1
|
Rana R, Natoli TA, Khandelwal P, Pissios P, Muhammad AB, Chipashvili V, Farrington KP, Zhou W, Zheng G, Bukanov NO, Pocai A, Magnone MC. VEPTP inhibition with an extracellular domain targeting antibody did not restore albuminuria in a mouse model of diabetic kidney disease. Physiol Rep 2024; 12:e70058. [PMID: 39324545 PMCID: PMC11425269 DOI: 10.14814/phy2.70058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/30/2024] [Accepted: 09/08/2024] [Indexed: 09/27/2024] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage kidney disease. DKD is a heterogeneous disease with complex pathophysiology where early endothelial dysfunction is associated with disease progression. The Tie2 receptor and Angiopoietin 1 and 2 ligands are critical for maintaining endothelial cell permeability and integrity. Tie2 signaling is negatively regulated by the endothelial specific transmembrane receptor Vascular Endothelial Protein Tyrosine Phosphatase (VEPTP). Genetic deletion of VEPTP protects from hypertension and diabetes induced renal injury in a mouse model of DKD. Here, we show that VEPTP inhibition with an extracellular domain targeting VEPTP antibody induced Tie2 phosphorylation and improved VEGF-A induced vascular permeability both in vitro and in vivo. Treatment with the VEPTP blocking antibody decreased the renal expression of endothelial activation markers (Angpt2, Edn1, and Icam1) but failed to improve kidney function in db/db uninephrectomized ReninAAV DKD mice.
Collapse
Affiliation(s)
- Rajashree Rana
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentSpring HousePennsylvaniaUSA
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentCambridgeMassachusettsUSA
| | - Thomas A. Natoli
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentSpring HousePennsylvaniaUSA
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentCambridgeMassachusettsUSA
| | - Puneet Khandelwal
- Biologics Discovery, Johnson & Johnson Research & DevelopmentSpring HousePennsylvaniaUSA
| | - Pavlos Pissios
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentSpring HousePennsylvaniaUSA
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentCambridgeMassachusettsUSA
| | - Abdul Bari Muhammad
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentSpring HousePennsylvaniaUSA
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentCambridgeMassachusettsUSA
| | - Vaja Chipashvili
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentSpring HousePennsylvaniaUSA
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentCambridgeMassachusettsUSA
| | - Krista P. Farrington
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentSpring HousePennsylvaniaUSA
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentCambridgeMassachusettsUSA
| | - Wen Zhou
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentSpring HousePennsylvaniaUSA
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentCambridgeMassachusettsUSA
| | - Gang Zheng
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentSpring HousePennsylvaniaUSA
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentCambridgeMassachusettsUSA
| | - Nikolay O. Bukanov
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentSpring HousePennsylvaniaUSA
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentCambridgeMassachusettsUSA
| | - Alessandro Pocai
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentSpring HousePennsylvaniaUSA
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentCambridgeMassachusettsUSA
| | - Maria Chiara Magnone
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentSpring HousePennsylvaniaUSA
- Cardiovascular and Metabolism, Johnson & Johnson Research & DevelopmentCambridgeMassachusettsUSA
| |
Collapse
|
2
|
Zhou Y, Yao Z, Lin Y, Zhang H. From Tyrosine Kinases to Tyrosine Phosphatases: New Therapeutic Targets in Cancers and Beyond. Pharmaceutics 2024; 16:888. [PMID: 39065585 PMCID: PMC11279542 DOI: 10.3390/pharmaceutics16070888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/20/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs) regulate the level of tyrosine phosphorylation in proteins. PTKs are key enzymes that catalyze the transfer of an ATP phosphoric acid to a tyrosine residue on target protein substrates. Protein tyrosine phosphatases (PTPs) are responsible for the dephosphorylation of tyrosine residues and play a role in countering PTK overactivity. As widespread oncogenes, PTKs were once considered to be promising targets for therapy. However, tyrosine kinase inhibitors (TKIs) now face a number of challenges, including drug resistance and toxic side effects. Treatment strategies now need to be developed from a new perspective. In this review, we assess the current state of TKIs and highlight the role of PTPs in cancer and other diseases. With the advances of allosteric inhibition and the development of multiple alternative proprietary drug strategies, the reputation of PTPs as "undruggable" targets has been overturned, and they are now considered viable therapeutic targets. We also discuss the strategies and prospects of PTP-targeted therapy, as well as its future development.
Collapse
Affiliation(s)
- Yu Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou 510632, China; (Y.Z.); (Z.Y.); (Y.L.)
| | - Zhimeng Yao
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou 510632, China; (Y.Z.); (Z.Y.); (Y.L.)
- Department of Urology Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510660, China
| | - Yusheng Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou 510632, China; (Y.Z.); (Z.Y.); (Y.L.)
- Department of Thoracic Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510660, China
| | - Hao Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, MOE Key Laboratory of Tumor Molecular Biology, and Institute of Precision Cancer Medicine and Pathology, School of Medicine, Jinan University, Guangzhou 510632, China; (Y.Z.); (Z.Y.); (Y.L.)
- Department of Pathology, Gongli Hospital of Shanghai Pudong New Area, Shanghai 200135, China
- Zhuhai Institute of Jinan University, Zhuhai 511436, China
| |
Collapse
|
3
|
Fonódi M, Nagy L, Boratkó A. Role of Protein Phosphatases in Tumor Angiogenesis: Assessing PP1, PP2A, PP2B and PTPs Activity. Int J Mol Sci 2024; 25:6868. [PMID: 38999976 PMCID: PMC11241275 DOI: 10.3390/ijms25136868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Tumor angiogenesis, the formation of new blood vessels to support tumor growth and metastasis, is a complex process regulated by a multitude of signaling pathways. Dysregulation of signaling pathways involving protein kinases has been extensively studied, but the role of protein phosphatases in angiogenesis within the tumor microenvironment remains less explored. However, among angiogenic pathways, protein phosphatases play critical roles in modulating signaling cascades. This review provides a comprehensive overview of the involvement of protein phosphatases in tumor angiogenesis, highlighting their diverse functions and mechanisms of action. Protein phosphatases are key regulators of cellular signaling pathways by catalyzing the dephosphorylation of proteins, thereby modulating their activity and function. This review aims to assess the activity of the protein tyrosine phosphatases and serine/threonine phosphatases. These phosphatases exert their effects on angiogenic signaling pathways through various mechanisms, including direct dephosphorylation of angiogenic receptors and downstream signaling molecules. Moreover, protein phosphatases also crosstalk with other signaling pathways involved in angiogenesis, further emphasizing their significance in regulating tumor vascularization, including endothelial cell survival, sprouting, and vessel maturation. In conclusion, this review underscores the pivotal role of protein phosphatases in tumor angiogenesis and accentuate their potential as therapeutic targets for anti-angiogenic therapy in cancer.
Collapse
Affiliation(s)
| | | | - Anita Boratkó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (M.F.); (L.N.)
| |
Collapse
|
4
|
Brouillard P, Murtomäki A, Leppänen VM, Hyytiäinen M, Mestre S, Potier L, Boon LM, Revencu N, Greene A, Anisimov A, Salo MH, Hinttala R, Eklund L, Quéré I, Alitalo K, Vikkula M. Loss-of-function mutations of the TIE1 receptor tyrosine kinase cause late-onset primary lymphedema. J Clin Invest 2024; 134:e173586. [PMID: 38820174 PMCID: PMC11245153 DOI: 10.1172/jci173586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 05/24/2024] [Indexed: 06/02/2024] Open
Abstract
Primary lymphedema (PL), characterized by tissue swelling, fat accumulation, and fibrosis, results from defects in lymphatic vessels or valves caused by mutations in genes involved in development, maturation, and function of the lymphatic vascular system. Pathogenic variants in various genes have been identified in about 30% of PL cases. By screening of a cohort of 755 individuals with PL, we identified two TIE1 (tyrosine kinase with immunoglobulin- and epidermal growth factor-like domains 1) missense variants and one truncating variant, all predicted to be pathogenic by bioinformatic algorithms. The TIE1 receptor, in complex with TIE2, binds angiopoietins to regulate the formation and remodeling of blood and lymphatic vessels. The premature stop codon mutant encoded an inactive truncated extracellular TIE1 fragment with decreased mRNA stability, and the amino acid substitutions led to decreased TIE1 signaling activity. By reproducing the two missense variants in mouse Tie1 via CRISPR/Cas9, we showed that both cause edema and are lethal in homozygous mice. Thus, our results indicate that TIE1 loss-of-function variants can cause lymphatic dysfunction in patients. Together with our earlier demonstration that ANGPT2 loss-of-function mutations can also cause PL, our results emphasize the important role of the ANGPT2/TIE1 pathway in lymphatic function.
Collapse
Affiliation(s)
- Pascal Brouillard
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium
| | - Aino Murtomäki
- Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Veli-Matti Leppänen
- Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Marko Hyytiäinen
- Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sandrine Mestre
- Department of Vascular Medicine, Centre de Référence des Maladies Lymphatiques et Vasculaires Rares, Inserm IDESP, CHU Montpellier, Université de Montpellier, Montpellier, France
| | - Lucas Potier
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium
| | - Laurence M. Boon
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium
- Center for Vascular Anomalies, Division of Plastic Surgery, Cliniques Universitaires Saint-Luc, University of Louvain, VASCERN-VASCA Reference Centre, Brussels, Belgium
| | - Nicole Revencu
- Center for Human Genetics, Cliniques Universitaires Saint-Luc, University of Louvain, Brussels, Belgium
| | - Arin Greene
- Department of Plastic and Oral Surgery, Lymphedema Program, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrey Anisimov
- Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Miia H. Salo
- Biocenter Oulu, Research Unit of Clinical Medicine and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Reetta Hinttala
- Biocenter Oulu, Research Unit of Clinical Medicine and Medical Research Center Oulu, University of Oulu and Oulu University Hospital, Oulu, Finland
| | - Lauri Eklund
- Oulu Center for Cell-Matrix Research, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Isabelle Quéré
- Department of Vascular Medicine, Centre de Référence des Maladies Lymphatiques et Vasculaires Rares, Inserm IDESP, CHU Montpellier, Université de Montpellier, Montpellier, France
| | - Kari Alitalo
- Wihuri Research Institute, Biomedicum Helsinki, Helsinki, Finland
- Translational Cancer Medicine Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, University of Louvain, Brussels, Belgium
- WELBIO department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
5
|
Vemuri K, de Alves Pereira B, Fuenzalida P, Subashi Y, Barbera S, van Hooren L, Hedlund M, Pontén F, Lindskog C, Olsson AK, Lugano R, Dimberg A. CD93 maintains endothelial barrier function and limits metastatic dissemination. JCI Insight 2024; 9:e169830. [PMID: 38441970 PMCID: PMC11128212 DOI: 10.1172/jci.insight.169830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 02/27/2024] [Indexed: 03/07/2024] Open
Abstract
Compromised vascular integrity facilitates extravasation of cancer cells and promotes metastatic dissemination. CD93 has emerged as a target for antiangiogenic therapy, but its importance for vascular integrity in metastatic cancers has not been evaluated. Here, we demonstrate that CD93 participates in maintaining the endothelial barrier and reducing metastatic dissemination. Primary melanoma growth was hampered in CD93-/- mice, but metastatic dissemination was increased and associated with disruption of adherens and tight junctions in tumor endothelial cells and elevated expression of matrix metalloprotease 9 at the metastatic site. CD93 directly interacted with vascular endothelial growth factor receptor 2 (VEGFR2) and its absence led to VEGF-induced hyperphosphorylation of VEGFR2 in endothelial cells. Antagonistic anti-VEGFR2 antibody therapy rescued endothelial barrier function and reduced the metastatic burden in CD93-/- mice to wild-type levels. These findings reveal a key role of CD93 in maintaining vascular integrity, which has implications for pathological angiogenesis and endothelial barrier function in metastatic cancer.
Collapse
Affiliation(s)
- Kalyani Vemuri
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, and
| | - Beatriz de Alves Pereira
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, and
| | - Patricia Fuenzalida
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, and
| | - Yelin Subashi
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, and
| | - Stefano Barbera
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, and
| | - Luuk van Hooren
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, and
| | - Marie Hedlund
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, and
| | - Fredrik Pontén
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, and
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, and
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Uppsala University Biomedical Center, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Roberta Lugano
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, and
| | - Anna Dimberg
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Science for Life Laboratory, and
| |
Collapse
|
6
|
Baluk P, Shirakura K, Vestweber D, McDonald DM. Heterogeneity of endothelial VE-PTP downstream polarization, Tie2 activation, junctional claudin-5, and permeability in the aorta and vena cava. Cell Tissue Res 2024; 395:81-103. [PMID: 38032480 PMCID: PMC10774230 DOI: 10.1007/s00441-023-03844-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/10/2023] [Indexed: 12/01/2023]
Abstract
Endothelial cells of mammalian blood vessels have multiple levels of heterogeneity along the vascular tree and among different organs. Further heterogeneity results from blood flow turbulence and variations in shear stress. In the aorta, vascular endothelial protein tyrosine phosphatase (VE-PTP), which dephosphorylates tyrosine kinase receptor Tie2 in the plasma membrane, undergoes downstream polarization and endocytosis in endothelial cells exposed to laminar flow and high shear stress. VE-PTP sequestration promotes Tie2 phosphorylation at tyrosine992 and endothelial barrier tightening. The present study characterized the heterogeneity of VE-PTP polarization, Tie2-pY992 and total Tie2, and claudin-5 in anatomically defined regions of endothelial cells in the mouse descending thoracic aorta, where laminar flow is variable and IgG extravasation is patchy. We discovered that VE-PTP and Tie2-pY992 had mosaic patterns, unlike the uniform distribution of total Tie2. Claudin-5 at tight junctions also had a mosaic pattern, whereas VE-cadherin at adherens junctions bordered all endothelial cells. Importantly, the amounts of Tie2-pY992 and claudin-5 in aortic endothelial cells correlated with downstream polarization of VE-PTP. VE-PTP and Tie2-pY992 also had mosaic patterns in the vena cava, but claudin-5 was nearly absent and extravasated IgG was ubiquitous. Correlation of Tie2-pY992 and claudin-5 with VE-PTP polarization supports their collective interaction in the regulation of endothelial barrier function in the aorta, yet differences between the aorta and vena cava indicate additional flow-related determinants of permeability. Together, the results highlight new levels of endothelial cell functional mosaicism in the aorta and vena cava, where blood flow dynamics are well known to be heterogeneous.
