR2 kinetics and promoted AKT signalling. Blockage of PI3K/AKT partly normalised flow-directed migration of ENG LOF ECs in vitro and reduced the severity of AVM in vivo. This demonstrates the requirement of ENG in flow-mediated migration and modulation of VEGFR2 signalling in vascular patterning.
Collapse
Affiliation(s)
- Yi Jin
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77 Stockholm, Sweden
| | - Lars Muhl
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77 Stockholm, Sweden
| | - Mikhail Burmakin
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77 Stockholm, Sweden
| | - Yixin Wang
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77 Stockholm, Sweden
| | - Anne-Claire Duchez
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77 Stockholm, Sweden
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Uppsala University, Dag Hammarskjölds väg 20, 751 85 Uppsala, Sweden.,Integrated Cardio Metabolic Centre (ICMC), Karolinska Institutet, Novum, Blickagången 6, SE14157 Huddinge, Sweden
| | - Helen M Arthur
- Institute of Genetic Medicine, International Centre for Life, Newcastle University, Newcastle upon Tyne NE1 3BZ, UK
| | - Lars Jakobsson
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Scheeles väg 2, 171 77 Stockholm, Sweden
| |
Collapse
|
169
|
Siqueira M, Francis D, Gisbert D, Gomes FCA, Stipursky J. Radial Glia Cells Control Angiogenesis in the Developing Cerebral Cortex Through TGF-β1 Signaling. Mol Neurobiol 2017; 55:3660-3675. [PMID: 28523566 DOI: 10.1007/s12035-017-0557-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 04/12/2017] [Indexed: 10/19/2022]
Abstract
Neuroangiogenesis in the developing central nervous system is controlled by interactions between endothelial cells (ECs) and radial glia (RG) neural stem cells, although RG-derived molecules implicated in these events are not fully known. Here, we investigated the role of RG-secreted TGF-β1, in angiogenesis in the developing cerebral cortex. By isolation of murine microcapillary brain endothelial cells (MBECs), we demonstrate that conditioned medium from RG cultures (RG-CM) promoted MBEC migration and formation of vessel-like structures in vitro, in a TGF-β1-dependent manner. These events were followed by endothelial regulation of GPR124 and BAI-1 gene expression by RG-CM. Proteome profile of RG-CM identified angiogenesis-related molecules IGFBP2/3, osteopontin, endostatin, SDF1, fractalkine, TIMP1/4, Ang-1, pentraxin3, and Cyr61, some of them modulated by TGF-β1 induction. In vivo gain and loss of function assays targeting RG cells demonstrates a specific TGF-β1-dependent control of blood vessels branching in the cerebral cortex. Together, our results point to TGF-β1 signaling pathway as a potential mediator of the RG-EC interactions and shed light to the key role of RG in paving the brain vascular network.
Collapse
Affiliation(s)
- Michele Siqueira
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Daniel Francis
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Diego Gisbert
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Joice Stipursky
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil. .,Laboratório de Neurobiologia Celular, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro - Centro de Ciências da Saúde, Bloco F, Sala F15, Ilha do Fundão, Rio de Janeiro, RJ, 21949-902, Brazil.
| |
Collapse
|
170
|
Tian H, Ketova T, Hardy D, Xu X, Gao X, Zijlstra A, Blobe GC. Endoglin Mediates Vascular Maturation by Promoting Vascular Smooth Muscle Cell Migration and Spreading. Arterioscler Thromb Vasc Biol 2017; 37:1115-1126. [PMID: 28450296 PMCID: PMC5444426 DOI: 10.1161/atvbaha.116.308859] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 04/19/2017] [Indexed: 11/16/2022]
Abstract
Supplemental Digital Content is available in the text. Objective— Endoglin, a transforming growth factor-β superfamily coreceptor, is predominantly expressed in endothelial cells and has essential roles in vascular development. However, whether endoglin is also expressed in vascular smooth muscle cells (VSMCs), especially in vivo, remains controversial. Furthermore, the roles of endoglin in VSMC biology remain largely unknown. Our objective was to examine the expression and determine the function of endoglin in VSMCs during angiogenesis. Approach and Results— Here, we determine that endoglin is robustly expressed in VSMCs. Using CRISPR/CAS9 knockout and short hairpin RNA knockdown in the VSMC/endothelial coculture model system, we determine that endoglin in VSMCs, but not in endothelial cells, promotes VSMCs recruitment by the endothelial cells both in vitro and in vivo. Using an unbiased bioinformatics analysis of RNA sequencing data and further study, we determine that, mechanistically, endoglin mediates VSMC recruitment by promoting VSMC migration and spreading on endothelial cells via increasing integrin/FAK pathway signaling, whereas endoglin has minimal effects on VSMC adhesion to endothelial cells. In addition, we further determine that loss of endoglin in VSMCs inhibits VSMC recruitment in vivo. Conclusions— These studies demonstrate that endoglin has an important role in VSMC recruitment and blood vessel maturation during angiogenesis and also provide novel insights into how discordant endoglin function in endothelial and VSMCs may regulate vascular maturation and angiogenesis.
Collapse
Affiliation(s)
- Hongyu Tian
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.);
| | - Tatiana Ketova
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.)
| | - Duriel Hardy
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.)
| | - Xiaojiang Xu
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.)
| | - Xia Gao
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.)
| | - Andries Zijlstra
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.)
| | - Gerard C Blobe
- From the Division of Medical Oncology, Department of Medicine (H.T., D.H., G.C.B.) and Department of Pharmacology and Cancer Biology (G.C.B.), Duke University Medical Center, Durham, NC; Department of Pathology, Microbiology, and Immunology (T.K., A.Z.) and Department of Cancer Biology (A.Z.), Vanderbilt University, Nashville, TN; Integrative Bioinformatics, National Institute of Environmental Health Sciences, Research Triangle Park, NC (X.X.); Department of Cell Biology, Duke University School of Medicine, Durham, NC (X.G.); and Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN (A.Z.);
| |
Collapse
|
171
|
Wagner MJ, Ravi V, Menter DG, Sood AK. Endothelial cell malignancies: new insights from the laboratory and clinic. NPJ Precis Oncol 2017; 1:11. [PMID: 29872699 PMCID: PMC5859470 DOI: 10.1038/s41698-017-0013-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/13/2017] [Indexed: 12/14/2022] Open
Abstract
Endothelial cell malignancies are rare in the Western world and range from intermediate grade hemangioendothelioma to Kaposi sarcoma to aggressive high-grade angiosarcoma that metastasize early and have a high rate of mortality. These malignancies are associated with dysregulation of normal endothelial cell signaling pathways, including the vascular endothelial growth factor, angiopoietin, and Notch pathways. Discoveries over the past two decades related to mechanisms of angiogenesis have led to the development of many drugs that intuitively would be promising therapeutic candidates for these endothelial-derived tumors. However, clinical efficacy of such drugs has been limited. New insights into the mechanisms that lead to dysregulated angiogenesis such as mutation or amplification in known angiogenesis related genes, viral infection, and chromosomal translocations have improved our understanding of the pathogenesis of endothelial malignancies and how they evade anti-angiogenesis drugs. In this review, we describe the major molecular alterations in endothelial cell malignancies and consider emerging opportunities for improving therapeutic efficacy against these rare but deadly tumors.
Collapse
Affiliation(s)
- Michael J Wagner
- 1Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Vinod Ravi
- 2Department of Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - David G Menter
- 3Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Anil K Sood
- 4Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA.,5Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA.,6Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| |
Collapse
|
172
|
TGF-β Family Signaling in the Control of Cell Proliferation and Survival. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022145. [PMID: 27920038 DOI: 10.1101/cshperspect.a022145] [Citation(s) in RCA: 451] [Impact Index Per Article: 56.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The transforming growth factor β (TGF-β) family controls many fundamental aspects of cellular behavior. With advances in the molecular details of the TGF-β signaling cascade and its cross talk with other signaling pathways, we now have a more coherent understanding of the cytostatic program induced by TGF-β. However, the molecular mechanisms are still largely elusive for other cellular processes that are regulated by TGF-β and determine a cell's proliferation and survival, apoptosis, dormancy, autophagy, and senescence. The difficulty in defining TGF-β's roles partly stems from the context-dependent nature of TGF-β signaling. Here, we review our current understanding and recent progress on the biological effects of TGF-β at the cellular level, with the hope of providing a framework for understanding how cells respond to TGF-β signals in specific contexts, and why disruption of such mechanisms may result in different human diseases including cancer.
Collapse
|
173
|
Endoglin: a novel target for therapeutic intervention in acute leukemias revealed in xenograft mouse models. Blood 2017; 129:2526-2536. [PMID: 28351936 DOI: 10.1182/blood-2017-01-763581] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/20/2017] [Indexed: 12/23/2022] Open
Abstract
Endoglin (CD105), a receptor of the transforming growth factor-β superfamily, has been reported to identify functional long-term repopulating hematopoietic stem cells, and has been detected in certain subtypes of acute leukemias. Whether this receptor plays a functional role in leukemogenesis remains unknown. We identified endoglin expression on the majority of blasts from patients with acute myeloid leukemia (AML) and acute B-lymphoblastic leukemia (B-ALL). Using a xenograft model, we find that CD105+ blasts are endowed with superior leukemogenic activity compared with the CD105- population. We test the effect of targeting this receptor using the monoclonal antibody TRC105, and find that in AML, TRC105 prevented the engraftment of primary AML blasts and inhibited leukemia progression following disease establishment, but in B-ALL, TRC105 alone was ineffective due to the shedding of soluble CD105. However, in both B-ALL and AML, TRC105 synergized with reduced intensity myeloablation to inhibit leukemogenesis, indicating that TRC105 may represent a novel therapeutic option for B-ALL and AML.
Collapse
|
174
|
Tania NP, Maarsingh H, T Bos IS, Mattiotti A, Prakash S, Timens W, Gunst QD, Jimenez-Borreguero LJ, Schmidt M, van den Hoff MJB, Gosens R. Endothelial follistatin-like-1 regulates the postnatal development of the pulmonary vasculature by modulating BMP/Smad signaling. Pulm Circ 2017; 7:219-231. [PMID: 28680581 PMCID: PMC5448549 DOI: 10.1177/2045893217702340] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 12/20/2016] [Indexed: 11/17/2022] Open
Abstract
Bone morphogenetic protein (BMP) signaling regulates vascular smooth muscle maturation, endothelial cell proliferation, and tube formation. The endogenous BMP antagonist Follistatin-like 1 (Fstl1) is highly expressed in pulmonary vascular endothelium of the developing mouse lung, suggesting a role in pulmonary vascular formation and vascular homeostasis. The aim of this study was to investigate the role of Fstl1 in the pulmonary vascular endothelium. To this aim, Fstl1 was conditionally deleted from endothelial and endothelial-derived cells using Tie2-cre driven Fstl1-KO mice (Fstl1-eKO mice). Endothelial-specific Fstl1 deletion was postnatally lethal, as ∼70% of Fstl1-eKO mice died at three weeks after birth. Deletion of Fstl1 from endothelium resulted in a reduction of right ventricular output at three weeks after birth compared with controls. This was associated with pulmonary vascular remodeling, as the percentage of actin-positive small pulmonary vessels was increased at three weeks in Fstl1-eKO mice compared with controls. Endothelial deletion of Fstl1 resulted in activation of Smad1/5/8 signaling and increased BMP/Smad-regulated gene expression of Jagged1, Endoglin, and Gata2 at one week after birth compared with controls. In addition, potent vasoconstrictor Endothelin-1, the expression of which is driven by Gata2, was increased in expression, both on the mRNA and protein levels, at one week after birth compared with controls. At three weeks, Jagged1 was reduced in the Fstl1-eKO mice whereas Endoglin and Endothelin-1 were unchanged. In conclusion, loss of endothelial Fstl1 in the lung is associated with elevated BMP-regulated genes, impaired small pulmonary vascular remodeling, and decreased right ventricular output.
Collapse
Affiliation(s)
- Navessa P Tania
- University of Groningen, Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Harm Maarsingh
- Palm Beach Atlantic University, Department of Pharmaceutical Sciences, Lloyd L. Gregory School of Pharmacy, West Palm Beach, FL, USA
| | - I Sophie T Bos
- University of Groningen, Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Andrea Mattiotti
- Academic Medical Center, Department of Anatomy, Embryology and Physiology, Amsterdam, The Netherlands
| | - Stuti Prakash
- Academic Medical Center, Department of Anatomy, Embryology and Physiology, Amsterdam, The Netherlands
| | - Wim Timens
- University of Groningen, University Medical Center Groningen, Department of Pathology and Medical Biology, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Quinn D Gunst
- Academic Medical Center, Department of Anatomy, Embryology and Physiology, Amsterdam, The Netherlands
| | | | - Martina Schmidt
- University of Groningen, Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| | - Maurice J B van den Hoff
- Academic Medical Center, Department of Anatomy, Embryology and Physiology, Amsterdam, The Netherlands
| | - Reinoud Gosens
- University of Groningen, Department of Molecular Pharmacology, Groningen Research Institute for Asthma and COPD, Groningen, The Netherlands
| |
Collapse
|
175
|
Farey JE, Fisher OM, Levert-Mignon AJ, Forner PM, Lord RV. Decreased Levels of Circulating Cancer-Associated Protein Biomarkers Following Bariatric Surgery. Obes Surg 2017; 27:578-585. [PMID: 27525640 PMCID: PMC5306243 DOI: 10.1007/s11695-016-2321-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Epidemiological studies have identified obesity as a major risk factor for cancer in humans, and trials have demonstrated a significant reduction in the incidence of cancer after bariatric surgery. The rapidity of weight loss after bariatric surgery provides an opportunity to identify the molecular changes associated with effective obesity treatment. Indirectly, this may provide some insights into the mechanisms that drive the association between obesity and cancer. We sought to measure circulating cancer-associated proteins before and after laparoscopic sleeve gastrectomy (LSG). METHODS We prospectively enrolled 15 patients undergoing LSG. Thirty-four plasma protein biomarkers thought to be associated with cancer processes were analyzed at baseline and following successful weight loss at 12 weeks using a multiplex bead-based assay. RESULTS Mean excess body weight loss was 44 % at 12-week follow-up. After LSG, a significant reduction in circulating plasma levels was observed for half (17/34) of the proteins assessed: VEGF-A, VEGF-C, VEGF-D, endoglin, PLGF, sFASL, IGFBP-1, IL-18, prolactin, EGF, TGFα, sCD40L, IL-18, TNFα, IL-6, HB-EGF, and PAI-1. Nonsignificant decreases were found for the remaining proteins. CONCLUSIONS Circulating cancer-related biomarker levels were reduced by surgical weight loss, and this benefit was achieved as early as 3 months after operation. The observed reduction in cancer biomarkers may be related to the reported decrease in cancer incidence following bariatric surgery.
Collapse
Affiliation(s)
- John Edward Farey
- Department of Surgery, University of Notre Dame Australia, School of Medicine, Sydney, Darlinghurst, NSW Australia
- Gastro-Oesophageal Cancer Research Program, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW Australia
| | - Oliver M. Fisher
- Department of Surgery, University of Notre Dame Australia, School of Medicine, Sydney, Darlinghurst, NSW Australia
- Gastro-Oesophageal Cancer Research Program, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW Australia
| | - Angelique J. Levert-Mignon
- Department of Surgery, University of Notre Dame Australia, School of Medicine, Sydney, Darlinghurst, NSW Australia
- Gastro-Oesophageal Cancer Research Program, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW Australia
| | - Patrice M. Forner
- Department of Surgery, University of Notre Dame Australia, School of Medicine, Sydney, Darlinghurst, NSW Australia
- Gastro-Oesophageal Cancer Research Program, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW Australia
| | - Reginald V. Lord
- Department of Surgery, University of Notre Dame Australia, School of Medicine, Sydney, Darlinghurst, NSW Australia
- Gastro-Oesophageal Cancer Research Program, St Vincent’s Centre for Applied Medical Research, Darlinghurst, NSW Australia
| |
Collapse
|
176
|
Yassin AM, El-Deeb NM, Metwaly AM, El Fawal GF, Radwan MM, Hafez EE. Induction of Apoptosis in Human Cancer Cells Through Extrinsic and Intrinsic Pathways by Balanites aegyptiaca Furostanol Saponins and Saponin-Coated SilverNanoparticles. Appl Biochem Biotechnol 2017; 182:1675-1693. [PMID: 28236195 DOI: 10.1007/s12010-017-2426-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/24/2017] [Indexed: 10/20/2022]
Abstract
The aim of this investigation is to examine the anticancer activities of Balanites aegyptiaca fruit extract with its biogenic silver nanoparticles (AgNPs) against colon and liver cancer cells. B. aegyptiaca aqueous extract was fractionated according to polarity and by biosynthesized AgNP. The cytotoxicity of the extract, semi-purified fractions, and the AgNPs was examined on noncancerous cell lines. The safer fraction was subjected to ultra-performance liquid chromatography-MS to identify the major active constituents. The anticancer activities of the nontoxic doses of all the used treatments were tested against HepG2 and CaCo2 cells. The nontoxic dose of the B. aegyptiaca (0.63 mg/ml) extract showed high anti-proliferative activities against HepG2 and CaCo2 with a percentage of 81 and 77%, respectively. The butanol fraction was safer than the other two fractions with 46.3 and 90.35% anti-proliferative activity against Caco2 and HepG2 cells, respectively. The nontoxic dose of AgNPs (0.63 mg/ml) inhibits both HepG2 and Caco2 cells with a percentage of 84.5 and 83.4%, respectively. In addition, AgNPs regulate the expression of certain genes with folding higher than that of crude extract. Saponin-coated AgNPs showed great abilities to select the most anticancer ingredient(s) from the B. aegyptiaca extract with a more safety pattern than the polarity gradient fractionation.