Collapse
Affiliation(s)
- Peter Baluk
- Department of Anatomy, Cardiovascular Research Institute, and UCSF Helen Diller Family Comprehensive Cancer Center, University of California, 513 Parnassus Avenue, Room S1349, San Francisco, CA, 94143-0452, USA
| | - Keisuke Shirakura
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, Münster, 48149, Germany
| | - Dietmar Vestweber
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, Münster, 48149, Germany
| | - Donald M McDonald
- Department of Anatomy, Cardiovascular Research Institute, and UCSF Helen Diller Family Comprehensive Cancer Center, University of California, 513 Parnassus Avenue, Room S1349, San Francisco, CA, 94143-0452, USA.
| |
Collapse
|
7
|
Wang Y, Halawa M, Chatterjee A, Eshwaran R, Qiu Y, Wibowo YC, Pan J, Wieland T, Feng Y. Sufficient Cav-1 levels in the endothelium are critical for the maintenance of the neurovascular unit in the retina. Mol Med 2023; 29:152. [PMID: 37923999 PMCID: PMC10623831 DOI: 10.1186/s10020-023-00749-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Caveolin-1 (Cav-1) is a pivotal protein in the plasma membrane. Studies on homozygous Cav-1 deficient mice revealed that Cav-1 is essential for endothelial function and angiogenesis in the retina. However, whether a reduction in Cav-1 content hampers the neurovascular unit (NVU) in the retina is unclear. Thus, this study examines the NVU in the retinas of heterozygous Cav-1 deficient (Cav-1+/-) mice and analyzes possible underlying mechanisms. METHODS The vascular, glial and neuronal components in the retina were evaluated using retinal morphometry, whole mount retinal immunofluorescence staining, histological analysis and optical coherence tomography. In addition, immunoblotting and immunofluorescence staining, subcellular fractionation, biotin labeling of cell surface proteins, and proximity ligation assay were employed to detect expression and localization of proteins in the retina or endothelial cells (ECs) upon knockdown of Cav-1 with Cav-1 siRNA. RESULTS Cav-1+/- retinas showed a significant reduction in pericyte coverage along with an increase in acellular capillaries compared to controls at 8 months of age, but not at 1 month. A significant loss and obvious morphological abnormalities of smooth muscle cells were observed in 8-month-old Cav-1+/- retinal arterioles. Macroglial and microglial cells were activated in the Cav-1+/- retinas. A transient significant delay in retinal angiogenesis was detected in Cav-1+/- retinas at p5, which was however no longer detectable at p10. The Cav-1+/- retinas displayed increased vascular permeability and a notable reduction in VEGFR2 content at 8 months. In vitro, siRNA-mediated knockdown experiments in ECs revealed that the loss of Cav-1 in ECs resulted in decreased levels of VEGFR2, VE-Cadherin and their interaction at the plasma membrane as well. CONCLUSION Our results indicate that a sufficient Cav-1 level over 50% of its normal abundance is vital for the proper localization of VEGFR2 and VE-cadherin, likely in a complex, at the plasma membrane, which is essential for the maintenance of normal NVU in the retina.
Collapse
Affiliation(s)
- Yixin Wang
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Mahmoud Halawa
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Anupriya Chatterjee
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Rachana Eshwaran
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Yi Qiu
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Yohanes Cakrapradipta Wibowo
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Jianyuan Pan
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
| | - Thomas Wieland
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Yuxi Feng
- Experimental Pharmacology Mannheim, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Ludolf-Krehl-Str. 13-17, 68167, Mannheim, Germany.
| |
Collapse
|
8
|
Nan W, He Y, Wang S, Zhang Y. Molecular mechanism of VE-cadherin in regulating endothelial cell behaviour during angiogenesis. Front Physiol 2023; 14:1234104. [PMID: 37601629 PMCID: PMC10433914 DOI: 10.3389/fphys.2023.1234104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
Vascular endothelial (VE)-cadherin, an endothelium-specific adhesion protein, is found in the junctions between endothelial cells (ECs). It's crucial to maintain the homogeneity of ECs. Keeping and controlling the contact between ECs is essential. In addition to its adhesive function, VE-cadherin plays important roles in vascular development, permeability, and tumour angiogenesis. Signal transfer, cytoskeletal reconstruction, and contractile integrating, which are crucial for constructing and maintaining monolayer integrity as well as for repair and regeneration, are the foundation of endothelial cell (EC) junctional dynamics. The molecular basis of adhesion junctions (AJs), which are closely related and work with actin filaments, is provided by the VE-cadherin-catenin complex. They can activate intracellular signals that drive ECs to react or communicate structural changes to junctions. An increasing number of molecules, including the vascular endothelial growth factor receptor 2 (VEGFR2) and vascular endothelial protein tyrosine phosphatase (VE-PTP), have been connected to VE-cadherin in addition to the conventional VE-cadherin-catenin complex. This review demonstrates significant progress in our understanding of the molecular mechanisms that affect VE-cadherin's function in the regulation of EC behaviour during angiogenesis. The knowledge of the molecular processes that control VE-cadherin's role in the regulation of EC behaviour during angiogenesis has recently advanced, as shown in this review.
Collapse
Affiliation(s)
- Weijin Nan
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Yuxi He
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
| | - Shurong Wang
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yan Zhang
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun, China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Qiu F, Jiang B, Lin Y, Li H, Li D, Luo M, Hui H, Miao H, Zhang Y. Dual Pigment Epithelium-derived Factor and Hepatocyte Growth Factor Overexpression: A New Therapy for Pulmonary Hypertension. Am J Respir Cell Mol Biol 2023; 69:87-98. [PMID: 37094101 DOI: 10.1165/rcmb.2022-0459oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/24/2023] [Indexed: 04/26/2023] Open
Abstract
Pulmonary hypertension (PH) is a disease characterized by advanced pulmonary vasculature remodeling that is thought to be curable only through lung transplantation. The application of angiogenic hepatocyte growth factor (HGF) is reported to be protective in PH through its anti-vascular remodeling effect, but excessive HGF-mediated immature neovascularization is not conducive to the restoration of pulmonary perfusion because of apparent vascular leakage. As a canonical antiangiogenic molecule, pigment epithelium-derived factor (PEDF) inhibits angiogenesis and reduces vascular permeability in a variety of diseases. However, the effect of PEDF on HGF-based PH treatment remains to be determined. In this study, monocrotaline-induced PH rats and endothelial cells isolated from rat and human PH lung tissues were used. We assessed PH progression, right cardiac function, and pulmonary perfusion in HGF- and/or PEDF-treated rats with PH. Additionally, the receptor and mechanism responsible for the role of PEDF in HGF-based PH therapy were investigated. In this study, we found that HGF and PEDF jointly prevent PH development and improve right cardiac function in rats with PH. Moreover, PEDF delivery increases the pulmonary perfusion in PH lungs and inhibits immature angiogenesis and vascular endothelial (VE)-cadherin junction disintegration induced by HGF without affecting the therapeutic inhibition of pulmonary vascular remodeling by HGF. Mechanistically, PEDF targets VE growth factor receptor 2 and suppresses its phosphorylation at Y951 and Y1175 but not Y1214. Finally, VE growth factor receptor 2/VE protein tyrosine phosphatase/VE-cadherin complex formation and Akt and Erk1/2 inactivation were observed in rat and human PH lung endothelial cells. Collectively, our data indicate that PEDF additively enhances the efficacy of HGF against PH, which may provide new insights into treatment strategies for clinical PH.
Collapse
Affiliation(s)
- Fan Qiu
- Department of Thoracic Cardiovascular Surgery and
| | - Bo Jiang
- Department of Thoracic Cardiovascular Surgery and
| | - Yangui Lin
- Department of Thoracic Cardiovascular Surgery and
| | - Huaming Li
- Department of Thoracic Cardiovascular Surgery and
| | - Dan Li
- Community Health Center, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Min Luo
- Department of Thoracic Cardiovascular Surgery and
| | | | - Haoran Miao
- Department of Thoracic Cardiovascular Surgery and
| | - Yiqian Zhang
- Department of Thoracic Cardiovascular Surgery and
| |
Collapse
|
10
|
Li Y, Liu P, Zhou Y, Maekawa H, Silva JB, Ansari MJ, Boubes K, Alia Y, Deb DK, Thomson BR, Jin J, Quaggin SE. Activation of Angiopoietin-Tie2 Signaling Protects the Kidney from Ischemic Injury by Modulation of Endothelial-Specific Pathways. J Am Soc Nephrol 2023; 34:969-987. [PMID: 36787763 PMCID: PMC10278803 DOI: 10.1681/asn.0000000000000098] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 01/19/2023] [Indexed: 02/16/2023] Open
Abstract
SIGNIFICANCE STATEMENT Ischemia-reperfusion AKI (IR-AKI) is common and causes significant morbidity. Effective treatments are lacking. However, preclinical studies suggest that inhibition of angiopoietin-Tie2 vascular signaling promotes injury, whereas activation of Tie2 is protective. We show that kidney ischemia leads to increased levels of the endothelial-specific phosphatase vascular endothelial protein tyrosine phosphatase (VE-PTP; PTPRB), which inactivates Tie2. Activation of Tie2 through VE-PTP deletion, or delivery of a novel angiopoietin mimetic (Hepta-ANG1), abrogated IR-AKI in mice. Single-cell RNAseq analysis showed Tie2 activation promotes increased Entpd1 expression, downregulation of FOXO1 target genes in the kidney vasculature, and emergence of a new subpopulation of glomerular endothelial cells. Our data provide a molecular basis and identify a candidate therapeutic to improve endothelial integrity and kidney function after IR-AKI. BACKGROUND Ischemia-reperfusion AKI (IR-AKI) is estimated to affect 2%-7% of all hospitalized patients. The significant morbidity and mortality associated with AKI indicates urgent need for effective treatments. Previous studies have shown activation of the vascular angiopoietin-Tie2 tyrosine kinase signaling pathway abrogates ischemia-reperfusion injury (IRI). We extended previous studies to (1) determine the molecular mechanism(s) underlying kidney injury and protection related to decreased or increased activation of Tie2, respectively, and (2) to test the hypothesis that deletion of the Tie2 inhibitory phosphatase vascular endothelial protein tyrosine phosphatase (VE-PTP) or injection of a new angiopoietin mimetic protects the kidney from IRI by common molecular mechanism(s). METHODS Bilateral IR-AKI was performed in VE-PTP wild-type or knockout mice and in C57BL/6J mice treated with Hepta-ANG1 or vehicle. Histologic, immunostaining, and single-cell RNA sequencing analyses were performed. RESULTS The phosphatase VE-PTP, which negatively regulates the angiopoietin-Tie2 pathway, was upregulated in kidney endothelial cells after IRI, and genetic deletion of VE-PTP in mice protected the kidney from IR-AKI. Injection of Hepta-ANG1 potently activated Tie2 and protected the mouse kidney from IRI. Single-cell RNAseq analysis of kidneys from Hepta-ANG1-treated and vehicle-treated mice identified endothelial-specific gene signatures and emergence of a new glomerular endothelial subpopulation associated with improved kidney function. Overlap was found between endothelial-specific genes upregulated by Hepta-ANG1 treatment and those downregulated in HUVECs with constitutive FOXO1 activation, including Entpd1 / ENTPD1 that modulates purinergic receptor signaling. CONCLUSIONS Our data support a key role of the endothelium in the development of IR-AKI, introduce Hepta-ANG1 as a putative new therapeutic biologic, and report a model to explain how IRI reduces Tie2 signaling and how Tie2 activation protects the kidney. PODCAST This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/JASN/2023_05_23_JSN_Ang_EP23_052323.mp3.