Collapse
Affiliation(s)
- Abdelrahman M Yassin
- Biopharmaceutical Product Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, New Borg El-Arab City, Alexandria, 21934, Egypt
| | - Nehal M El-Deeb
- Biopharmaceutical Product Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, New Borg El-Arab City, Alexandria, 21934, Egypt.
| | - Ahmed M Metwaly
- Department of Pharmacognosy, Faculty of Pharmacy, Al-Azhar University, Cairo, 11371, Egypt
| | - Gomaa F El Fawal
- Polymeric Materials Research Department, Advanced Technology and New Materials Research Institute (ATNMRI), City of Scientific Research and Technological Applications, New Borg El-Arab City, Alexandria, 21934, Egypt
| | - Mohamed M Radwan
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, Oxford, MS, 38677, USA.,Department of Pharmacognosy, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Elsayed E Hafez
- Department of Plant Protection and Biomolecular Diagnosis, ALCRI, City of Scientific Research and Technological Applications, New Borg El-Arab City, Alexandria, 21934, Egypt
| |
Collapse
|
177
|
Albiñana V, Zafra MP, Colau J, Zarrabeitia R, Recio-Poveda L, Olavarrieta L, Pérez-Pérez J, Botella LM. Mutation affecting the proximal promoter of Endoglin as the origin of hereditary hemorrhagic telangiectasia type 1. BMC MEDICAL GENETICS 2017; 18:20. [PMID: 28231770 PMCID: PMC5324315 DOI: 10.1186/s12881-017-0380-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 02/09/2017] [Indexed: 11/23/2022]
Abstract
Background Hereditary hemorrhagic telangiectasia (HHT) is a vascular multi-organ system disorder. Its diagnostic criteria include epistaxis, telangiectases in mucocutaneous sites, arteriovenous malformations (AVMs), and familial inheritance. HHT is transmitted as an autosomal dominant condition, caused in 85% of cases by mutations in either Endoglin (ENG) or Activin receptor-like kinase (ACVRL1/ACVRL1/ALK1) genes. Pathogenic mutations have been described in exons, splice junctions and, in a few cases with ENG mutations, in the proximal promoter, which creates a new ATG start site. However, no mutations affecting transcription regulation have been described to date in HHT, and this type of mutation is rarely identified in the literature on rare diseases. Methods Sequencing data from a family with HHT lead to single nucleotide change, c.-58G > A. The functionality and pathogenicity of this change was analyzed by in vitro mutagenesis, quantitative PCR and Gel shift assay. Student t test was used for statistical significance. Results A single nucleotide change, c.-58G > A, in the proximal ENG promoter co-segregated with HHT clinical features in an HHT family. This mutation was present in the proband and in 2 other symptomatic members, whereas 2 asymptomatic relatives did not harbor the mutation. Analysis of RNA from activated monocytes from the probands and the healthy brother revealed reduced ENG mRNA expression in the HHT patient (p = 0.005). Site-directed mutagenesis of the ENG promoter resulted in a three-fold decrease in luciferase activity of the mutant c.-58A allele compared to wild type (p = 0.005). Finally, gel shift assay identified a DNA-protein specific complex. Conclusions The novel ENG c.-58G > A substitution in the ENG promoter co-segregates with HHT symptoms in a family and appears to affect the transcriptional regulation of the gene, resulting in reduced ENG expression. ENG c.-58G > A may therefore be a pathogenic HHT mutation leading to haploinsufficiency of Endoglin and HHT symptoms. To the best of our knowledge, this is the first report of a pathogenic mutation in HHT involving the binding site for a transcription factor in the promoter of ENG.
Collapse
Affiliation(s)
- Virginia Albiñana
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Ramiro de Maeztu 9, Madrid, 28040, Spain
| | - Ma Paz Zafra
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Ramiro de Maeztu 9, Madrid, 28040, Spain
| | - Jorge Colau
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Ramiro de Maeztu 9, Madrid, 28040, Spain
| | - Roberto Zarrabeitia
- HHT Spanish Unit, Hospital Sierrallana and Centro de InvestigacionBiomedica en Red de Enfermedades Raras (CIBERER), Torrelavega, Santander, Spain
| | - Lucia Recio-Poveda
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Ramiro de Maeztu 9, Madrid, 28040, Spain
| | | | | | - Luisa M Botella
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Ramiro de Maeztu 9, Madrid, 28040, Spain.
| |
Collapse
|
178
|
Decreased Endoglin expression in the pulmonary vasculature of nitrofen-induced congenital diaphragmatic hernia rat model. Pediatr Surg Int 2017; 33:263-268. [PMID: 27822781 DOI: 10.1007/s00383-016-4004-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/25/2016] [Indexed: 10/20/2022]
Abstract
AIM OF THE STUDY Pulmonary hypertension (PH) remains a therapeutical challenge in neonates born with congenital diaphragmatic hernia (CDH). Endoglin (Eng), an auxiliary receptor component of the transforming growth factor β (TGFβ) signalling pathway, is expressed mainly by endothelial cells and has been found to be involved in angiogenesis and vascular remodelling. Genetic studies have linked TGFβ and Eng mutations to human arterial PH and other cardiovascular syndromes. Eng interacts with the TGFβ receptors 1 and 2 (Tgfβr1, Tgfβr2). We designed this study to investigate the hypothesis that Eng is altered in the pulmonary vasculature of rats with nitrofen-induced CDH subjected to its interdependency with Tgfβr1 and Tgfβr2. METHODS After ethical approval (Rec 913b), time-pregnant Sprague-Dawley rats received either nitrofen or olive oil on gestational day (D9). The foetuses (n = 22) were sacrificed and divided into CDH and control group on D21. Gene and protein expressions of Eng, Tgfβr1 and Tgfβr2 were assessed via qRT-PCR and western blotting. Immunofluorescence staining for Eng was combined with CD34 to evaluate Eng expression in the pulmonary vasculature. MAIN RESULTS Relative mRNA levels of Eng, Tgfβr1 and Tgfβr2 were significantly downregulated in CDH lungs compared to controls (Eng CDH 0.341 ± 0.022, Eng Ctrl 0.471 ± 0.031, p = 0.0015; Tgfβr1 CDH 0.161 ± 0.008, Tgfβr1 Ctrl 0.194 ± 0.01, p = 0.0114; Tgfβr2 CDH 0.896 ± 0.099, Tgfβr2 Ctrl 1.379 ± 0.081, p = 0.0006) Western blotting confirmed the reduced pulmonary protein expression of these three proteins in the CDH lungs. A markedly diminished endothelial expression of Eng in the pulmonary vasculature of nitrofen-exposed foetuses compared to controls was seen in laser scanning confocal-microscopy. CONCLUSION This study demonstrates for the first time a reduced expression of Endoglin in the pulmonary vasculature of nitrofen-induced CDH. Abnormal Eng/Tgfβr1/Tgfβr2 signalling may contribute to impaired vascular remodelling and development of PH in this CDH animal model.
Collapse
|
179
|
Jarad M, Kuczynski EA, Morrison J, Viloria-Petit AM, Coomber BL. Release of endothelial cell associated VEGFR2 during TGF-β modulated angiogenesis in vitro. BMC Cell Biol 2017; 18:10. [PMID: 28114883 PMCID: PMC5260130 DOI: 10.1186/s12860-017-0127-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/18/2017] [Indexed: 01/11/2023] Open
Abstract
Background Sprouting angiogenesis requires vascular endothelial proliferation, migration and morphogenesis. The process is regulated by soluble factors, principally vascular endothelial growth factor (VEGF), and via bidirectional signaling through the Jagged/Notch system, leading to assignment of tip cell and stalk cell identity. The cytokine transforming growth factor beta (TGF-β) can either stimulate or inhibit angiogenesis via its differential surface receptor signaling. Here we evaluate changes in expression of angiogenic signaling receptors when bovine aortic endothelial cells were exposed to TGF-β1 under low serum conditions. Results TGF-β1 induced a dose dependent inhibition of tip cell assignment and subsequent angiogenesis on Matrigel, maximal at 5.0 ng/ml. This occurred via ALK5-dependent pathways and was accompanied by significant upregulation of the TGF-β co-receptor endoglin, and SMAD2 phosphorylation, but no alteration in Smad1/5 activation. TGF-β1 also induced ALK5-dependent downregulation of Notch1 but not of its ligand delta-like ligand 4. Cell associated VEGFR2 (but not VEGFR1) was significantly downregulated and accompanied by reciprocal upregulation of VEGFR2 in conditioned medium. Quantitative polymerase chain reaction analysis revealed that this soluble VEGFR2 was not generated by a selective shift in mRNA isoform transcription. This VEGFR2 in conditioned medium was full-length protein and was associated with increased soluble HSP-90, consistent with a possible shedding of microvesicles/exosomes. Conclusions Taken together, our results suggest that endothelial cells exposed to TGF-β1 lose both tip and stalk cell identity, possibly mediated by loss of VEGFR2 signaling. The role of these events in physiological and pathological angiogenesis requires further investigation. Electronic supplementary material The online version of this article (doi:10.1186/s12860-017-0127-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M Jarad
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada
| | - E A Kuczynski
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada
| | - J Morrison
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada
| | - A M Viloria-Petit
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada
| | - B L Coomber
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, OVC Room 3645, Guelph, N1G 2W1, ON, Canada.
| |
Collapse
|
180
|
Cruz-Solbes AS, Youker K. Epithelial to Mesenchymal Transition (EMT) and Endothelial to Mesenchymal Transition (EndMT): Role and Implications in Kidney Fibrosis. Results Probl Cell Differ 2017; 60:345-372. [PMID: 28409352 DOI: 10.1007/978-3-319-51436-9_13] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Tubulointerstitial injury is one of the hallmarks of renal disease. In particular, interstitial fibrosis has a prominent role in the development and progression of kidney injury. Collagen-producing fibroblasts are responsible for the ECM deposition. However, the origin of those activated fibroblasts is not clear. This chapter will discuss in detail the concept of epithelial to mesenchymal transition (EMT) and endothelial to mesenchymal transition (EndMT) in the context of fibrosis and kidney disease. In short, EMT and EndMT involve a change in cell shape, loss of polarity and increased motility associated with increased collagen production. Thus, providing a new source of fibroblasts. However, many controversies exist regarding the existence of EMT and EndMT in kidney disease, as well as its burden and role in disease development. The aim of this chapter is to provide an overview of the concepts and profibrotic pathways and to present the evidence that has been published in favor and against EMT and EndMT.