Collapse
Affiliation(s)
- Yanyang Li
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- The Feinberg Cardiovascular and Renal Research Institute, Chicago, Illinois
| | - Pan Liu
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- The Feinberg Cardiovascular and Renal Research Institute, Chicago, Illinois
| | - Yalu Zhou
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- The Feinberg Cardiovascular and Renal Research Institute, Chicago, Illinois
| | - Hiroshi Maekawa
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - John B. Silva
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Mohammed Javeed Ansari
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Khaled Boubes
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Yazan Alia
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Dilip K. Deb
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | | | - Jing Jin
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- The Feinberg Cardiovascular and Renal Research Institute, Chicago, Illinois
| | - Susan E. Quaggin
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- The Feinberg Cardiovascular and Renal Research Institute, Chicago, Illinois
| |
Collapse
|
11
|
Beter M, Abdollahzadeh A, Pulkkinen HH, Huang H, Orsenigo F, Magnusson PU, Ylä-Herttuala S, Tohka J, Laakkonen JP. SproutAngio: an open-source bioimage informatics tool for quantitative analysis of sprouting angiogenesis and lumen space. Sci Rep 2023; 13:7279. [PMID: 37142637 PMCID: PMC10160097 DOI: 10.1038/s41598-023-33090-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 04/06/2023] [Indexed: 05/06/2023] Open
Abstract
Three-dimensional image analyses are required to improve the understanding of the regulation of blood vessel formation and heterogeneity. Currently, quantitation of 3D endothelial structures or vessel branches is often based on 2D projections of the images losing their volumetric information. Here, we developed SproutAngio, a Python-based open-source tool, for fully automated 3D segmentation and analysis of endothelial lumen space and sprout morphology. To test the SproutAngio, we produced a publicly available in vitro fibrin bead assay dataset with a gradually increasing VEGF-A concentration ( https://doi.org/10.5281/zenodo.7240927 ). We demonstrate that our automated segmentation and sprout morphology analysis, including sprout number, length, and nuclei number, outperform the widely used ImageJ plugin. We also show that SproutAngio allows a more detailed and automated analysis of the mouse retinal vasculature in comparison to the commonly used radial expansion measurement. In addition, we provide two novel methods for automated analysis of endothelial lumen space: (1) width measurement from tip, stalk and root segments of the sprouts and (2) paired nuclei distance analysis. We show that these automated methods provided important additional information on the endothelial cell organization in the sprouts. The pipelines and source code of SproutAngio are publicly available ( https://doi.org/10.5281/zenodo.7381732 ).
Collapse
Affiliation(s)
- M Beter
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, P.O.Box 1627, 70211, Kuopio, Finland
| | - A Abdollahzadeh
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, P.O.Box 1627, 70211, Kuopio, Finland
| | - H H Pulkkinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, P.O.Box 1627, 70211, Kuopio, Finland
| | - H Huang
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - F Orsenigo
- Vascular Biology Unit, IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy
| | - P U Magnusson
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - S Ylä-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, P.O.Box 1627, 70211, Kuopio, Finland
- Heart Center, Kuopio University Hospital, Kuopio, Finland
- Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland
| | - J Tohka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, P.O.Box 1627, 70211, Kuopio, Finland
| | - J P Laakkonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neulaniementie 2, P.O.Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
12
|
Lee E, Lee EA, Kong E, Chon H, Llaiqui-Condori M, Park CH, Park BY, Kang NR, Yoo JS, Lee HS, Kim HS, Park SH, Choi SW, Vestweber D, Lee JH, Kim P, Lee WS, Kim I. An agonistic anti-Tie2 antibody suppresses the normal-to-tumor vascular transition in the glioblastoma invasion zone. Exp Mol Med 2023; 55:470-484. [PMID: 36828931 PMCID: PMC9981882 DOI: 10.1038/s12276-023-00939-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 02/26/2023] Open
Abstract
Tumor progression is intimately associated with the vasculature, as tumor proliferation induces angiogenesis and tumor cells metastasize to distant organs via blood vessels. However, whether tumor invasion is associated with blood vessels remains unknown. As glioblastoma (GBM) is featured by aggressive invasion and vascular abnormalities, we characterized the onset of vascular remodeling in the diffuse tumor infiltrating zone by establishing new spontaneous GBM models with robust invasion capacity. Normal brain vessels underwent a gradual transition to severely impaired tumor vessels at the GBM periphery over several days. Increasing vasodilation from the tumor periphery to the tumor core was also found in human GBM. The levels of vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2) showed a spatial correlation with the extent of vascular abnormalities spanning the tumor-invading zone. Blockade of VEGFR2 suppressed vascular remodeling at the tumor periphery, confirming the role of VEGF-VEGFR2 signaling in the invasion-associated vascular transition. As angiopoietin-2 (ANGPT2) was expressed in only a portion of the central tumor vessels, we developed a ligand-independent tunica interna endothelial cell kinase 2 (Tie2)-activating antibody that can result in Tie2 phosphorylation in vivo. This agonistic anti-Tie2 antibody effectively normalized the vasculature in both the tumor periphery and tumor center, similar to the effects of VEGFR2 blockade. Mechanistically, this antibody-based Tie2 activation induced VE-PTP-mediated VEGFR2 dephosphorylation in vivo. Thus, our study reveals that the normal-to-tumor vascular transition is spatiotemporally associated with GBM invasion and may be controlled by Tie2 activation via a novel mechanism of action.
Collapse
Affiliation(s)
- Eunhyeong Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Eun-Ah Lee
- R&D Center, PharmAbcine Inc., Daejeon, 34047, Republic of Korea
| | - Eunji Kong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Haemin Chon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Melissa Llaiqui-Condori
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Cheon Ho Park
- R&D Center, PharmAbcine Inc., Daejeon, 34047, Republic of Korea
| | - Beom Yong Park
- R&D Center, PharmAbcine Inc., Daejeon, 34047, Republic of Korea
| | - Nu Ri Kang
- R&D Center, PharmAbcine Inc., Daejeon, 34047, Republic of Korea
| | - Jin-San Yoo
- R&D Center, PharmAbcine Inc., Daejeon, 34047, Republic of Korea
| | - Hyun-Soo Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, 61463, Republic of Korea
| | - Sung-Hong Park
- Department of Bio and Brain Engineering, KAIST, Daejeon, 34141, Republic of Korea
| | - Seung-Won Choi
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Dietmar Vestweber
- Max Planck Institute for Molecular Biomedicine, D-48149, Muenster, Germany
| | - Jeong Ho Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.,BioMedical Research Center, KAIST, Daejeon, 34141, Republic of Korea.,SoVarGen, Inc., Daejeon, 34051, Republic of Korea
| | - Pilhan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea.,Graduate School of Nanoscience and Technology, Daejeon, 34141, Republic of Korea.,KI for Health Science and Technology, KAIST, Daejeon, 34141, Republic of Korea
| | - Weon Sup Lee
- R&D Center, PharmAbcine Inc., Daejeon, 34047, Republic of Korea.
| | - Injune Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea. .,BioMedical Research Center, KAIST, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
13
|
Zhou L, Zhang W, Xiang Y, Qian Z, Zhou J, Ni L, Feng Y, Gao B. The apatinib and pemetrexed combination has antitumor and antiangiogenic effects against NSCLC. Open Life Sci 2023; 18:20220533. [PMID: 36910471 PMCID: PMC9993329 DOI: 10.1515/biol-2022-0533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 05/17/2022] [Accepted: 11/02/2022] [Indexed: 03/09/2023] Open
Abstract
Chemotherapy for advanced non-small-cell lung cancer (NSCLC) remains the first treatment choice. Angiogenesis inhibitors are effective for lung cancer treatment. This study explored whether chemotherapy combined with angiogenesis inhibitors could achieve better efficacy in NSCLC. The zebrafish A549 xenograft model was used to investigate the combined effect of apatinib and chemotherapeutic agents in NSCLC. Apatinib combined with pemetrexed demonstrated the highest antitumor effect compared with apatinib combined with gemcitabine or paclitaxel in vitro. In the zebrafish A549 xenograft model, apatinib and pemetrexed, alone or in combination, showed significant inhibition of tumor growth. Co-treatment with apatinib and pemetrexed demonstrated the best antitumor effects, suggesting that the combination of apatinib and pemetrexed might be a promising alternative therapy for patients with lung cancer. Apatinib combined with pemetrexed had enhanced antitumor effects compared with either one alone in the zebrafish model of NSCLC.
Collapse
Affiliation(s)
- Ling Zhou
- Department of Respiratory Medicine, Rui Jin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Wenchao Zhang
- Department of Allergy, Henan Provincial People's Hospital, Henan University, Zhengzhou 450000, China
| | - Yi Xiang
- Department of Respiratory Medicine, Rui Jin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Zijun Qian
- Department of Respiratory Medicine, Rui Jin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Jianping Zhou
- Department of Respiratory Medicine, Rui Jin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Lei Ni
- Department of Respiratory Medicine, Rui Jin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Yun Feng
- Department of Respiratory Medicine, Rui Jin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| | - Beili Gao
- Department of Respiratory Medicine, Rui Jin Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai 200025, China
| |
Collapse
|
14
|
Luxen M, van Meurs M, Molema G. Unlocking the Untapped Potential of Endothelial Kinase and Phosphatase Involvement in Sepsis for Drug Treatment Design. Front Immunol 2022; 13:867625. [PMID: 35634305 PMCID: PMC9136877 DOI: 10.3389/fimmu.2022.867625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Abstract
Sepsis is a devastating clinical condition that can lead to multiple organ failure and death. Despite advancements in our understanding of molecular mechanisms underlying sepsis and sepsis-associated multiple organ failure, no effective therapeutic treatment to directly counteract it has yet been established. The endothelium is considered to play an important role in sepsis. This review highlights a number of signal transduction pathways involved in endothelial inflammatory activation and dysregulated endothelial barrier function in response to sepsis conditions. Within these pathways – NF-κB, Rac1/RhoA GTPases, AP-1, APC/S1P, Angpt/Tie2, and VEGF/VEGFR2 – we focus on the role of kinases and phosphatases as potential druggable targets for therapeutic intervention. Animal studies and clinical trials that have been conducted for this purpose are discussed, highlighting reasons why they might not have resulted in the expected outcomes, and which lessons can be learned from this. Lastly, opportunities and challenges that sepsis and sepsis-associated multiple organ failure research are currently facing are presented, including recommendations on improved experimental design to increase the translational power of preclinical research to the clinic.
Collapse
Affiliation(s)
- Matthijs Luxen
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- *Correspondence: Matthijs Luxen,
| | - Matijs van Meurs
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Critical Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Grietje Molema
- Department of Pathology and Medical Biology, Medical Biology Section, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
15
|
Corti F, Ristori E, Rivera-Molina F, Toomre D, Zhang J, Mihailovic J, Zhuang ZW, Simons M. Syndecan-2 selectively regulates VEGF-induced vascular permeability. NATURE CARDIOVASCULAR RESEARCH 2022; 1:518-528. [PMID: 36212522 PMCID: PMC9544384 DOI: 10.1038/s44161-022-00064-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 04/06/2022] [Indexed: 02/03/2023]
Abstract
Vascular endothelial growth factor (VEGF)- driven increase in vascular permeability is a key feature of many disease states associated with inflammation and ischemic injury, contributing significantly to morbidity and mortality in these settings. Despite its importance, no specific regulators that preferentially control VEGF-dependent increase in permeability versus its other biological activities, have been identified. Here we report that a proteoglycan Syndecan-2 (Sdc2) regulates the interaction between a transmembrane phosphatase DEP1 and VEGFR2 by controlling cell surface levels of DEP1. In the absence of Sdc2 or the presence of an antibody that blocks Sdc2-DEP1 interaction, increased plasma membrane DEP1 levels promote selective dephosphorylation of the VEGFR2 Y951 site that is involved in permeability control. Either an endothelial-specific Sdc2 deletion or a treatment with an anti-Sdc2 antibody result in a highly significant reduction in stroke size due to a decrease in intracerebral edema.
Collapse
Affiliation(s)
- F Corti
- Yale Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - E Ristori
- Yale Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - F Rivera-Molina
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - D Toomre
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - J Zhang
- Yale Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - J Mihailovic
- Department of Radiology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Z W Zhuang
- Yale Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| | - M Simons
- Yale Cardiovascular Research Center Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06511, USA
| |
Collapse
|
16
|
βIV-spectrin as a stalk cell-intrinsic regulator of VEGF signaling. Nat Commun 2022; 13:1326. [PMID: 35288568 PMCID: PMC8921520 DOI: 10.1038/s41467-022-28933-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 02/14/2022] [Indexed: 11/25/2022] Open
Abstract
Defective angiogenesis underlies over 50 malignant, ischemic and inflammatory disorders yet long-term therapeutic applications inevitably fail, thus highlighting the need for greater understanding of the vast crosstalk and compensatory mechanisms. Based on proteomic profiling of angiogenic endothelial components, here we report βIV-spectrin, a non-erythrocytic cytoskeletal protein, as a critical regulator of sprouting angiogenesis. Early loss of endothelial-specific βIV-spectrin promotes embryonic lethality in mice due to hypervascularization and hemorrhagic defects whereas neonatal depletion yields higher vascular density and tip cell populations in developing retina. During sprouting, βIV-spectrin expresses in stalk cells to inhibit their tip cell potential by enhancing VEGFR2 turnover in a manner independent of most cell-fate determining mechanisms. Rather, βIV-spectrin recruits CaMKII to the plasma membrane to directly phosphorylate VEGFR2 at Ser984, a previously undefined phosphoregulatory site that strongly induces VEGFR2 internalization and degradation. These findings support a distinct spectrin-based mechanism of tip-stalk cell specification during vascular development. Defective angiogenesis remains a high source of morbidity in multiple disorders. Here they show that βIV-spectrin, a membrane-associated cytoskeletal protein, is essential for regulation of endothelial tip cell populations and VEGF signaling during sprouting angiogenesis.
Collapse
|
17
|
Waters SB, Dominguez JR, Cho HD, Sarich NA, Malik AB, Yamada KH. KIF13B-mediated VEGFR2 trafficking is essential for vascular leakage and metastasis in vivo. Life Sci Alliance 2022; 5:e202101170. [PMID: 34670814 PMCID: PMC8548263 DOI: 10.26508/lsa.202101170] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 11/24/2022] Open
Abstract
VEGF-A induces vascular leakage and angiogenesis via activating the cell surface localized receptor VEGF receptor 2 (VEGFR2). The amount of available VEGFR2 at the cell surface is however tightly regulated by trafficking of VEGFR2 by kinesin family 13 B (KIF13B), a plus-end kinesin motor, to the plasma membrane of endothelial cells (ECs). Competitive inhibition of interaction between VEGFR2 and KIF13B by a peptide kinesin-derived angiogenesis inhibitor (KAI) prevented pathological angiogenesis in models of cancer and eye disease associated with defective angiogenesis. Here, we show the protective effects of KAI in VEGF-A-induced vascular leakage and cancer metastasis. Using an EC-specific KIF13B knockout (Kif13b iECKO ) mouse model, we demonstrated the function of EC expressed KIF13B in mediating VEGF-A-induced vascular leakage, angiogenesis, tumor growth, and cancer metastasis. Thus, KIF13B-mediated trafficking of VEGFR2 to the endothelial surface has an essential role in pathological angiogenesis induced by VEGF-A, and is therefore a potential therapeutic target.
Collapse
Affiliation(s)
- Stephen B Waters
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Joseph R Dominguez
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Hyun-Dong Cho
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Nicolene A Sarich
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Asrar B Malik
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
| | - Kaori H Yamada
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL, USA
- Department of Ophthalmology and Visual Sciences, University of Illinois College of Medicine, Chicago, IL, USA
| |
Collapse
|
18
|
Wang S, Zhou Z, Li J, Wang Y, Li H, Lv R, Xu G, Zhang J, Bi J, Huo R. Identification of ACTA2 as a Key Contributor to Venous Malformation. Front Cell Dev Biol 2021; 9:755409. [PMID: 34858981 PMCID: PMC8630574 DOI: 10.3389/fcell.2021.755409] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 09/29/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives: Proteomics and high connotation functional gene screening (HCS) were used to screen key functional genes that play important roles in the pathogenesis of venous malformation. Furthermore, this study was conducted to analyze and explore their possible functions, establish a gene mutation zebrafish model, and perform a preliminary study to explore their possible pathogenic mechanisms in venous malformation. Methods: Pathological and normal tissues from patients with disseminated venous malformation were selected for Tandem Mass Tag (TMT) proteomics analysis to identify proteins that were differentially expressed. Based on bioinformatics analysis, 20 proteins with significant differential expression were selected for HCS to find key driver genes and characterize the expression of these genes in patients with venous malformations. In vitro experiments were then performed using human microvascular endothelial cells (HMEC-1). A gene mutant zebrafish model was also constructed for in vivo experiments to explore gene functions and pathogenic mechanisms. Results: The TMT results showed a total of 71 proteins that were differentially expressed as required, with five of them upregulated and 66 downregulated. Based on bioinformatics and proteomics results, five highly expressed genes and 15 poorly expressed genes were selected for functional screening by RNAi technology. HCS screening identified ACTA2 as the driver gene. Quantitative polymerase chain reaction (qPCR) and western blot were used to detect the expression of ACTA2 in the pathological tissues of patients with venous malformations and in control tissues, and the experimental results showed a significantly lower expression of ACTA2 in venous malformation tissues (P < 0.05). Cell assays on the human microvascular endothelial cells (HMEC-1) model showed that cell proliferation, migration, invasion, and angiogenic ability were all significantly increased in the ACTA2 over-expression group (P < 0.05), and that overexpression of ACTA2 could improve the inhibitory effect on vascular endothelial cell proliferation. We constructed an ACTA2-knockdown zebrafish model and found that the knockdown of ACTA2 resulted in defective vascular development, disruption of vascular integrity, and malformation of micro vein development in zebrafish. Further qPCR assays revealed that the knockdown of ACTA2 inhibited the Dll4/notch1 signaling pathway, Ephrin-B2 signaling pathway, and vascular integrity-related molecules and activated the Hedgehog signaling pathway. Conclusion: This study revealed that ACTA2 deficiency is an important factor in the pathogenesis of venous malformation, resulting in the disruption of vascular integrity and malformed vascular development. ACTA2 can be used as a potential biomarker for the treatment and prognosis of venous malformations.
Collapse
Affiliation(s)
- Song Wang
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zifu Zhou
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jing Li
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu Wang
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hongwen Li
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Renrong Lv
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guangqi Xu
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jian Zhang
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianhai Bi
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ran Huo
- Department of Burn and Plastic Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Burn and Plastic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
19
|
Luo H, Zhang Y, Deng Y, Li L, Sheng Z, Yu Y, Lin Y, Chen X, Feng P. Nxhl Controls Angiogenesis by Targeting VE-PTP Through Interaction With Nucleolin. Front Cell Dev Biol 2021; 9:728821. [PMID: 34733844 PMCID: PMC8558974 DOI: 10.3389/fcell.2021.728821] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/08/2021] [Indexed: 11/13/2022] Open
Abstract
Precise regulation of angiogenesis is required for organ development, wound repair, and tumor progression. Here, we identified a novel gene, nxhl (New XingHuo light), that is conserved in vertebrates and that plays a crucial role in vascular integrity and angiogenesis. Bioinformatic analysis uncovered its essential roles in development based on co-expression with several key developmental genes. Knockdown of nxhl in zebrafish causes global and pericardial edema, loss of blood circulation, and vascular defects characterized by both reduced vascularization in intersegmental vessels and decreased sprouting in the caudal vein plexus. The nxhl gene also affects human endothelial cell behavior in vitro. We found that nxhl functions in part by targeting VE-PTP through interaction with NCL (nucleolin). Loss of ptprb (a VE-PTP ortholo) in zebrafish resulted in defects similar to nxhl knockdown. Moreover, nxhl deficiency attenuates tumor invasion and proteins (including VE-PTP and NCL) associated with angiogenesis and EMT. These findings illustrate that nxhl can regulate angiogenesis via a novel nxhl-NCL-VE-PTP axis, providing a new therapeutic target for modulating vascular formation and function, especially for cancer treatment.
Collapse
Affiliation(s)
- Honglin Luo
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China.,Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Yongde Zhang
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Yanfei Deng
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Lequn Li
- Department of Hepatobiliary Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Zhaoan Sheng
- College of Animal Science and Technology, Guangxi University, Nanning, China
| | - Yanling Yu
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Yong Lin
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Xiaohan Chen
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| | - Pengfei Feng
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Academy of Fishery Sciences, Nanning, China
| |
Collapse
|
20
|
Hou S, Niu G, Liu X, Bourbon PM, Zhang D, Cui P, Zhao K, Zhao D, Zeng H. A novel transcriptional complex on the VE-cadherin promoter regulated the downregulation of VE-cadherin in the Down Syndrome Candidate Region 1 isoform 1L-mediated angiogenesis. Microvasc Res 2021; 138:104209. [PMID: 34146582 PMCID: PMC9295908 DOI: 10.1016/j.mvr.2021.104209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 05/18/2021] [Accepted: 06/10/2021] [Indexed: 11/26/2022]
Abstract
Angiogenesis is critical for many diseases. Previously, we reported that Down Syndrome Candidate Region 1 isoform 1L (DSCR1-1L) was one of the most up-regulated genes in endothelial cells induced by VEGF and histamine, and regulated endothelial cell proliferation and Matrigel angiogenesis in mice. However, it was not known whether DSCR1-1L regulated angiogenesis in vivo and what was the molecular mechanism underlying it. In this study, gene knockdown and overexpression models were established to study the role of DSCR1-1L in angiogenesis in vivo. Further, the downstream regulatory target of DSCR1-1L was explored with molecular biological methods in vascular endothelial cells. We found that DSCR1-1L shRNAs significantly inhibited angiogenesis induced by VEGF in mice (p < 0.0001). In the gain-of-function assay, overexpression of DSCR1-1L cDNA in mouse endothelium of EC-FH-DSCR1-1L transgenic mice was sufficient to induce angiogenesis significantly (p < 0.01). DSCR1-1L regulated angiogenesis in the early stage by down-regulation of the VE-cadherin expression through targeting its transcription, but not mRNA stability. Three DSCR1-1L-targeted DNA elements in the VE-cadherin promoter were identified by promoter reporter assays, among which, a novel specific transcriptional complex was found. The DNA sequence (CTTCTG) in the VE-cadherin promoter was identified to directly interact with proteins by Electrophoresis Mobility Shift Assays and DNase I footprint assay. Hence, DSCR1-1L is an excellent therapeutic target for angiogenic diseases through down-regulating the formation of a novel transcriptional complex on the VE-cadherin promoter. DSCR1-1L shRNAs and cDNA have the potential to be developed for clinical application. Our results also contribute significantly to the field of mechanistic studies.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cadherins/genetics
- Cadherins/metabolism
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Down-Regulation
- Female
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Intracellular Signaling Peptides and Proteins/genetics
- Intracellular Signaling Peptides and Proteins/metabolism
- Male
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/genetics
- Melanoma, Experimental/metabolism
- Mice, Nude
- Mice, Transgenic
- Muscle Proteins/genetics
- Muscle Proteins/metabolism
- Neovascularization, Pathologic
- Neovascularization, Physiologic
- Promoter Regions, Genetic
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Shiqiang Hou
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China
| | - Gengming Niu
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Department of General Surgery, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Xin Liu
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Pierre M Bourbon
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dongmei Zhang
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Key Laboratory of Chinese Internal Medicine, Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, PR China
| | - Pengfei Cui
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA; Pancreatic Disease Institute, Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Kevin Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dezheng Zhao
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Huiyan Zeng
- Center for Vascular Biology Research and Division of Gastroenterology, Departments of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
21
|
Adrian E, Treľová D, Filová E, Kumorek M, Lobaz V, Poreba R, Janoušková O, Pop-Georgievski O, Lacík I, Kubies D. Complexation of CXCL12, FGF-2 and VEGF with Heparin Modulates the Protein Release from Alginate Microbeads. Int J Mol Sci 2021; 22:11666. [PMID: 34769095 PMCID: PMC8583835 DOI: 10.3390/ijms222111666] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/20/2021] [Accepted: 10/25/2021] [Indexed: 12/19/2022] Open
Abstract
Long-term delivery of growth factors and immunomodulatory agents is highly required to support the integrity of tissue in engineering constructs, e.g., formation of vasculature, and to minimize immune response in a recipient. However, for proteins with a net positive charge at the physiological pH, controlled delivery from negatively charged alginate (Alg) platforms is challenging due to electrostatic interactions that can hamper the protein release. In order to regulate such interactions between proteins and the Alg matrix, we propose to complex proteins of interest in this study - CXCL12, FGF-2, VEGF - with polyanionic heparin prior to their encapsulation into Alg microbeads of high content of α-L-guluronic acid units (high-G). This strategy effectively reduced protein interactions with Alg (as shown by model ITC and SPR experiments) and, depending on the protein type, afforded control over the protein release for at least one month. The released proteins retained their in vitro bioactivity: CXCL12 stimulated the migration of Jurkat cells, and FGF-2 and VEGF induced proliferation and maturation of HUVECs. The presence of heparin also intensified protein biological efficiency. The proposed approach for encapsulation of proteins with a positive net charge into high-G Alg hydrogels is promising for controlled long-term protein delivery under in vivo conditions.
Collapse
Affiliation(s)
- Edyta Adrian
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
- Department of Chemical Engineering, University of Chemistry and Technology, Technicka 5, 166 28 Prague, Czech Republic
| | - Dušana Treľová
- Polymer Institute of the Slovak Academy of Sciences, Dubravska cesta 9, 845 41 Bratislava, Slovakia; (D.T.); (I.L.)
| | - Elena Filová
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic;
| | - Marta Kumorek
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
| | - Volodymyr Lobaz
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
| | - Rafal Poreba
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
| | - Olga Janoušková
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
| | - Ognen Pop-Georgievski
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
| | - Igor Lacík
- Polymer Institute of the Slovak Academy of Sciences, Dubravska cesta 9, 845 41 Bratislava, Slovakia; (D.T.); (I.L.)