Collapse
|
181
|
The expanding role of neuropilin: regulation of transforming growth factor-β and platelet-derived growth factor signaling in the vasculature. Curr Opin Hematol 2016; 23:260-7. [PMID: 26849476 DOI: 10.1097/moh.0000000000000233] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW Long recognized for its role in regulation of vascular endothelial growth factor signaling, neuropilin (Nrp)1 has emerged as a modulator of additional signaling pathways critical for vascular development and function. Here we review two novel functions of Nrp1 in blood vessels: regulation of transforming growth factor-β (TGFβ) signaling in endothelial cells and regulation of platelet-derived growth factor (PDGF) signaling in vascular smooth muscle cells. RECENT FINDINGS Novel mouse models demonstrate that Nrp1 fulfills vascular functions independent of vascular endothelial growth factor signaling. These include modulation of TGFβ-dependent inhibition of endothelial sprouting during developmental angiogenesis and PDGF signaling in vascular smooth muscle cells during development and disease. SUMMARY Broadening our understanding of how and where Nrp1 functions in the vasculature is critical for the development of targeted therapeutics for cancer and vascular diseases such as atherosclerosis and retinopathies.
Collapse
|
182
|
Núñez-Gómez E, Pericacho M, Ollauri-Ibáñez C, Bernabéu C, López-Novoa JM. The role of endoglin in post-ischemic revascularization. Angiogenesis 2016; 20:1-24. [PMID: 27943030 DOI: 10.1007/s10456-016-9535-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 11/29/2016] [Indexed: 12/12/2022]
Abstract
Following arterial occlusion, blood vessels respond by forming a new network of functional capillaries (angiogenesis), by reorganizing preexisting capillaries through the recruitment of smooth muscle cells to generate new arteries (arteriogenesis) and by growing and remodeling preexisting collateral arterioles into physiologically relevant arteries (collateral development). All these processes result in the recovery of organ perfusion. The importance of endoglin in post-occlusion reperfusion is sustained by several observations: (1) endoglin expression is increased in vessels showing active angiogenesis/remodeling; (2) genetic endoglin haploinsufficiency in humans causes deficient angiogenesis; and (3) the reduction of endoglin expression by gene disruption or the administration of endoglin-neutralizing antibodies reduces angiogenesis and revascularization. However, the precise role of endoglin in the several processes associated with revascularization has not been completely elucidated and, in some cases, the function ascribed to endoglin by different authors is controversial. The purpose of this review is to organize in a critical way the information available for the role of endoglin in several phenomena (angiogenesis, arteriogenesis and collateral development) associated with post-ischemic revascularization.
Collapse
Affiliation(s)
- Elena Núñez-Gómez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Miguel Pericacho
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Claudia Ollauri-Ibáñez
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain.,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain
| | - Carmelo Bernabéu
- Centro de Investigaciones Biológicas, Spanish National Research Council (CIB, CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - José M López-Novoa
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain. .,Biomedical Research Institute of Salamanca (IBSAL), Salamanca, Spain.
| |
Collapse
|
183
|
Xu P, Lin X, Feng XH. Posttranslational Regulation of Smads. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a022087. [PMID: 27908935 DOI: 10.1101/cshperspect.a022087] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Transforming growth factor β (TGF-β) family signaling dictates highly complex programs of gene expression responses, which are extensively regulated at multiple levels and vary depending on the physiological context. The formation, activation, and destruction of two major functional complexes in the TGF-β signaling pathway (i.e., the TGF-β receptor complexes and the Smad complexes that act as central mediators of TGF-β signaling) are direct targets for posttranslational regulation. Dysfunction of these complexes often leads or contributes to pathogenesis in cancer and fibrosis and in cardiovascular, and autoimmune diseases. Here we discuss recent insights into the roles of posttranslational modifications in the functions of the receptor-activated Smads in the common Smad4 and inhibitory Smads, and in the control of the physiological responses to TGF-β. It is now evident that these modifications act as decisive factors in defining the intensity and versatility of TGF-β responsiveness. Thus, the characterization of posttranslational modifications of Smads not only sheds light on how TGF-β controls physiological and pathological processes but may also guide us to manipulate the TGF-β responses for therapeutic benefits.
Collapse
Affiliation(s)
- Pinglong Xu
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xia Lin
- Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030
| | - Xin-Hua Feng
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China.,Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Texas 77030.,Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
184
|
Ola R, Dubrac A, Han J, Zhang F, Fang JS, Larrivée B, Lee M, Urarte AA, Kraehling JR, Genet G, Hirschi KK, Sessa WC, Canals FV, Graupera M, Yan M, Young LH, Oh PS, Eichmann A. PI3 kinase inhibition improves vascular malformations in mouse models of hereditary haemorrhagic telangiectasia. Nat Commun 2016; 7:13650. [PMID: 27897192 PMCID: PMC5141347 DOI: 10.1038/ncomms13650] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 10/20/2016] [Indexed: 12/26/2022] Open
Abstract
Activin receptor-like kinase 1 (ALK1) is an endothelial serine-threonine kinase receptor for bone morphogenetic proteins (BMPs) 9 and 10. Inactivating mutations in the ALK1 gene cause hereditary haemorrhagic telangiectasia type 2 (HHT2), a disabling disease characterized by excessive angiogenesis with arteriovenous malformations (AVMs). Here we show that inducible, endothelial-specific homozygous Alk1 inactivation and BMP9/10 ligand blockade both lead to AVM formation in postnatal retinal vessels and internal organs including the gastrointestinal (GI) tract in mice. VEGF and PI3K/AKT signalling are increased on Alk1 deletion and BMP9/10 ligand blockade. Genetic deletion of the signal-transducing Vegfr2 receptor prevents excessive angiogenesis but does not fully revert AVM formation. In contrast, pharmacological PI3K inhibition efficiently prevents AVM formation and reverts established AVMs. Thus, Alk1 deletion leads to increased endothelial PI3K pathway activation that may be a novel target for the treatment of vascular lesions in HHT2.
Collapse
Affiliation(s)
- Roxana Ola
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Alexandre Dubrac
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Jinah Han
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Feng Zhang
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Jennifer S. Fang
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Bruno Larrivée
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Monica Lee
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Ana A. Urarte
- Vascular Signalling Laboratory, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Jan R. Kraehling
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Gael Genet
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Karen K. Hirschi
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - William C. Sessa
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Francesc V. Canals
- Translation Research Laboratory, Catalan Institute of Oncology, Idibell, Barcelona 08908, Spain
| | - Mariona Graupera
- Vascular Signalling Laboratory, Institut d'Investigació Biomèdica de Bellvitge, L'Hospitalet de Llobregat, Barcelona 08908, Spain
| | - Minhong Yan
- Molecular Oncology, Genentech, Inc., South San Francisco, California 94080-4990, USA
| | - Lawrence H. Young
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Paul S. Oh
- Department of Physiology and Functional Genomics, University of Florida College of Medicine, PO Box 100274, Gainesville, Florida 32610, USA
| | - Anne Eichmann
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06511, USA
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
- Inserm U970, Paris Cardiovascular Research Center, Paris 75015, France
| |
Collapse
|
185
|
Rapp J, Kiss E, Meggyes M, Szabo-Meleg E, Feller D, Smuk G, Laszlo T, Sarosi V, Molnar TF, Kvell K, Pongracz JE. Increased Wnt5a in squamous cell lung carcinoma inhibits endothelial cell motility. BMC Cancer 2016; 16:915. [PMID: 27876017 PMCID: PMC5120464 DOI: 10.1186/s12885-016-2943-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 11/09/2016] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Angiogenesis is important both in normal tissue function and disease and represents a key target in lung cancer (LC) therapy. Unfortunately, the two main subtypes of non-small-cell lung cancers (NSCLC) namely, adenocarcinoma (AC) and squamous cell carcinoma (SCC) respond differently to anti-angiogenic e.g. anti-vascular endothelial growth factor (VEGF)-A treatment with life-threatening side effects, often pulmonary hemorrhage in SCC. The mechanisms behind such adverse reactions are still largely unknown, although peroxisome proliferator activator receptor (PPAR) gamma as well as Wnt-s have been named as molecular regulators of the process. As the Wnt microenvironments in NSCLC subtypes are drastically different, we hypothesized that the particularly high levels of non-canonical Wnt5a in SCC might be responsible for alterations in blood vessel growth and result in serious adverse reactions. METHODS PPARgamma, VEGF-A, Wnt5a, miR-27b and miR-200b levels were determined in resected adenocarcinoma and squamous cell carcinoma samples by qRT-PCR and TaqMan microRNA assay. The role of PPARgamma in VEGF-A expression, and the role of Wnts in overall regulation was investigated using PPARgamma knock-out mice, cancer cell lines and fully human, in vitro 3 dimensional (3D), distal lung tissue aggregates. PPARgamma mRNA and protein levels were tested by qRT-PCR and immunohistochemistry, respectively. PPARgamma activity was measured by a PPRE reporter system. The tissue engineered lung tissues expressing basal level and lentivirally delivered VEGF-A were treated with recombinant Wnts, chemical Wnt pathway modifiers, and were subjected to PPARgamma agonist and antagonist treatment. RESULTS PPARgamma down-regulation and VEGF-A up-regulation are characteristic to both AC and SCC. Increased VEGF-A levels are under direct control of PPARgamma. PPARgamma levels and activity, however, are under Wnt control. Imbalance of both canonical (in AC) and non-canonical (in SCC) Wnts leads to PPARgamma down-regulation. While canonical Wnts down-regulate PPARgamma directly, non-canonical Wnt5a increases miR27b that is known regulator of PPARgamma. CONCLUSION During carcinogenesis the Wnt microenvironment alters, which can downregulate PPARgamma leading to increased VEGF-A expression. Differences in the Wnt microenvironment in AC and SCC of NSCLC lead to PPARgamma decrease via mechanisms that differentially alter endothelial cell motility and branching which in turn can influence therapeutic response.