- Centre for Advanced Materials Application of the Slovak Academy of Sciences, Dubravska cesta 9, 845 11 Bratislava, Slovakia
| | - Dana Kubies
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
| |
Collapse
|
22
|
Waters SB, Zhou C, Nguyen T, Zelkha R, Lee H, Kazlauskas A, Rosenblatt MI, Malik AB, Yamada KH. VEGFR2 Trafficking by KIF13B Is a Novel Therapeutic Target for Wet Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2021; 62:5. [PMID: 33533881 PMCID: PMC7862734 DOI: 10.1167/iovs.62.2.5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Purpose Vascular endothelial growth factor (VEGF) and its receptor VEGFR2 are promising therapeutic targets for wet age-related macular degeneration (AMD). As a topically applicable option, we developed the peptide KAI to selectively interfere with VEGFR2 trafficking to the cell surface where it receives VEGF. This study sought to determine the efficacy of KAI in the mouse model of choroidal neovascularization (CNV). Methods The specificity of KAI was tested by surface plasmon resonance. The drug delivery was analyzed by cryosection and the ELISA after treatment of KAI eyedrop to the mouse eyes. For the laser-induced CNV model, mice with laser-induced ruptures in Bruch's membrane received daily treatment of KAI eyedrop or control peptide. The other groups of mice received intravitreal injection of anti-VEGF or IgG control. After two weeks, CNV was quantified and compared. Results First, we showed the specificity and high affinity of KAI to VEGFR2. Next, biodistribution revealed successful delivery of KAI eyedrop to the back of the mouse eyes. KAI significantly reduced the disease progression in laser-induced CNV. The comparison with current therapy suggests that KAI eyedrop is as effective as current therapy to prevent CNV in wet AMD. Moreover, the genetic deletion of a kinesin KIF13B, which mediates VEGFR2 trafficking to the cell surface, confirmed the pivotal role of KIF13B in disease progression of wet AMD and neovascularization from choroidal vessels. Conclusions Taken together, pharmacologic inhibition and genetic deletion complementarily suggest the therapeutic possibility of targeting VEGFR2 trafficking to inhibit pathological angiogenesis in wet AMD.
Collapse
Affiliation(s)
- Stephen B Waters
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, United States
| | - Christopher Zhou
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, United States
| | - Tara Nguyen
- Department of Ophthalmology and Visual Sciences, University of Illinois College of Medicine, Chicago, Illinois, United States
| | - Ruth Zelkha
- Department of Ophthalmology and Visual Sciences, University of Illinois College of Medicine, Chicago, Illinois, United States
| | - Hyun Lee
- Biophysics Core & Department of Pharmaceutical Sciences, University of Illinois College of Medicine, Chicago, Illinois, United States
| | - Andrius Kazlauskas
- Department of Ophthalmology and Visual Sciences, University of Illinois College of Medicine, Chicago, Illinois, United States.,Department of Physiology and Biophysics, University of Illinois College of Medicine, Chicago, Illinois, United States
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, University of Illinois College of Medicine, Chicago, Illinois, United States
| | - Asrar B Malik
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, United States
| | - Kaori H Yamada
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, United States.,Department of Ophthalmology and Visual Sciences, University of Illinois College of Medicine, Chicago, Illinois, United States
| |
Collapse
|
23
|
Cornelissen A, Guo L, Fernandez R, Kelly MC, Janifer C, Kuntz S, Sakamoto A, Jinnouchi H, Sato Y, Paek KH, Kolodgie FD, Romero ME, Surve D, Virmani R, Finn AV. Endothelial Recovery in Bare Metal Stents and Drug-Eluting Stents on a Single-Cell Level. Arterioscler Thromb Vasc Biol 2021; 41:2277-2292. [PMID: 34162228 DOI: 10.1161/atvbaha.121.316472] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Anne Cornelissen
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.).,Department of Cardiology, University Hospital RWTH Aachen, Germany (A.C.)
| | - Liang Guo
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Raquel Fernandez
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Michael C Kelly
- Single Cell Analysis Facility, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD (M.C.K.)
| | - Christine Janifer
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Salome Kuntz
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Atsushi Sakamoto
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Hiroyuki Jinnouchi
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Yu Sato
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Ka Hyun Paek
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Frank D Kolodgie
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Maria E Romero
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Dipti Surve
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Renu Virmani
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.)
| | - Aloke V Finn
- CVPath Institute, Gaithersburg, MD (A.C., L.G., R.F., C.J., S.K., A.S., H.J., Y.S., K.H.P., F.D.K., M.E.R., D.S., R.V., A.V.F.).,University of Maryland, School of Medicine, Baltimore (A.V.F.)
| |
Collapse
|
24
|
Siragusa M, Oliveira Justo AF, Malacarne PF, Strano A, Buch A, Withers B, Peters KG, Fleming I. VE-PTP inhibition elicits eNOS phosphorylation to blunt endothelial dysfunction and hypertension in diabetes. Cardiovasc Res 2021; 117:1546-1556. [PMID: 32653904 DOI: 10.1093/cvr/cvaa213] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 06/18/2020] [Accepted: 07/06/2020] [Indexed: 12/11/2022] Open
Abstract
AIMS Receptor-type vascular endothelial protein tyrosine phosphatase (VE-PTP) dephosphorylates Tie-2 as well as CD31, VE-cadherin, and vascular endothelial growth factor receptor 2 (VEGFR2). The latter form a signal transduction complex that mediates the endothelial cell response to shear stress, including the activation of the endothelial nitric oxide (NO) synthase (eNOS). As VE-PTP expression is increased in diabetes, we investigated the consequences of VE-PTP inhibition (using AKB-9778) on blood pressure in diabetic patients and the role of VE-PTP in the regulation of eNOS activity and vascular reactivity. METHODS AND RESULTS In diabetic patients AKB-9778 significantly lowered systolic and diastolic blood pressure. This could be linked to elevated NO production, as AKB increased NO generation by cultured endothelial cells and elicited the NOS inhibitor-sensitive relaxation of endothelium-intact rings of mouse aorta. At the molecular level, VE-PTP inhibition increased the phosphorylation of eNOS on Tyr81 and Ser1177 (human sequence). The PIEZO1 activator Yoda1, which was used to mimic the response to shear stress, also increased eNOS Tyr81 phosphorylation, an effect that was enhanced by VE-PTP inhibition. Two kinases, i.e. abelson-tyrosine protein kinase (ABL)1 and Src were identified as eNOS Tyr81 kinases as their inhibition and down-regulation significantly reduced the basal and Yoda1-induced tyrosine phosphorylation and activity of eNOS. VE-PTP, on the other hand, formed a complex with eNOS in endothelial cells and directly dephosphorylated eNOS Tyr81 in vitro. Finally, phosphorylation of eNOS on Tyr80 (murine sequence) was found to be reduced in diabetic mice and diabetes-induced endothelial dysfunction (isolated aortic rings) was blunted by VE-PTP inhibition. CONCLUSIONS VE-PTP inhibition enhances eNOS activity to improve endothelial function and decrease blood pressure indirectly, through the activation of Tie-2 and the CD31/VE-cadherin/VEGFR2 complex, and directly by dephosphorylating eNOS Tyr81. VE-PTP inhibition, therefore, represents an attractive novel therapeutic option for diabetes-induced endothelial dysfunction and hypertension.
Collapse
MESH Headings
- Aniline Compounds/therapeutic use
- Animals
- Antihypertensive Agents/therapeutic use
- Blood Pressure/drug effects
- Cells, Cultured
- Diabetes Mellitus/drug therapy
- Diabetes Mellitus/enzymology
- Diabetes Mellitus/genetics
- Diabetes Mellitus/physiopathology
- Disease Models, Animal
- Endothelial Cells/drug effects
- Endothelial Cells/enzymology
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/physiopathology
- Enzyme Inhibitors/therapeutic use
- Humans
- Hypertension/drug therapy
- Hypertension/enzymology
- Hypertension/genetics
- Hypertension/physiopathology
- Mice, Inbred C57BL
- Mice, Transgenic
- Nitric Oxide/metabolism
- Nitric Oxide Synthase Type III/genetics
- Nitric Oxide Synthase Type III/metabolism
- Phosphorylation
- Receptor-Like Protein Tyrosine Phosphatases, Class 3/antagonists & inhibitors
- Receptor-Like Protein Tyrosine Phosphatases, Class 3/genetics
- Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism
- Signal Transduction
- Sulfonic Acids/therapeutic use
- Treatment Outcome
- United States
- Mice
Collapse
Affiliation(s)
- Mauro Siragusa
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany
| | - Alberto Fernando Oliveira Justo
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | | | - Anna Strano
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Akshay Buch
- Aerpio Pharmaceuticals, Inc., Cincinnati, OH, USA
| | | | | | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
- German Center for Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany
| |
Collapse
|
25
|
Cai A, Chatziantoniou C, Calmont A. Vascular Permeability: Regulation Pathways and Role in Kidney Diseases. Nephron Clin Pract 2021; 145:297-310. [PMID: 33744890 DOI: 10.1159/000514314] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 01/08/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Vascular permeability (VP) is a fundamental aspect of vascular biology. A growing number of studies have revealed that many signalling pathways govern VP in both physiological and pathophysiological conditions. Furthermore, emerging evidence identifies VP alteration as a pivotal pathogenic factor in acute kidney injury, chronic kidney disease, diabetic kidney disease, and other proteinuric diseases. Therefore, perceiving the connections between these pathways and the aetiology of kidney disease is an important task as such knowledge may trigger the development of novel therapeutic or preventive medical approaches. In this regard, the discussion summarizing VP-regulating pathways and associating them with kidney diseases is highly warranted. SUMMARY Major pathways of VP regulation comprise angiogenic factors including vascular endothelial growth factor/VEGFR, angiopoietin/Tie, and class 3 semaphorin/neuropilin and inflammatory factors including histamine, platelet-activating factor, and leukocyte extravasation. These pathways mainly act on vascular endothelial cadherin to modulate adherens junctions of endothelial cells (ECs), thereby augmenting VP via the paracellular pathway. Elevated VP in diverse kidney diseases involves EC apoptosis, imbalanced regulatory factors, and many other pathophysiological events, which in turn exacerbates renal structural and functional disorders. Measures improving VP effectively ameliorate the diseased kidney in terms of tissue injury, endothelial dysfunction, kidney function, and long-term prognosis. Key Messages: (1) Angiogenic factors, inflammatory factors, and adhesion molecules represent major pathways that regulate VP. (2) Vascular hyperpermeability links various pathophysiological processes and plays detrimental roles in multiple kidney diseases.
Collapse
Affiliation(s)
- Anxiang Cai
- Unité mixte Inserm - Sorbonne Université, UMR_S1155, Tenon Hospital, Paris, France,
| | | | - Amélie Calmont
- Unité mixte Inserm - Sorbonne Université, UMR_S1155, Tenon Hospital, Paris, France
| |
Collapse
|
26
|
Yoodee S, Peerapen P, Plumworasawat S, Thongboonkerd V. ARID1A knockdown in human endothelial cells directly induces angiogenesis by regulating angiopoietin-2 secretion and endothelial cell activity. Int J Biol Macromol 2021; 180:1-13. [PMID: 33675830 DOI: 10.1016/j.ijbiomac.2021.02.218] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 02/10/2021] [Accepted: 02/24/2021] [Indexed: 12/29/2022]
Abstract
AT-rich interactive domain 1A (ARID1A) is a novel tumor suppressor gene found in several human cells and its loss/defect is commonly observed in many cancers. However, its roles in angiogenesis, which is one of the hallmarks for tumor progression, remained unclear. Herein, we demonstrated the direct effects of ARID1A knockdown in human endothelial cells by lentivirus-based short-hairpin RNA (shRNA) (shARID1A) on angiogenesis. Functional assays revealed that shARID1A significantly enhanced cell proliferation and migration/invasion and endothelial tube formation compared with the control cells transfected with scramble shRNA (shControl). Additionally, the shARID1A-transfected cells had significantly increased podosome formation and secretion of angiopoietin-2 (ANG2), a key angiogenic factor. Moreover, neutralization of ANG2 with monoclonal anti-ANG2 antibody strongly reduced cell proliferation and migration/invasion and endothelial tube formation in the shARID1A-transfected cells. These findings indicate that down-regulation of ARID1A in human endothelial cells directly induces angiogenesis by regulating angiopoietin-2 secretion and endothelial cell activity.
Collapse
Affiliation(s)
- Sunisa Yoodee
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Paleerath Peerapen
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sirikanya Plumworasawat
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Office for Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| |
Collapse
|
27
|
Vestweber D. Vascular Endothelial Protein Tyrosine Phosphatase Regulates Endothelial Function. Physiology (Bethesda) 2021; 36:84-93. [PMID: 33595386 DOI: 10.1152/physiol.00026.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Vascular endothelial protein tyrosine phosphatase (VE-PTP) is a receptor-type PTP (RPTP), predominantly expressed in vascular endothelial cells. It regulates embryonic and tumor angiogenesis and controls vascular permeability and homeostasis in inflammation. Major substrates are the tyrosine kinase receptor Tie-2 and the adhesion molecule VE-cadherin. This review describes how VE-PTP controls vascular functions by its various substrates and the therapeutic potential of VE-PTP in various pathophysiological settings.
Collapse
|
28
|
Zink J, Frye M, Frömel T, Carlantoni C, John D, Schreier D, Weigert A, Laban H, Salinas G, Stingl H, Günther L, Popp R, Hu J, Vanhollebeke B, Schmidt H, Acker-Palmer A, Renné T, Fleming I, Benz PM. EVL regulates VEGF receptor-2 internalization and signaling in developmental angiogenesis. EMBO Rep 2021; 22:e48961. [PMID: 33512764 PMCID: PMC7857432 DOI: 10.15252/embr.201948961] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 12/03/2020] [Accepted: 12/09/2020] [Indexed: 12/11/2022] Open
Abstract
Endothelial tip cells are essential for VEGF‐induced angiogenesis, but underlying mechanisms are elusive. The Ena/VASP protein family, consisting of EVL, VASP, and Mena, plays a pivotal role in axon guidance. Given that axonal growth cones and endothelial tip cells share many common features, from the morphological to the molecular level, we investigated the role of Ena/VASP proteins in angiogenesis. EVL and VASP, but not Mena, are expressed in endothelial cells of the postnatal mouse retina. Global deletion of EVL (but not VASP) compromises the radial sprouting of the vascular plexus in mice. Similarly, endothelial‐specific EVL deletion compromises the radial sprouting of the vascular plexus and reduces the endothelial tip cell density and filopodia formation. Gene sets involved in blood vessel development and angiogenesis are down‐regulated in EVL‐deficient P5‐retinal endothelial cells. Consistently, EVL deletion impairs VEGF‐induced endothelial cell proliferation and sprouting, and reduces the internalization and phosphorylation of VEGF receptor 2 and its downstream signaling via the MAPK/ERK pathway. Together, we show that endothelial EVL regulates sprouting angiogenesis via VEGF receptor‐2 internalization and signaling.
Collapse
Affiliation(s)
- Joana Zink
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Maike Frye
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Timo Frömel
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Claudia Carlantoni
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David John
- German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany.,Insitute for Cardiovascular Regeneration, Goethe University, Frankfurt am Main, Germany
| | - Danny Schreier
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Weigert
- Institute of Biochemistry I-Pathobiochemistry, Faculty of Medicine, Goethe-University, Frankfurt am Main, Germany
| | - Hebatullah Laban
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Gabriela Salinas
- NGS-Integrative Genomics Core Unit (NIG), Institute of Human Genetics, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Heike Stingl
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Lea Günther
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Rüdiger Popp
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Jiong Hu
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Benoit Vanhollebeke
- Laboratory of Neurovascular Signaling, ULB Neuroscience Institute Department of Molecular Biology, University of Brussels, Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Brussels, Belgium
| | - Hannes Schmidt
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Amparo Acker-Palmer
- Institute of Cell Biology and Neuroscience and Buchmann Institute for Molecular Life Sciences, Goethe University, Frankfurt am Main, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingrid Fleming
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| | - Peter M Benz
- Centre for Molecular Medicine, Institute for Vascular Signalling, Goethe University, Frankfurt am Main, Germany.,German Centre of Cardiovascular Research (DZHK), Partner site Rhein-Main, Frankfurt am Main, Germany
| |
Collapse
|
29
|
Mercier C, Rousseau M, Geraldes P. Growth Factor Deregulation and Emerging Role of Phosphatases in Diabetic Peripheral Artery Disease. Front Cardiovasc Med 2021; 7:619612. [PMID: 33490120 PMCID: PMC7817696 DOI: 10.3389/fcvm.2020.619612] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/10/2020] [Indexed: 01/25/2023] Open
Abstract
Peripheral artery disease is caused by atherosclerosis of lower extremity arteries leading to the loss of blood perfusion and subsequent critical ischemia. The presence of diabetes mellitus is an important risk factor that greatly increases the incidence, the progression and the severity of the disease. In addition to accelerated disease progression, diabetic patients are also more susceptible to develop serious impairment of their walking abilities through an increased risk of lower limb amputation. Hyperglycemia is known to alter the physiological development of collateral arteries in response to ischemia. Deregulation in the production of several critical pro-angiogenic factors has been reported in diabetes along with vascular cell unresponsiveness in initiating angiogenic processes. Among the multiple molecular mechanisms involved in the angiogenic response, protein tyrosine phosphatases are potent regulators by dephosphorylating pro-angiogenic tyrosine kinase receptors. However, evidence has indicated that diabetes-induced deregulation of phosphatases contributes to the progression of several micro and macrovascular complications. This review provides an overview of growth factor alterations in the context of diabetes and peripheral artery disease, as well as a description of the role of phosphatases in the regulation of angiogenic pathways followed by an analysis of the effects of hyperglycemia on the modulation of protein tyrosine phosphatase expression and activity. Knowledge of the role of phosphatases in diabetic peripheral artery disease will help the development of future therapeutics to locally regulate phosphatases and improve angiogenesis.
Collapse
Affiliation(s)
- Clément Mercier
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Marina Rousseau
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Pedro Geraldes
- Department of Medicine, Division of Endocrinology, Research Center of the Centre Hospitalier Universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
30
|
Vázquez R, Riveiro ME, Berenguer-Daizé C, O'Kane A, Gormley J, Touzelet O, Rezai K, Bekradda M, Ouafik L. Targeting Adrenomedullin in Oncology: A Feasible Strategy With Potential as Much More Than an Alternative Anti-Angiogenic Therapy. Front Oncol 2021; 10:589218. [PMID: 33489885 PMCID: PMC7815935 DOI: 10.3389/fonc.2020.589218] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/02/2020] [Indexed: 12/18/2022] Open
Abstract
The development, maintenance and metastasis of solid tumors are highly dependent on the formation of blood and lymphatic vessels from pre-existing ones through a series of processes that are respectively known as angiogenesis and lymphangiogenesis. Both are mediated by specific growth-stimulating molecules, such as the vascular endothelial growth factor (VEGF) and adrenomedullin (AM), secreted by diverse cell types which involve not only the cancerogenic ones, but also those constituting the tumor stroma (i.e., macrophages, pericytes, fibroblasts, and endothelial cells). In this sense, anti-angiogenic therapy represents a clinically-validated strategy in oncology. Current therapeutic approaches are mainly based on VEGF-targeting agents, which, unfortunately, are usually limited by toxicity and/or tumor-acquired resistance. AM is a ubiquitous peptide hormone mainly secreted in the endothelium with an important involvement in blood vessel development and cardiovascular homeostasis. In this review, we will introduce the state-of-the-art in terms of AM physiology, while putting a special focus on its pro-tumorigenic role, and discuss its potential as a therapeutic target in oncology. A large amount of research has evidenced AM overexpression in a vast majority of solid tumors and a correlation between AM levels and disease stage, progression and/or vascular density has been observed. The analysis presented here indicates that the involvement of AM in the pathogenesis of cancer arises from: 1) direct promotion of cell proliferation and survival; 2) increased vascularization and the subsequent supply of nutrients and oxygen to the tumor; 3) and/or alteration of the cell phenotype into a more aggressive one. Furthermore, we have performed a deep scrutiny of the pathophysiological prominence of each of the AM receptors (AM1 and AM2) in different cancers, highlighting their differential locations and functions, as well as regulatory mechanisms. From the therapeutic point of view, we summarize here an exhaustive series of preclinical studies showing a reduction of tumor angiogenesis, metastasis and growth following treatment with AM-neutralizing antibodies, AM receptor antagonists, or AM receptor interference. Anti-AM therapy is a promising strategy to be explored in oncology, not only as an anti-angiogenic alternative in the context of acquired resistance to VEGF treatment, but also as a potential anti-metastatic approach.
Collapse
Affiliation(s)
- Ramiro Vázquez
- Preclinical Department, Early Drug Development Group (E2DG), Boulogne-Billancourt, France.,Center for Genomic Science of IIT@SEMM, Fondazione Istituto Italiano di Tecnologia (IIT), Milan, Italy
| | - Maria E Riveiro
- Preclinical Department, Early Drug Development Group (E2DG), Boulogne-Billancourt, France
| | | | - Anthony O'Kane
- Discovery and Scientific Affairs Department, Fusion Antibodies plc., Belfast, United Kingdom
| | - Julie Gormley
- Discovery and Scientific Affairs Department, Fusion Antibodies plc., Belfast, United Kingdom
| | - Olivier Touzelet
- Discovery and Scientific Affairs Department, Fusion Antibodies plc., Belfast, United Kingdom
| | - Keyvan Rezai
- Department of Radio-Pharmacology, Institute Curie-René Huguenin Hospital, Saint-Cloud, France
| | - Mohamed Bekradda
- Preclinical Department, Early Drug Development Group (E2DG), Boulogne-Billancourt, France
| | - L'Houcine Ouafik
- Aix Marseille University, CNRS, INP, Institute of NeuroPhysiopathology, Marseille, France.,APHM, CHU Nord, Service de Transfert d'Oncologie Biologique, Marseille, France
| |
Collapse
|
31
|
Koh BI, Lee HJ, Kwak PA, Yang MJ, Kim JH, Kim HS, Koh GY, Kim I. VEGFR2 signaling drives meningeal vascular regeneration upon head injury. Nat Commun 2020; 11:3866. [PMID: 32737287 PMCID: PMC7395111 DOI: 10.1038/s41467-020-17545-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 07/06/2020] [Indexed: 01/04/2023] Open
Abstract
Upon severe head injury (HI), blood vessels of the meninges and brain parenchyma are inevitably damaged. While limited vascular regeneration of the injured brain has been studied extensively, our understanding of meningeal vascular regeneration following head injury is quite limited. Here, we identify key pathways governing meningeal vascular regeneration following HI. Rapid and complete vascular regeneration in the meninges is predominantly driven by VEGFR2 signaling. Substantial increase of VEGFR2 is observed in both human patients and mouse models of HI, and endothelial cell-specific deletion of Vegfr2 in the latter inhibits meningeal vascular regeneration. We further identify the facilitating, stabilizing and arresting roles of Tie2, PDGFRβ and Dll4 signaling, respectively, in meningeal vascular regeneration. Prolonged inhibition of this angiogenic process following HI compromises immunological and stromal integrity of the injured meninges. These findings establish a molecular framework for meningeal vascular regeneration after HI, and may guide development of wound healing therapeutics. Severe head injury results in critical damage of blood vessels of the meninges and brain parenchyma. Here, the authors describe key pathways governing meningeal vascular regeneration following head injury, characterizing the differential roles of VEGFR2, Tie2, Dll4 and PDGFRβ signaling.
Collapse
Affiliation(s)
- Bong Ihn Koh
- KI for Bio-century, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Hyuek Jong Lee
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, 34141, Republic of Korea
| | - Pil Ae Kwak
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, 34141, Republic of Korea
| | - Myung Jin Yang
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, 34141, Republic of Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Ju-Hee Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju, 61463, Republic of Korea
| | - Gou Young Koh
- KI for Bio-century, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea. .,Center for Vascular Research, Institute for Basic Science (IBS), Daejeon, 34141, Republic of Korea. .,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.
| | - Injune Kim
- KI for Bio-century, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea. .,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
32
|
Yanagida K, Engelbrecht E, Niaudet C, Jung B, Gaengel K, Holton K, Swendeman S, Liu CH, Levesque MV, Kuo A, Fu Z, Smith LEH, Betsholtz C, Hla T. Sphingosine 1-Phosphate Receptor Signaling Establishes AP-1 Gradients to Allow for Retinal Endothelial Cell Specialization. Dev Cell 2020; 52:779-793.e7. [PMID: 32059774 DOI: 10.1016/j.devcel.2020.01.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 12/09/2019] [Accepted: 01/16/2020] [Indexed: 12/17/2022]
Abstract
Transcriptional mechanisms that drive angiogenesis and organotypic vascular endothelial cell specialization are poorly understood. Here, we show that retinal endothelial sphingosine 1-phosphate receptors (S1PRs), which restrain vascular endothelial growth factor (VEGF)-induced angiogenesis, spatially restrict expression of JunB, a member of the activator protein 1 (AP-1) family of transcription factors (TFs). Mechanistically, VEGF induces JunB expression at the sprouting vascular front while S1PR-dependent vascular endothelial (VE)-cadherin assembly suppresses JunB expression in the nascent vascular network, thus creating a gradient of this TF. Endothelial-specific JunB knockout mice showed diminished expression of neurovascular guidance genes and attenuated retinal vascular network progression. In addition, endothelial S1PR signaling is required for normal expression of β-catenin-dependent genes such as TCF/LEF1 and ZIC3 TFs, transporters, and junctional proteins. These results show that S1PR signaling restricts JunB function to the expanding vascular front, thus creating an AP-1 gradient and enabling organotypic endothelial cell specialization of the vascular network.
Collapse
Affiliation(s)
- Keisuke Yanagida
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Eric Engelbrecht
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Colin Niaudet
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Bongnam Jung
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Konstantin Gaengel
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Steven Swendeman
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Catherine H Liu
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Michel V Levesque
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Andrew Kuo
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; ICMC (Integrated Cardio Metabolic Centre), Karolinska Institutet, Novum, Huddinge, Sweden
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
33
|
Abstract
Lethal features of sepsis and acute respiratory distress syndrome (ARDS) relate to the health of small blood vessels. For example, alveolar infiltration with proteinaceous fluid is often driven by breach of the microvascular barrier. Spontaneous thrombus formation within inflamed microvessels exacerbates organ ischemia, and in its final stages, erupts into overt disseminated intravascular coagulation. Disruption of an endothelial signaling axis, the Angiopoietin-Tie2 pathway, may mediate the abrupt transition from microvascular integrity to pathologic disruption. This review summarizes preclinical and clinical results that implicate the Tie2 pathway as a promising target to restore microvascular health in sepsis and ARDS.