Collapse
MESH Headings
- Adenocarcinoma/blood supply
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Animals
- Biomarkers, Tumor
- Carcinoma, Non-Small-Cell Lung/blood supply
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Squamous Cell/blood supply
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Movement
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lung Neoplasms/blood supply
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- MicroRNAs/genetics
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- PPAR gamma/physiology
- Tumor Cells, Cultured
- Tumor Microenvironment
- Vascular Endothelial Growth Factor A/metabolism
- Wnt-5a Protein/metabolism
Collapse
Affiliation(s)
- J Rapp
- Department of Pharmaceutical Biotechnology, School of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, 7624, Hungary
- János Szentágothai Research Centre, University of Pécs, 20 Ifjúság Str, Pecs, 7622, Hungary
- Humeltis Ltd, János Szentágothai Research Center, University of Pécs, 20 Ifjúság Str, Pécs, 7622, Hungary
| | - E Kiss
- Department of Pharmaceutical Biotechnology, School of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, 7624, Hungary
- János Szentágothai Research Centre, University of Pécs, 20 Ifjúság Str, Pecs, 7622, Hungary
- Humeltis Ltd, János Szentágothai Research Center, University of Pécs, 20 Ifjúság Str, Pécs, 7622, Hungary
| | - M Meggyes
- Medical Microbiology and Immunity, University of Pécs, 12 Szigeti Str, Pécs, 7624, Hungary
- Humeltis Ltd, János Szentágothai Research Center, University of Pécs, 20 Ifjúság Str, Pécs, 7622, Hungary
| | - E Szabo-Meleg
- Biophysics, University of Pécs, 12 Szigeti Str, Pécs, 7624, Hungary
- János Szentágothai Research Centre, University of Pécs, 20 Ifjúság Str, Pecs, 7622, Hungary
| | - D Feller
- Department of Pharmaceutical Biotechnology, School of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, 7624, Hungary
- János Szentágothai Research Centre, University of Pécs, 20 Ifjúság Str, Pecs, 7622, Hungary
- Humeltis Ltd, János Szentágothai Research Center, University of Pécs, 20 Ifjúság Str, Pécs, 7622, Hungary
| | - G Smuk
- Pathology, University of Pécs, 12 Szigeti Str, Pécs, 7624, Hungary
| | - T Laszlo
- Pathology, University of Pécs, 12 Szigeti Str, Pécs, 7624, Hungary
| | - V Sarosi
- Internal Medicine, Pulmonology, University of Pécs, 2 Rakoczi Str, Pécs, 7623, Hungary
| | - T F Molnar
- Operational Medicine, University of Pécs, 12 Szigeti Str, Pécs, 7624, Hungary
- Department of Surgery, Thoracic Surgery Unit, Petz A Hospital, 2-4 Vasvari Str, Győr, 9023, Hungary
| | - K Kvell
- Department of Pharmaceutical Biotechnology, School of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, 7624, Hungary
- János Szentágothai Research Centre, University of Pécs, 20 Ifjúság Str, Pecs, 7622, Hungary
| | - J E Pongracz
- Department of Pharmaceutical Biotechnology, School of Pharmacy, University of Pecs, 2 Rokus Str, Pecs, 7624, Hungary.
- János Szentágothai Research Centre, University of Pécs, 20 Ifjúság Str, Pecs, 7622, Hungary.
- Humeltis Ltd, János Szentágothai Research Center, University of Pécs, 20 Ifjúság Str, Pécs, 7622, Hungary.
| |
Collapse
|
186
|
Vorstenbosch J, Nguyen CM, Zhou S, Seo YJ, Siblini A, Finnson KW, Bizet AA, Tran SD, Philip A. Overexpression of CD109 in the Epidermis Differentially Regulates ALK1 Versus ALK5 Signaling and Modulates Extracellular Matrix Synthesis in the Skin. J Invest Dermatol 2016; 137:641-649. [PMID: 27866969 DOI: 10.1016/j.jid.2016.09.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 09/25/2016] [Indexed: 12/16/2022]
Abstract
Transforming growth factor-β (TGF-β) is a multifunctional growth factor involved in many physiological processes including wound healing and inflammation. Excessive TGF-β signaling in the skin has been implicated in fibrotic skin disorders such as keloids and scleroderma. We previously identified CD109 as a TGF-β co-receptor and inhibitor of TGF-β signaling and have shown that transgenic mice overexpressing CD109 in the epidermis display decreased scarring. In certain cell types, in addition to the canonical type I receptor, ALK5, which activates Smad2/3, TGF-β can signal through another type I receptor, ALK1, which activates Smad1/5. Here we demonstrate that ALK1 is expressed and co-localizes with CD109 in mouse keratinocytes and that mice overexpressing CD109 in the epidermis display enhanced ALK1-Smad1/5 signaling but decreased ALK5-Smad2/3 signaling, TGF-β expression, and extracellular matrix production in the skin when compared with wild-type littermates. Furthermore, treatment with conditioned media from isolated keratinocytes or epidermal explants from CD109 transgenic mouse skin leads to a decrease in extracellular matrix production in mouse skin fibroblasts. Taken together, our findings suggest that CD109 differentially regulates TGF-β-induced ALK1-Smad1/5 versus ALK5-Smad2/3 pathways, leading to decreased extracellular matrix production in the skin and that epidermal CD109 expression regulates dermal function through a paracrine mechanism.
Collapse
Affiliation(s)
- Joshua Vorstenbosch
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Christopher M Nguyen
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Shufeng Zhou
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - You Jung Seo
- Faculty of Dentistry, McGill University, McGill University, Montreal, Canada
| | - Aya Siblini
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Kenneth W Finnson
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Albane A Bizet
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada
| | - Simon D Tran
- Faculty of Dentistry, McGill University, McGill University, Montreal, Canada
| | - Anie Philip
- Division of Plastic Surgery, Department of Surgery, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
187
|
Doerr M, Morrison J, Bergeron L, Coomber BL, Viloria-Petit A. Differential effect of hypoxia on early endothelial–mesenchymal transition response to transforming growth beta isoforms 1 and 2. Microvasc Res 2016; 108:48-63. [DOI: 10.1016/j.mvr.2016.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Revised: 07/31/2016] [Accepted: 08/04/2016] [Indexed: 01/16/2023]
|
188
|
Blair LA, Haven AK, Bauer NN. Circulating microparticles in severe pulmonary arterial hypertension increase intercellular adhesion molecule-1 expression selectively in pulmonary artery endothelium. Respir Res 2016; 17:133. [PMID: 27765042 PMCID: PMC5073933 DOI: 10.1186/s12931-016-0445-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Accepted: 10/05/2016] [Indexed: 01/27/2023] Open
Abstract
Background Microparticles (MPs) stimulate inflammatory adhesion molecule expression in systemic vascular diseases, however it is unknown whether circulating MPs stimulate localized ICAM-1 expression in the heterogeneically distinct pulmonary endothelium during pulmonary arterial hypertension (PAH). Pulmonary vascular lesions with infiltrating inflammatory cells in PAH form in the pulmonary arteries and arterioles, but not the microcirculation. Therefore, we sought to determine whether circulating MPs from PAH stimulate pulmonary artery endothelial cell-selective ICAM-1 expression. Results Pulmonary artery endothelial cells (PAECs) were exposed to MPs isolated from the circulation of a rat model of severe PAH. During late-stage (8-weeks) PAH, but not early-stage (3-weeks), an increase in ICAM-1 was observed. To determine whether PAH MP-induced ICAM-1 was selective for a specific segment of the pulmonary circulation, pulmonary microvascular endothelial cells (PMVECs) were exposed to late-stage PAH MPs and no increase in ICAM-1 was detected. A select population of circulating MPs, the late-stage endoglin + MPs, were used to assess their ability to stimulate ICAM-1 and it was determined that the endoglin + MPs were sufficient to promote ICAM-1 increases in the whole cell, but not surface only expression. Conclusions Late-stage, but not early-stage, MPs in a model of severe PAH selectively induce ICAM-1 in pulmonary artery endothelium, but not pulmonary microcirculation. Further, the selected endoglin + PAH MPs, but not endoglin + MPs from control, are sufficient to promote whole cell ICAM-1 in PAECs. The implications of this work are that MPs in late-stage PAH are capable of inducing ICAM-1 expression selectively in the pulmonary artery. ICAM-1 likely plays a significant role in the observed inflammatory cell recruitment, specifically to vascular lesions in the pulmonary artery and not the pulmonary microcirculation. Electronic supplementary material The online version of this article (doi:10.1186/s12931-016-0445-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Leslie A Blair
- Department of Pharmacology, College of Medicine, University of South Alabama, 5851 USA Drive North, MSB 3340, Mobile, AL, 36688, USA.,Center for Lung Biology, College of Medicine, University of South Alabama, 5851 USA Drive North, MSB 3340, Mobile, AL, 36688, USA
| | - April K Haven
- Department of Pharmacology, College of Medicine, University of South Alabama, 5851 USA Drive North, MSB 3340, Mobile, AL, 36688, USA.,Center for Lung Biology, College of Medicine, University of South Alabama, 5851 USA Drive North, MSB 3340, Mobile, AL, 36688, USA
| | - Natalie N Bauer
- Department of Pharmacology, College of Medicine, University of South Alabama, 5851 USA Drive North, MSB 3340, Mobile, AL, 36688, USA. .,Center for Lung Biology, College of Medicine, University of South Alabama, 5851 USA Drive North, MSB 3340, Mobile, AL, 36688, USA.