Collapse
Affiliation(s)
- Kelsey D Sack
- Department of Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, RN330C, Boston, MA 02215, USA
| | - John A Kellum
- Department of Critical Care Medicine, CRISMA Center, University of Pittsburgh, University of Pittsburgh, School of Medicine, 3347 Forbes Avenue, Suite 220, Room 202, Pittsburgh, PA 15213, USA
| | - Samir M Parikh
- Department of Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, RN330C, Boston, MA 02215, USA.
| |
Collapse
|
34
|
Bloch E, Sikorski EL, Pontoriero D, Day EK, Berger BW, Lazzara MJ, Thévenin D. Disrupting the transmembrane domain-mediated oligomerization of protein tyrosine phosphatase receptor J inhibits EGFR-driven cancer cell phenotypes. J Biol Chem 2019; 294:18796-18806. [PMID: 31676686 DOI: 10.1074/jbc.ra119.010229] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/21/2019] [Indexed: 12/19/2022] Open
Abstract
Receptor protein tyrosine phosphatases (RPTPs) play critical regulatory roles in mammalian signal transduction. However, the structural basis for the regulation of their catalytic activity is not fully understood, and RPTPs are generally not therapeutically targetable. This knowledge gap is partially due to the lack of known natural ligands or selective agonists of RPTPs. Contrary to what is known from structure-function studies of receptor tyrosine kinases (RTKs), RPTP activities have been reported to be suppressed by dimerization, which may prevent RPTPs from accessing their RTK substrates. We report here that homodimerization of protein tyrosine phosphatase receptor J (PTPRJ, also known as DEP-1) is regulated by specific transmembrane (TM) residues. We found that disrupting these interactions destabilizes homodimerization of full-length PTPRJ in cells, reduces the phosphorylation of the known PTPRJ substrate epidermal growth factor receptor (EGFR) and of other downstream signaling effectors, antagonizes EGFR-driven cell phenotypes, and promotes substrate access. We demonstrate these observations in human cancer cells using mutational studies and identified a peptide that binds to the PTPRJ TM domain and represents the first example of an allosteric agonist of RPTPs. The results of our study provide fundamental structural and functional insights into how PTPRJ activity is tuned by TM interactions in cells. Our findings also open up opportunities for developing peptide-based agents that could be used as tools to probe RPTPs' signaling mechanisms or to manage cancers driven by RTK signaling.
Collapse
Affiliation(s)
- Elizabeth Bloch
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015
| | - Eden L Sikorski
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015
| | - David Pontoriero
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Evan K Day
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Bryan W Berger
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Matthew J Lazzara
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia 22903
| | - Damien Thévenin
- Department of Chemistry, Lehigh University, Bethlehem, Pennsylvania 18015.
| |
Collapse
|
35
|
Thomson BR, Carota IA, Souma T, Soman S, Vestweber D, Quaggin SE. Targeting the vascular-specific phosphatase PTPRB protects against retinal ganglion cell loss in a pre-clinical model of glaucoma. eLife 2019; 8:48474. [PMID: 31621585 PMCID: PMC6874417 DOI: 10.7554/elife.48474] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 10/11/2019] [Indexed: 12/14/2022] Open
Abstract
Elevated intraocular pressure (IOP) due to insufficient aqueous humor outflow through the trabecular meshwork and Schlemm’s canal (SC) is the most important risk factor for glaucoma, a leading cause of blindness worldwide. We previously reported loss of function mutations in the receptor tyrosine kinase TEK or its ligand ANGPT1 cause primary congenital glaucoma in humans and mice due to failure of SC development. Here, we describe a novel approach to enhance canal formation in these animals by deleting a single allele of the gene encoding the phosphatase PTPRB during development. Compared to Tek haploinsufficient mice, which exhibit elevated IOP and loss of retinal ganglion cells, Tek+/-;Ptprb+/- mice have elevated TEK phosphorylation, which allows normal SC development and prevents ocular hypertension and RGC loss. These studies provide evidence that PTPRB is an important regulator of TEK signaling in the aqueous humor outflow pathway and identify a new therapeutic target for treatment of glaucoma.
Collapse
Affiliation(s)
- Benjamin R Thomson
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, United States.,Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Isabel A Carota
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, United States.,Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Tomokazu Souma
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, United States.,Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Saily Soman
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, United States.,Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, United States
| | | | - Susan E Quaggin
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, United States.,Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, United States
| |
Collapse
|
36
|
Regulation of VEGFR2 trafficking and signaling by Rab GTPase-activating proteins. Sci Rep 2019; 9:13342. [PMID: 31527750 PMCID: PMC6746989 DOI: 10.1038/s41598-019-49646-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 08/29/2019] [Indexed: 12/19/2022] Open
Abstract
Vascular endothelial growth factor receptor-2 (VEGFR2) and its ligands (VEGFs) are crucial players in vasculogenesis and angiogenesis. General blocking of this signaling system with antibodies or small molecule inhibitors is an established strategy to treat cancer and age-related macular degeneration. Nevertheless, the activated receptor can signal to discrete downstream signaling pathways and the equilibrium between these pathways is modulated by coreceptors and distinct isoforms of VEGF. Here we investigated the influence of Rab GTPase activating proteins (RabGAPs) on VEGFR2 signaling, tube formation, and migration of endothelial cells. We demonstrate that members of the TBC1D10 subfamily of RabGAPs have opposite effects. Whereas TBC1D10A leads to increased Erk1/2 signaling, TBC1D10B lowered Erk1/2 and p38 signaling and reduced tube formation in vitro. TBC1D10A is a RabGAP acting on RAB13 that was shown before to play a role in angiogenesis and we could indeed show colocalization of these two proteins with VEGFR2 in activated cells. In addition, we observed that cells expressing TBC1D10B show lower expression of VEGFR2 and NRP1 on filopodia of activated cells. Taken together, our systematic analysis of influence of RabGAPs on VEGFR2 signaling identifies the TBC1D10 subfamily members as modulators of angiogenesis.
Collapse
|
37
|
Braun LJ, Zinnhardt M, Vockel M, Drexler HC, Peters K, Vestweber D. VE-PTP inhibition stabilizes endothelial junctions by activating FGD5. EMBO Rep 2019; 20:e47046. [PMID: 31267715 PMCID: PMC6607018 DOI: 10.15252/embr.201847046] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 04/01/2019] [Accepted: 04/18/2019] [Indexed: 01/06/2023] Open
Abstract
Inhibition of VE-PTP, an endothelial receptor-type tyrosine phosphatase, triggers phosphorylation of the tyrosine kinase receptor Tie-2, which leads to the suppression of inflammation-induced vascular permeability. Analyzing the underlying mechanism, we show here that inhibition of VE-PTP and activation of Tie-2 induce tyrosine phosphorylation of FGD5, a GTPase exchange factor (GEF) for Cdc42, and stimulate its translocation to cell contacts. Interfering with the expression of FGD5 blocks the junction-stabilizing effect of VE-PTP inhibition in vitro and in vivo. Likewise, FGD5 is required for strengthening cortical actin bundles and inhibiting radial stress fiber formation, which are each stimulated by VE-PTP inhibition. We identify Y820 of FGD5 as the direct substrate for VE-PTP. The phosphorylation of FGD5-Y820 is required for the stabilization of endothelial junctions and for the activation of Cdc42 by VE-PTP inhibition but is dispensable for the recruitment of FGD5 to endothelial cell contacts. Thus, activation of FGD5 is a two-step process that comprises membrane recruitment and phosphorylation of Y820. These steps are necessary for the junction-stabilizing effect stimulated by VE-PTP inhibition and Tie-2 activation.
Collapse
Affiliation(s)
- Laura J Braun
- Max Planck Institute of Molecular BiomedicineMünsterGermany
| | | | - Matthias Vockel
- Max Planck Institute of Molecular BiomedicineMünsterGermany
- Present address:
Institute for Human GeneticsUniversity of MünsterMünsterGermany
| | | | | | | |
Collapse
|
38
|
Kim DY, Park JA, Kim Y, Noh M, Park S, Lie E, Kim E, Kim YM, Kwon YG. SALM4 regulates angiogenic functions in endothelial cells through VEGFR2 phosphorylation at Tyr1175. FASEB J 2019; 33:9842-9857. [PMID: 31170000 PMCID: PMC6704462 DOI: 10.1096/fj.201802516rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Angiogenesis depends on VEGF-mediated signaling. However, the regulatory mechanisms and functions of individual VEGF receptor 2 (VEGFR2) phosphorylation sites remain unclear. Here, we report that synaptic adhesion-like molecule 4 (SALM4) regulates a specific VEGFR2 phosphorylation site. SALM4 silencing in HUVECs and Salm4 knockout (KO) in lung endothelial cells (ECs) of Salm4−/− mice suppressed phosphorylation of VEGFR2 tyrosine (Y) 1175 (Y1173 in mice) and downstream signaling upon VEGF-A stimulation. However, VEGFR2 phosphorylation at Y951 (Y949 in mice) and Y1214 (Y1212 in mice) remained unchanged. Knockdown and KO of SALM4 inhibited VEGF-A–induced angiogenic functions of ECs. SALM4 depletion reduced endothelial leakage, sprouting, and migratory activities. Furthermore, in an ischemia and reperfusion (I/R) model, brain injury was attenuated in Salm4−/− mice compared with wild-type (WT) mice. In brain lysates after I/R, VEGFR2 phosphorylation at Y949, Y1173, and Y1212 were induced in WT brains, but only Y1173 phosphorylation of VEGFR2 was reduced in Salm4−/− brains. Taken together, our results demonstrate that SALM4 specifically regulates VEGFR2 phosphorylation at Y1175 (Y1173 in mice), thereby fine-tuning VEGF signaling in ECs.—Kim, D. Y., Park, J. A., Kim, Y., Noh, M., Park, S., Lie, E., Kim, E., Kim, Y.-M., Kwon, Y.-G. SALM4 regulates angiogenic functions in endothelial cells through VEGFR2 phosphorylation at Tyr1175.
Collapse
Affiliation(s)
- Dong Young Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Jeong Ae Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Yeomyung Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Minyoung Noh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Songyi Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Eunkyung Lie
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology, Daejeon, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute for Science and Technology, Daejeon, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon-si, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| |
Collapse
|
39
|
LeBlanc ME, Saez-Torres KL, Cano I, Hu Z, Saint-Geniez M, Ng YS, D'Amore PA. Glycocalyx regulation of vascular endothelial growth factor receptor 2 activity. FASEB J 2019; 33:9362-9373. [PMID: 31141406 DOI: 10.1096/fj.201900011r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
We have previously shown that knockdown of endomucin (EMCN), an integral membrane glycocalyx glycoprotein, prevents VEGF-induced proliferation, migration, and tube formation in vitro and angiogenesis in vivo. In the endothelium, VEGF mediates most of its angiogenic effects through VEGF receptor 2 (VEGFR2). To understand the role of EMCN, we examined the effect of EMCN depletion on VEGFR2 endocytosis and activation. Results showed that although VEGF stimulation promoted VEGFR2 internalization in control endothelial cells (ECs), loss of EMCN prevented VEGFR2 endocytosis. Cell surface analysis revealed a decrease in VEGFR2 following VEGF stimulation in control but not siRNA directed against EMCN-transfected ECs. EMCN depletion resulted in heightened phosphorylation following VEGF stimulation with an increase in total VEGFR2 protein. These results indicate that EMCN modulates VEGFR2 endocytosis and activity and point to EMCN as a potential therapeutic target.-LeBlanc, M. E., Saez-Torres, K. L., Cano, I., Hu, Z., Saint-Geniez, M., Ng, Y.-S., D'Amore, P. A. Glycocalyx regulation of vascular endothelial growth factor receptor 2 activity.
Collapse
Affiliation(s)
- Michelle E LeBlanc
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Kahira L Saez-Torres
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Issahy Cano
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Zhengping Hu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Magali Saint-Geniez
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Yin-Shan Ng
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, Massachusetts, USA.,Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Pathology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Abstract
Angiogenic blood vessel growth is essential to ensure organs receive adequate blood supply to support normal organ function and homeostasis. Angiogenesis involves a complex series of cellular events through which new vessels grow out from existing vasculature. Growth factor signaling, layered over a range of other signaling inputs, orchestrates this process. The response of endothelial cells (ECs) to growth factor signals must be carefully controlled through feedback mechanisms to prevent excessive vessel growth, remodeling or destabilization. In this article, we summarize recent findings describing how ECs respond to growth factor signals during blood vessel development and homeostasis and how perturbation of these responses can lead to disease.
Collapse
Affiliation(s)
- Zoe L Grant
- a The Walter and Eliza Hall Institute of Medical Research , Parkville , Australia
- b Department of Medical Biology, University of Melbourne , Parkville , Australia
| | - Leigh Coultas
- a The Walter and Eliza Hall Institute of Medical Research , Parkville , Australia
- b Department of Medical Biology, University of Melbourne , Parkville , Australia
| |
Collapse
|
41
|
Kim J, Park JR, Choi J, Park I, Hwang Y, Bae H, Kim Y, Choi W, Yang JM, Han S, Chung TY, Kim P, Kubota Y, Augustin HG, Oh WY, Koh GY. Tie2 activation promotes choriocapillary regeneration for alleviating neovascular age-related macular degeneration. SCIENCE ADVANCES 2019; 5:eaau6732. [PMID: 30788433 PMCID: PMC6374104 DOI: 10.1126/sciadv.aau6732] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 01/03/2019] [Indexed: 05/28/2023]
Abstract
Choriocapillary loss is a major cause of neovascular age-related macular degeneration (NV-AMD). Although vascular endothelial growth factor (VEGF) blockade for NV-AMD has shown beneficial outcomes, unmet medical needs for patients refractory or tachyphylactic to anti-VEGF therapy exist. In addition, the treatment could exacerbate choriocapillary rarefaction, necessitating advanced treatment for fundamental recovery from NV-AMD. In this study, Tie2 activation by angiopoietin-2-binding and Tie2-activating antibody (ABTAA) presents a therapeutic strategy for NV-AMD. Conditional Tie2 deletion impeded choriocapillary maintenance, rendering eyes susceptible to NV-AMD development. Moreover, in a NV-AMD mouse model, ABTAA not only suppressed choroidal neovascularization (CNV) and vascular leakage but also regenerated the choriocapillaris and relieved hypoxia. Conversely, VEGF blockade degenerated the choriocapillaris and exacerbated hypoxia, although it suppressed CNV and vascular leakage. Together, we establish that angiopoietin-Tie2 signaling is critical for choriocapillary maintenance and that ABTAA represents an alternative, combinative therapeutic strategy for NV-AMD by alleviating anti-VEGF adverse effects.