| |
Collapse
|
189
|
Krishnan S, Szabo E, Burghardt I, Frei K, Tabatabai G, Weller M. Modulation of cerebral endothelial cell function by TGF-β in glioblastoma: VEGF-dependent angiogenesis versus endothelial mesenchymal transition. Oncotarget 2016; 6:22480-95. [PMID: 26090865 PMCID: PMC4673177 DOI: 10.18632/oncotarget.4310] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/03/2015] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma are among the most angiogenic tumors. The molecular mechanisms that control blood vessel formation by endothelial cells (EC) in glioblastoma remain incompletely understood. Transforming growth factor-β (TGF-β) is a key regulatory cytokine that has proinvasive and stemness-maintaining autocrine properties in glioblastoma and confers immunosuppression to the tumor microenvironment. Here we characterize potential pro- and anti-angiogenic activities of TGF-β in the context of glioblastoma in vitro, using human brain-derived microvascular endothelial cells (hCMEC/D3) and glioblastoma-derived endothelial cells (GMEC) as model systems. We find that TGF-β induces vascular endothelial growth factor (VEGF) and placental growth factor (PlGF) mRNA expression and protein release in a TGF-β receptor (TβR) II / activin-like kinase (ALK)-5-dependent manner under normoxia and hypoxia, defining potential indirect proangiogenic activity of TGF-β in glioblastoma. In parallel, exogenous TGF-β has also inhibitory effects on EC properties and induces endothelial-mesenchymal transition (EndMT) in hCMEC and GMEC. Accordingly, direct inhibition of endogenous TGF-β/ALK-5 signalling increases EC properties such as tube formation, von-Willebrand factor (vWF) and claudin (CLDN) 5 expression. Yet, the supernatant of TGF-β-stimulated hCMEC and GMEC strongly promotes EC-related gene expression and tube formation in a cediranib-sensitive manner. These observations shed light on the complex pro- and anti-angiogenic pathways involving the cross-talk between TGF-β and VEGF/PLGF signalling in glioblastoma which may involve parallel stimulation of angiogenesis and EndMT in distinct target cell populations.
Collapse
Affiliation(s)
- Shanmugarajan Krishnan
- Laboratory of Molecular Neuro-Oncology, Department of Neurology and Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Emese Szabo
- Laboratory of Molecular Neuro-Oncology, Department of Neurology and Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Isabel Burghardt
- Laboratory of Molecular Neuro-Oncology, Department of Neurology and Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Karl Frei
- Laboratory of Molecular Neuro-Oncology, Department of Neurosurgery and Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Ghazaleh Tabatabai
- Laboratory of Molecular Neuro-Oncology, Department of Neurology and Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland.,Interdisciplinary Division of Neuro-Oncology, Departments of Vascular Neurology and Neurosurgery, University Hospital Tübingen, Tübingen, Germany
| | - Michael Weller
- Laboratory of Molecular Neuro-Oncology, Department of Neurology and Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
190
|
Reichman D, Man L, Park L, Lis R, Gerhardt J, Rosenwaks Z, James D. Notch hyper-activation drives trans-differentiation of hESC-derived endothelium. Stem Cell Res 2016; 17:391-400. [PMID: 27643563 DOI: 10.1016/j.scr.2016.09.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 09/01/2016] [Accepted: 09/13/2016] [Indexed: 12/28/2022] Open
Abstract
During development, endothelial cells (EC) display tissue-specific attributes that are unique to each vascular bed, as well as generic signaling mechanisms that are broadly applied to create a patent circulatory system. We have previously utilized human embryonic stem cells (hESC) to generate tissue-specific EC sub-types (Rafii et al., 2013) and identify pathways that govern growth and trans-differentiation potential of hESC-derived ECs (James et al., 2010). Here, we elucidate a novel Notch-dependent mechanism that induces endothelial to mesenchymal transition (EndMT) in confluent monolayer cultures of hESC-derived ECs. We demonstrate density-dependent induction of EndMT that can be rescued by the Notch signaling inhibitor DAPT and identify a positive feedback signaling mechanism in hESC-ECs whereby trans-activation of Notch by DLL4 ligand induces elevated expression and surface presentation of DLL4. Increased Notch activation in confluent hESC-EC monolayer cultures induces areas of EndMT containing transitional cells that are marked by increased Jagged1 expression and reduced Notch signal integration. Jagged1 loss of function in monolayer hESC-ECs induces accelerated feedback stimulation of Notch signaling, increased expression of cell-autonomous, cis-inhibitory DLL4, and EndMT. These data elucidate a novel interplay of Notch ligands in modulating pathway activation during both expansion and EndMT of hESC-derived ECs.
Collapse
Affiliation(s)
- David Reichman
- Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY 10065, United States
| | - Limor Man
- Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY 10065, United States
| | - Laura Park
- Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY 10065, United States
| | - Raphael Lis
- Tri-Institutional Stem Cell Derivation Laboratory, Weill Cornell Medical College, New York, NY 10065, United States
| | - Jeannine Gerhardt
- Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY 10065, United States
| | - Zev Rosenwaks
- Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY 10065, United States
| | - Daylon James
- Center for Reproductive Medicine and Infertility, Weill Cornell Medical College, New York, NY 10065, United States; Tri-Institutional Stem Cell Derivation Laboratory, Weill Cornell Medical College, New York, NY 10065, United States.
| |
Collapse
|
191
|
Immunohistochemical Expression of CD105 and TGF-β1 in Oral Squamous Cell Carcinoma and Adjacent Apparently Normal Oral Mucosa and its Correlation With Clinicopathologic Features. Appl Immunohistochem Mol Morphol 2016; 24:35-41. [PMID: 25710582 DOI: 10.1097/pai.0000000000000152] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Angiogenesis in oral squamous cell carcinomas (OSCC) is essential for its growth, invasion, and metastasis. This entails a shift in the balance between proangiogenic and antiangiogenic factors. CD105 and TGF-β1 are 2 such proangiogenic factors wherein CD105 exerts its angiogenic effect by binding to and modulating the TGF-β1 pathway. A total of 50 resected specimens of OSCC were considered. One tissue specimen was taken from tumor proper and another specimen from adjacent apparently normal mucosa (AANM). Both tissues were immunohistochemically stained using CD105 and TGF-β1 antibodies. The expression of each antibody was individually assessed and then compared. Pearson χ test was used for statistical comparison of expression. CD105 was significantly expressed in OSCC as compared with AANM and also correlated with increasing TNM stage. The mean microvessel density was higher in OSCC. TGF-β1 was significantly expressed in epithelium of OSCC as compared with AANM. On comparing expression of TGF-β1 and CD105, 79.54% of endothelial cells expressed positivity for both molecules. Both CD105 and TGF-β1 were increased in OSCC, although based on our results CD105 alone can be used as a prognostic marker. On the basis of immunohistochemical expression of CD105 and TGF-β1 in endothelial cells, our results demonstrate that CD105 acts as one of the receptors of TGF-β1 on endothelial cells and induces the angiogenic pathway in OSCC.
Collapse
|
192
|
Jin M, Lee J, Lee KY, Jin Z, Pak JH, Kim HS. Alteration of TGF-β-ALK-Smad signaling in hyperoxia-induced bronchopulmonary dysplasia model of newborn rats. Exp Lung Res 2016; 42:354-364. [PMID: 27618520 DOI: 10.1080/01902148.2016.1226448] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Bronchopulmonary dysplasia (BPD) is a main chronic lung disease commonly occurs in preterm infants. BPD is characterized by impaired alveolarization and vascularization of the developing lung. Transforming growth factor-β (TGF-β) signaling pathway is known to play an important role during lung vascular development. In the present study, we examined whether the regulation of TGF-β-ALK-Smad signaling pathway influence on the disruption of pulmonary vascular development in newborn rats as hyperoxia-induced BPD model. MATERIALS AND METHODS Newborn rats were continuously exposed to 21% or 85% O2 for 7 days, and subsequently kept in normoxic condition for another 14 days. Lung tissues harvested at each time point were evaluated for the expression of TGF-β1, ALK1, ALK5, phosphorylated Smad1/5, phosphorylated Smad2/3, VEGF, and endoglin, as accessed by both biochemical and immunohistological analyses. RESULTS Double-fluorescence immunohistochemical staining indicated these molecules were mainly expressed in pulmonary endothelial cells. The expression of TGF-β1 and ALK5 mRNA and protein were significantly increased in D5 hyperoxia group, while that of ALK1 mRNA and protein were significantly decreased. The level of phosphorylated Smad1/5 was significantly decreased in D7 hyperoxia group, whereas that of phosphorylated Smad2/3 was oppositely increased. In addition, the expression of vascular endothelial growth factor (VEGF) mRNA was increased at D1 with subsequent decrease in D7 hyperoxia group. There was no significantly difference in endoglin expression in entire experimental period. CONCLUSION These results indicate that exposure to hyperoxia altered the balance between TGF-β-ALK1-Smad1/5 and TGF-β-ALK5-Smad2/3 pathways in pulmonary endothelial cells, which may ultimately lead to the development of BPD.
Collapse
Affiliation(s)
- Meihua Jin
- a Department of Pediatrics , Yanbian University Hospital , Yanji , Jilin Province , China
| | - Juyoung Lee
- b Department of Pediatrics , Inha University College of Medicine , Incheon , Korea
| | - Kyung-Yup Lee
- c Department of Pediatrics , Seoul National University College of Medicine , Seoul , Korea
| | - Zhengyong Jin
- a Department of Pediatrics , Yanbian University Hospital , Yanji , Jilin Province , China
| | - Jhang Ho Pak
- d Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine , Seoul , Korea
| | - Han-Suk Kim
- c Department of Pediatrics , Seoul National University College of Medicine , Seoul , Korea
| |
Collapse
|
193
|
Ottley EC, Nicholson HD, Gold EJ. Activin A regulates microRNAs and gene expression in LNCaP cells. Prostate 2016; 76:951-63. [PMID: 27018851 DOI: 10.1002/pros.23184] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 03/11/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Prostate cancer (PCa) is an increasing health issue worldwide. For patients with advanced castration-resistant PCa (CRPC) treatment options are limited and overall survival is relatively short. Paired with this, non-invasive diagnostic options are yet to be established. Activins are members of the TGF-β superfamily and have been linked to prostate physiology. For instance, activin A is an inhibitor of growth in the prostate. A novel class of non-coding RNA, microRNAs (miRNAs) have been intrinsically linked to a range of cellular processes and carcinogenesis. No studies have investigated the impact of activin A on miRNA expression in PCa cell lines. Hence, the objective of this study was to determine the effect of activin A on miRNA expression and downstream target genes in PCa. METHODS Activin-sensitive (LNCaP) and insensitive (PC3) prostate cells were treated with 50 ng/ml of activin A for 72 hr. To examine miRNA expression following treatment, SYBR RT-qPCR miRNA arrays were used in conjunction with TaqMan RT-qPCR. MiRPath-TarBase analysis was conducted using the miRNAs that were significantly altered following activin A treatment of LNCaP cells to highlight enriched target genes within biological pathways. Highlighted target genes were assessed using pathway-focused TGF-β and cell cycle SYBR RT-qPCR arrays. RESULTS Activin A treatment altered nine miRNAs in LNCaP cells: miR-222-3p, miR-15b-5p, miR-93-5p, miR-18a-5p, and let-7i-5p were significantly decreased, while miR-30a/30d-5p, let-7c, and miR-196b-5p were significantly increased versus media control. In PC3 cells five miRNAs were altered: miR-130a-3p, miR-7-5p, and miR-140-3p were significantly decreased while miR-191-5p and miR-26a-5p were significantly increased versus media control. MiRPath-TarBase analysis highlighted that the miRNAs significantly altered in LNCaP cells targeted genes contained in activin A-related KEGG pathways. Furthermore, when LNCaP cells were treated with activin A the expression of the targeted genes was the inverse of the expression of activin A-mediated miRNAs. CONCLUSIONS This study demonstrated the ability of activin A to modulate miRNA expression in PCa cell lines and suggests a correlative relationship between miRNA expression and downstream target genes in LNCaP cells. This study provides impetus for further studies into activin A and miRNAs in PCa. Prostate 76:951-963, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
|
194
|
Abstract
Transforming growth factor β (TGF-β) family members signal via heterotetrameric complexes of type I and type II dual specificity kinase receptors. The activation and stability of the receptors are controlled by posttranslational modifications, such as phosphorylation, ubiquitylation, sumoylation, and neddylation, as well as by interaction with other proteins at the cell surface and in the cytoplasm. Activation of TGF-β receptors induces signaling via formation of Smad complexes that are translocated to the nucleus where they act as transcription factors, as well as via non-Smad pathways, including the Erk1/2, JNK and p38 MAP kinase pathways, and the Src tyrosine kinase, phosphatidylinositol 3'-kinase, and Rho GTPases.
Collapse
Affiliation(s)
- Carl-Henrik Heldin
- Ludwig Institute for Cancer Research Ltd., Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden
| | - Aristidis Moustakas
- Ludwig Institute for Cancer Research Ltd., Science for Life Laboratory, Uppsala University, SE-751 24 Uppsala, Sweden Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Uppsala University, SE-751 23 Uppsala, Sweden
| |
Collapse
|
195
|
Abstract
The mechanisms underlying discrimination between "self" and "non-self", a central immunological principle, require careful consideration in immune oncology therapeutics where eliciting anti-cancer immunity must be weighed against the risk of autoimmunity due to the self origin of tumors. Whole cell vaccines are one promising immunotherapeutic avenue whereby a myriad of tumor antigens are introduced in an immunogenic context with the aim of eliciting tumor rejection. Despite the possibility collateral damage to healthy tissues, cancer immunotherapy can be designed such that off target autoimmunity remains limited in scope and severity or completely non-existent. Here we provide an immunological basis for reconciling the safety of cancer vaccines, focusing on tumor endothelial cell vaccines, by discussing the following topics: (a) Antigenic differences between neoplastic and healthy tissues that can be leveraged in cancer vaccine design; (b) The layers of tolerance that control T cell responses directed against antigens expressed in healthy tissues and tumors; and, (c) The hierarchy of antigenic epitope selection and display in response to whole cell vaccines, and how antigen processing and presentation can afford a degree of selectivity against tumors. We conclude with an example of early clinical data utilizing ValloVax™, an immunogenic placental endothelial cell vaccine that is being advanced to target the tumor endothelium of diverse cancers, and we report on the safety and efficacy of ValloVax™ for inducing immunity against tumor endothelial antigens.
Collapse
|
196
|
Gkatzis K, Thalgott J, Dos-Santos-Luis D, Martin S, Lamandé N, Carette MF, Disch F, Snijder RJ, Westermann CJ, Mager JJ, Oh SP, Miquerol L, Arthur HM, Mummery CL, Lebrin F. Interaction Between ALK1 Signaling and Connexin40 in the Development of Arteriovenous Malformations. Arterioscler Thromb Vasc Biol 2016; 36:707-17. [PMID: 26821948 DOI: 10.1161/atvbaha.115.306719] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/20/2016] [Indexed: 01/01/2023]
Abstract
OBJECTIVE To determine the role of Gja5 that encodes for the gap junction protein connexin40 in the generation of arteriovenous malformations in the hereditary hemorrhagic telangiectasia type 2 (HHT2) mouse model. APPROACH AND RESULTS We identified GJA5 as a target gene of the bone morphogenetic protein-9/activin receptor-like kinase 1 signaling pathway in human aortic endothelial cells and importantly found that connexin40 levels were particularly low in a small group of patients with HHT2. We next took advantage of the Acvrl1(+/-) mutant mice that develop lesions similar to those in patients with HHT2 and generated Acvrl1(+/-); Gja5(EGFP/+) mice. Gja5 haploinsufficiency led to vasodilation of the arteries and rarefaction of the capillary bed in Acvrl1(+/-) mice. At the molecular level, we found that reduced Gja5 in Acvrl1(+/-) mice stimulated the production of reactive oxygen species, an important mediator of vessel remodeling. To normalize the altered hemodynamic forces in Acvrl1(+/-); Gja5(EGFP/+) mice, capillaries formed transient arteriovenous shunts that could develop into large malformations when exposed to environmental insults. CONCLUSIONS We identified GJA5 as a potential modifier gene for HHT2. Our findings demonstrate that Acvrl1 haploinsufficiency combined with the effects of modifier genes that regulate vessel caliber is responsible for the heterogeneity and severity of the disease. The mouse models of HHT have led to the proposal that 3 events-heterozygosity, loss of heterozygosity, and angiogenic stimulation-are necessary for arteriovenous malformation formation. Here, we present a novel 3-step model in which pathological vessel caliber and consequent altered blood flow are necessary events for arteriovenous malformation development.
Collapse
MESH Headings
- Activin Receptors, Type I/genetics
- Activin Receptors, Type I/metabolism
- Activin Receptors, Type II/genetics
- Activin Receptors, Type II/metabolism
- Animals
- Arteriovenous Malformations/enzymology
- Arteriovenous Malformations/genetics
- Arteriovenous Malformations/pathology
- Cells, Cultured
- Connexins/genetics
- Connexins/metabolism
- Disease Models, Animal
- Endothelial Cells/enzymology
- Genetic Predisposition to Disease
- Haploinsufficiency
- Humans
- Mice, Mutant Strains
- Mice, Transgenic
- Neovascularization, Pathologic
- Phenotype
- RNA Interference
- Reactive Oxygen Species/metabolism
- Retinal Vessels/enzymology
- Retinal Vessels/pathology
- Signal Transduction
- Telangiectasia, Hereditary Hemorrhagic/enzymology
- Telangiectasia, Hereditary Hemorrhagic/genetics
- Telangiectasia, Hereditary Hemorrhagic/pathology
- Transfection
- Vascular Remodeling
- Gap Junction alpha-5 Protein
Collapse
Affiliation(s)
- Konstantinos Gkatzis
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Jérémy Thalgott
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Damien Dos-Santos-Luis
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Sabrina Martin
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Noël Lamandé
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Marie France Carette
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Frans Disch
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Repke J Snijder
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Cornelius J Westermann
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Johannes J Mager
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - S Paul Oh
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Lucile Miquerol
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Helen M Arthur
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Christine L Mummery
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.)
| | - Franck Lebrin
- From the Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands (K.G., C.L.M.); CNRS Unité mixte de recherche 7241/INSERM U1050, Center for Interdisciplinary Research in Biology, Collège de France, Paris cedex 05, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); MEMOLIFE Laboratory of Excellence, Paris Sciences et Lettres Research University, Paris, France (J.T., D.D.-S.-L., S.M., N.L., F.L.); Department of Radiology, AP-HP, Tenon Hospital, Paris, France (M.F.C.); Sorbonne Universités, UPMC University, Paris, France (M.F.C.); St. Antonius Hospital, Nieuwegein, The Netherlands (F.D., R.J.S., C.J.W., J.J.M.); Department of Physiology and Functional Genomics, College of Medicine, University of Florida, Gainesville (S.P.O.); Aix Marseille Université, CNRS IBDM UMR 7288, Marseille cedex 09, France (L.M.); and Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom (H.M.A.).
| |
Collapse
|
197
|
Mice Lacking Endoglin in Macrophages Show an Impaired Immune Response. PLoS Genet 2016; 12:e1005935. [PMID: 27010826 PMCID: PMC4806930 DOI: 10.1371/journal.pgen.1005935] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/24/2016] [Indexed: 12/26/2022] Open
Abstract
Endoglin is an auxiliary receptor for members of the TGF-β superfamily and plays an important role in the homeostasis of the vessel wall. Mutations in endoglin gene (ENG) or in the closely related TGF-β receptor type I ACVRL1/ALK1 are responsible for a rare dominant vascular dysplasia, the Hereditary Hemorrhagic Telangiectasia (HHT), or Rendu-Osler-Weber syndrome. Endoglin is also expressed in human macrophages, but its role in macrophage function remains unknown. In this work, we show that endoglin expression is triggered during the monocyte-macrophage differentiation process, both in vitro and during the in vivo differentiation of blood monocytes recruited to foci of inflammation in wild-type C57BL/6 mice. To analyze the role of endoglin in macrophages in vivo, an endoglin myeloid lineage specific knock-out mouse line (Engfl/flLysMCre) was generated. These mice show a predisposition to develop spontaneous infections by opportunistic bacteria. Engfl/flLysMCre mice also display increased survival following LPS-induced peritonitis, suggesting a delayed immune response. Phagocytic activity is impaired in peritoneal macrophages, altering one of the main functions of macrophages which contributes to the initiation of the immune response. We also observed altered expression of TGF-β1 target genes in endoglin deficient peritoneal macrophages. Overall, the altered immune activity of endoglin deficient macrophages could help to explain the higher rate of infectious diseases seen in HHT1 patients. Endoglin is a transmembrane protein and an auxiliary receptor for TGF-β with an important role in the homeostasis of the vessel wall. However, endoglin was originally identified as a human cell surface antigen expressed in a pre-B leukemic cell line. Mutations in ENG are responsible for the Hereditary Hemorrhagic Telangiectasia type 1 (HHT1) or Rendu-Osler-Weber syndrome. HHT is a rare disease, with a prevalence of 1/5,000 to 1/8,000. It is an autosomal dominant disorder characterized by a multisystemic vascular dysplasia, recurrent hemorrhages and arteriovenous malformations in internal organs. Interestingly, endoglin expression is also triggered during the monocyte-macrophage differentiation process. In our laboratory, we described that up-regulation of endoglin during in vitro differentiation of blood monocytes is age-dependent and impaired in monocytes from HHT patients, suggesting a role of endoglin in macrophages. In the present work, we first analyzed endoglin expression during differentiation of peripheral blood monocytes to macrophages under in vitro and in vivo conditions. Next, to investigate endoglin’s role in macrophage function in vivo, a myeloid-lineage specific endoglin knock-out mouse line was generated (Engfl/flLysMCre). Endoglin deficiency in macrophages predisposed animals to spontaneous infections and led to delayed endotoxin-induced mortality. Phagocytic activity by peritoneal macrophages was reduced in the absence of endoglin and altered expression of TGF-β target genes was consistent with an altered balance of TGF-β signaling. The results show a novel role for endoglin in mouse macrophages, which if analogous in human macrophages, may explain, at least in part, the increased infection rates seen in HHT patients.
Collapse
|
198
|
Clark CR, Robinson JY, Sanchez NS, Townsend TA, Arrieta JA, Merryman WD, Trykall DZ, Olivey HE, Hong CC, Barnett JV. Common pathways regulate Type III TGFβ receptor-dependent cell invasion in epicardial and endocardial cells. Cell Signal 2016; 28:688-98. [PMID: 26970186 DOI: 10.1016/j.cellsig.2016.03.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 02/23/2016] [Accepted: 03/08/2016] [Indexed: 11/29/2022]
Abstract
Epithelial-Mesenchymal Transformation (EMT) and the subsequent invasion of epicardial and endocardial cells during cardiac development is critical to the development of the coronary vessels and heart valves. The transformed cells give rise to cardiac fibroblasts and vascular smooth muscle cells or valvular interstitial cells, respectively. The Type III Transforming Growth Factor β (TGFβR3) receptor regulates EMT and cell invasion in both cell types, but the signaling mechanisms downstream of TGFβR3 are not well understood. Here we use epicardial and endocardial cells in in vitro cell invasion assays to identify common mechanisms downstream of TGFβR3 that regulate cell invasion. Inhibition of NF-κB activity blocked cell invasion in epicardial and endocardial cells. NF-κB signaling was found to be dysregulated in Tgfbr3(-/-) epicardial cells which also show impaired cell invasion in response to ligand. TGFβR3-dependent cell invasion is also dependent upon Activin Receptor-Like Kinase (ALK) 2, ALK3, and ALK5 activity. A TGFβR3 mutant that contains a threonine to alanine substitution at residue 841 (TGFβR3-T841A) induces ligand-independent cell invasion in both epicardial and endocardial cells in vitro. These findings reveal a role for NF-κB signaling in the regulation of epicardial and endocardial cell invasion and identify a mutation in TGFβR3 which stimulates ligand-independent signaling.
Collapse
Affiliation(s)
- Cynthia R Clark
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States.
| | - Jamille Y Robinson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States.
| | - Nora S Sanchez
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States.
| | - Todd A Townsend
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States.
| | - Julian A Arrieta
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States.
| | - W David Merryman
- Dept. of Biomedical Engineering, Vanderbilt University, Nashville, TN 37212.
| | - David Z Trykall
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States.
| | - Harold E Olivey
- Dept. of Biology, Indiana University-Northwest, Gary, IN 46408, United States.
| | - Charles C Hong
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, United States; Research Medicine, Veterans Affairs TVHS, Nashville, TN 37212, United States.
| | - Joey V Barnett
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, United States; Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, United States.
| |
Collapse
|
199
|
Park-Windhol C, D'Amore PA. Disorders of Vascular Permeability. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2016; 11:251-81. [PMID: 26907525 DOI: 10.1146/annurev-pathol-012615-044506] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The endothelial barrier maintains vascular and tissue homeostasis and modulates many physiological processes, such as angiogenesis. Vascular barrier integrity can be disrupted by a variety of soluble permeability factors, and changes in barrier function can exacerbate tissue damage during disease progression. Understanding endothelial barrier function is critical for vascular homeostasis. Many of the signaling pathways promoting vascular permeability can also be triggered during disease, resulting in prolonged or uncontrolled vascular leak. It is believed that recovery of the normal vasculature requires diminishing this hyperpermeable state. Although the molecular mechanisms governing vascular leak have been studied over the last few decades, recent advances have identified new therapeutic targets that have begun to show preclinical and clinical promise. These approaches have been successfully applied to an increasing number of disease conditions. New perspectives regarding how vascular leak impacts the progression of various diseases are highlighted in this review.
Collapse
Affiliation(s)
- Cindy Park-Windhol
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts 02114; , .,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115
| | - Patricia A D'Amore
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts 02114; , .,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts 02115.,Department of Pathology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
200
|
Yan XC, Cao J, Liang L, Wang L, Gao F, Yang ZY, Duan JL, Chang TF, Deng SM, Liu Y, Dou GR, Zhang J, Zheng QJ, Zhang P, Han H. miR-342-5p Is a Notch Downstream Molecule and Regulates Multiple Angiogenic Pathways Including Notch, Vascular Endothelial Growth Factor and Transforming Growth Factor β Signaling. J Am Heart Assoc 2016; 5:JAHA.115.003042. [PMID: 26857067 PMCID: PMC4802463 DOI: 10.1161/jaha.115.003042] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Endothelial cells (ECs) form blood vessels through angiogenesis that is regulated by coordination of vascular endothelial growth factor (VEGF), Notch, transforming growth factor β, and other signals, but the detailed molecular mechanisms remain unclear. Methods and Results Small RNA sequencing initially identified miR‐342‐5p as a novel downstream molecule of Notch signaling in ECs. Reporter assay, quantitative reverse transcription polymerase chain reaction and Western blot analysis indicated that miR‐342‐5p targeted endoglin and modulated transforming growth factor β signaling by repressing SMAD1/5 phosphorylation in ECs. Transfection of miR‐342‐5p inhibited EC proliferation and lumen formation and reduced angiogenesis in vitro and in vivo, as assayed by using a fibrin beads–based sprouting assay, mouse aortic ring culture, and intravitreal injection of miR‐342‐5p agomir in P3 pups. Moreover, miR‐342‐5p promoted the migration of ECs, accompanied by reduced endothelial markers and increased mesenchymal markers, indicative of increased endothelial–mesenchymal transition. Transfection of endoglin at least partially reversed endothelial–mesenchymal transition induced by miR‐342‐5p. The expression of miR‐342‐5p was upregulated by transforming growth factor β, and inhibition of miR‐342‐5p attenuated the inhibitory effects of transforming growth factor β on lumen formation and sprouting by ECs. In addition, VEGF repressed miR‐342‐5p expression, and transfection of miR‐342‐5p repressed VEGFR2 and VEGFR3 expression and VEGF‐triggered Akt phosphorylation in ECs. miR‐342‐5p repressed angiogenesis in a laser‐induced choroidal neovascularization model in mice, highlighting its clinical potential. Conclusions miR‐342‐5p acts as a multifunctional angiogenic repressor mediating the effects and interaction among angiogenic pathways.
Collapse
Affiliation(s)
- Xian-Chun Yan
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jing Cao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Liang Liang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Li Wang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fang Gao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zi-Yan Yang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Juan-Li Duan
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tian-Fang Chang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - San-Ming Deng
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuan Liu
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Guo-Rui Dou
- Department of Ophthalmology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jian Zhang
- Department of Respiratory Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qi-Jun Zheng
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ping Zhang
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hua Han
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|