Collapse
Affiliation(s)
- Jaeryung Kim
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Jang Ryul Park
- Department of Mechanical Engineering, KAIST, Daejeon 34141, Republic of Korea
- KI for Health Science and Technology (KIHST), KAIST, Daejeon 34141, Republic of Korea
| | - Jeongwoon Choi
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Intae Park
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Yoonha Hwang
- KI for Health Science and Technology (KIHST), KAIST, Daejeon 34141, Republic of Korea
- Graduate School of Nanoscience and Technology, KAIST, Daejeon 34141, Republic of Korea
| | - Hosung Bae
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Yongjoo Kim
- Department of Mechanical Engineering, KAIST, Daejeon 34141, Republic of Korea
- KI for Health Science and Technology (KIHST), KAIST, Daejeon 34141, Republic of Korea
| | - WooJhon Choi
- KI for Health Science and Technology (KIHST), KAIST, Daejeon 34141, Republic of Korea
| | - Jee Myung Yang
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Sangyeul Han
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Tae-Young Chung
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Pilhan Kim
- KI for Health Science and Technology (KIHST), KAIST, Daejeon 34141, Republic of Korea
- Graduate School of Nanoscience and Technology, KAIST, Daejeon 34141, Republic of Korea
| | - Yoshiaki Kubota
- The Laboratory of Vascular Biology, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Hellmut G. Augustin
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim 68167, Germany
- Division of Vascular Oncology and Metastasis, German Cancer Research Center, DKFZ-ZMBH Alliance, Heidelberg 69121, Germany
| | - Wang-Yuhl Oh
- Department of Mechanical Engineering, KAIST, Daejeon 34141, Republic of Korea
- KI for Health Science and Technology (KIHST), KAIST, Daejeon 34141, Republic of Korea
| | - Gou Young Koh
- Center for Vascular Research, Institute for Basic Science (IBS), Daejeon 34141, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
42
|
Cao J, Schnittler H. Putting VE-cadherin into JAIL for junction remodeling. J Cell Sci 2019; 132:132/1/jcs222893. [DOI: 10.1242/jcs.222893] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT
Junction dynamics of endothelial cells are based on the integration of signal transduction, cytoskeletal remodeling and contraction, which are necessary for the formation and maintenance of monolayer integrity, but also enable repair and regeneration. The VE-cadherin–catenin complex forms the molecular basis of the adherence junctions and cooperates closely with actin filaments. Several groups have recently described small actin-driven protrusions at the cell junctions that are controlled by the Arp2/3 complex, contributing to cell junction regulation. We identified these protrusions as the driving force for VE-cadherin dynamics, as they directly induce new VE-cadherin-mediated adhesion sites, and have accordingly referred to these structures as junction-associated intermittent lamellipodia (JAIL). JAIL extend over only a few microns and thus provide the basis for a subcellular regulation of adhesion. The local (subcellular) VE-cadherin concentration and JAIL formation are directly interdependent, which enables autoregulation. Therefore, this mechanism can contribute a subcellularly regulated adaptation of cell contact dynamics, and is therefore of great importance for monolayer integrity and relative cell migration during wound healing and angiogenesis, as well as for inflammatory responses. In this Review, we discuss the mechanisms and functions underlying these actin-driven protrusions and consider their contribution to the dynamic regulation of endothelial cell junctions.
Collapse
Affiliation(s)
- Jiahui Cao
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster Germany
| | - Hans Schnittler
- Institute of Anatomy and Vascular Biology, Westfälische Wilhelms-Universität Münster, Münster Germany
| |
Collapse
|
43
|
Abstract
Tumor blood vessel formation (angiogenesis) is essential for tumor growth and metastasis. Two main endothelial ligand–receptor pathways regulating angiogenesis are vascular endothelial growth factor (VEGF) receptor and angiopoietin-TIE receptor pathways. The angiopoietin-TIE pathway is required for the remodeling and maturation of the blood and lymphatic vessels during embryonic development after VEGF and VEGF-C mediated development of the primary vascular plexus. Angiopoietin-1 (ANGPT1) stabilizes the vasculature after angiogenic processes, via tyrosine kinase with immunoglobulin-like and EGF-like domains 2 (TIE2) activation. In contrast, ANGPT2 is upregulated at sites of vascular remodeling. ANGPT2 is secreted by activated endothelial cells in inflammation, promoting vascular destabilization. ANGPT2 has been found to be expressed in many human cancers. Intriguingly, in preclinical models inhibition of ANGPT2 has provided promising results in preventing tumor angiogenesis, tumor growth, and metastasis, making it an attractive candidate to target in tumors. However, until now the first ANGPT2 targeting therapies have been less effective in clinical trials than in experimental models. Additionally, in preclinical models combined therapy against ANGPT2 and VEGF or immune checkpoint inhibitors has been superior to monotherapies, and these pathways are also targeted in early clinical trials. In order to improve current anti-angiogenic therapies and successfully exploit ANGPT2 as a target for cancer treatment, the biology of the angiopoietin-TIE pathway needs to be profoundly clarified.
Collapse
Affiliation(s)
- Dieter Marmé
- Tumor Biology Center, Freiburg, Baden-Württemberg Germany
| |
Collapse
|
44
|
Lampugnani MG, Dejana E, Giampietro C. Vascular Endothelial (VE)-Cadherin, Endothelial Adherens Junctions, and Vascular Disease. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a029322. [PMID: 28851747 DOI: 10.1101/cshperspect.a029322] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Endothelial cell-cell adherens junctions (AJs) supervise fundamental vascular functions, such as the control of permeability and transmigration of circulating leukocytes, and the maintenance of existing vessels and formation of new ones. These processes are often dysregulated in pathologies. However, the evidence that links dysfunction of endothelial AJs to human pathologies is mostly correlative. In this review, we present an update of the molecular organization of AJ complexes in endothelial cells (ECs) that is mainly based on observations from experimental models. Furthermore, we report in detail on a human pathology, cerebral cavernous malformation (CCM), which is initiated by loss-of-function mutations in the genes that encode the three cytoplasmic components of AJs (CCM1, CCM2, and CCM3). At present, these represent a unique example of mutations in components of endothelial AJs that cause human disease. We describe also how studies into the defects of AJs in CCM are shedding light on the crucial regulatory mechanisms and signaling activities of these endothelial structures. Although these observations are specific for CCM, they support the concept that dysfunction of endothelial AJs can directly contribute to human pathologies.
Collapse
Affiliation(s)
- Maria Grazia Lampugnani
- Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute of Molecular Oncology, 20139 Milan, Italy.,Mario Negri Institute for Pharmacological Research, 20156 Milan, Italy
| | - Elisabetta Dejana
- Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute of Molecular Oncology, 20139 Milan, Italy.,Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
| | - Costanza Giampietro
- Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute of Molecular Oncology, 20139 Milan, Italy
| |
Collapse
|
45
|
Carbajosa G, Malki K, Lawless N, Wang H, Ryder JW, Wozniak E, Wood K, Mein CA, Dobson RJB, Collier DA, O'Neill MJ, Hodges AK, Newhouse SJ. Loss of Trem2 in microglia leads to widespread disruption of cell coexpression networks in mouse brain. Neurobiol Aging 2018; 69:151-166. [PMID: 29906661 PMCID: PMC6075941 DOI: 10.1016/j.neurobiolaging.2018.04.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/26/2018] [Accepted: 04/28/2018] [Indexed: 12/19/2022]
Abstract
Rare heterozygous coding variants in the triggering receptor expressed in myeloid cells 2 (TREM2) gene, conferring increased risk of developing late-onset Alzheimer's disease, have been identified. We examined the transcriptional consequences of the loss of Trem2 in mouse brain to better understand its role in disease using differential expression and coexpression network analysis of Trem2 knockout and wild-type mice. We generated RNA-Seq data from cortex and hippocampus sampled at 4 and 8 months. Using brain cell-type markers and ontology enrichment, we found subnetworks with cell type and/or functional identity. We primarily discovered changes in an endothelial gene-enriched subnetwork at 4 months, including a shift toward a more central role for the amyloid precursor protein gene, coupled with widespread disruption of other cell-type subnetworks, including a subnetwork with neuronal identity. We reveal an unexpected potential role of Trem2 in the homeostasis of endothelial cells that goes beyond its known functions as a microglial receptor and signaling hub, suggesting an underlying link between immune response and vascular disease in dementia.
Collapse
Affiliation(s)
- Guillermo Carbajosa
- Department of Biostatistics and Health Informatics, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK.
| | | | | | - Hong Wang
- Eli Lilly and Company, Indianapolis, IN, USA
| | | | - Eva Wozniak
- Barts and the London Genome Centre, John Vane Science Centre, Barts and the London School of Medicine and Dentistry, London, UK
| | - Kristie Wood
- Barts and the London Genome Centre, John Vane Science Centre, Barts and the London School of Medicine and Dentistry, London, UK
| | - Charles A Mein
- Barts and the London Genome Centre, John Vane Science Centre, Barts and the London School of Medicine and Dentistry, London, UK
| | - Richard J B Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK; NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK; Farr Institute of Health Informatics Research, UCL Institute of Health Informatics, University College London, London, UK
| | | | | | - Angela K Hodges
- Maurice Wohl Clinical Neuroscience Institute James Black Centre Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Stephen J Newhouse
- Department of Biostatistics and Health Informatics, Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK; NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust and King's College London, London, UK; Farr Institute of Health Informatics Research, UCL Institute of Health Informatics, University College London, London, UK
| |
Collapse
|
46
|
Szymborska A, Gerhardt H. Hold Me, but Not Too Tight-Endothelial Cell-Cell Junctions in Angiogenesis. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a029223. [PMID: 28851748 DOI: 10.1101/cshperspect.a029223] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Endothelial cell-cell junctions must perform seemingly incompatible tasks during vascular development-providing stable connections that prevent leakage, while allowing dynamic cellular rearrangements during sprouting, anastomosis, lumen formation, and functional remodeling of the vascular network. This review aims to highlight recent insights into the molecular mechanisms governing endothelial cell-cell adhesion in the context of vascular development.
Collapse
Affiliation(s)
- Anna Szymborska
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin
| | - Holger Gerhardt
- Integrative Vascular Biology Laboratory, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), 13125, Berlin, Germany.,Vascular Patterning Laboratory, Center for Cancer Biology, VIB, Department of Oncology, KU Leuven, 3000 Leuven, Belgium.,DZHK (German Centre for Cardiovascular Research), partner site Berlin.,Berlin Institute of Health (BIH), 10178 Berlin, Germany
| |
Collapse
|
47
|
Rosca AM, Mitroi DN, Cismasiu V, Badea R, Necula-Petrareanu G, Preda MB, Niculite C, Tutuianu R, Szedlacsek S, Burlacu A. Collagen regulates the ability of endothelial progenitor cells to protect hypoxic myocardium through a mechanism involving miR-377/VE-PTP axis. J Cell Mol Med 2018; 22:4700-4708. [PMID: 30044046 PMCID: PMC6156385 DOI: 10.1111/jcmm.13712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/03/2018] [Indexed: 12/25/2022] Open
Abstract
The possibility to employ stem/progenitor cells in the cardiovascular remodelling after myocardial infarction is one of the main queries of regenerative medicine. To investigate whether endothelial progenitor cells (EPCs) participate in the restoration of hypoxia-affected myocardium, we used a co-culture model that allowed the intimate interaction between EPCs and myocardial slices, mimicking stem cell transplantation into the ischaemic heart. On this model, we showed that EPCs engrafted to some extent and only transiently survived into the host tissue, yet produced visible protective effects, in terms of angiogenesis and protection against apoptosis and identified miR-377-VE-PTP axis as being involved in the protective effects of EPCs in hypoxic myocardium. We also showed that collagen, the main component of the myocardial scar, was important for these protective effects by preserving VE-PTP levels, which were otherwise diminished by miR-377. By this, a good face of the scar is revealed, which was so far perceived as having only detrimental impact on the exogenously delivered stem/progenitor cells by affecting not only the engraftment, but also the general protective effects of stem cells.
Collapse
Affiliation(s)
- Ana-Maria Rosca
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Daniel Nicolae Mitroi
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | | | - Rodica Badea
- Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | | | - Mihai Bogdan Preda
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | | | - Raluca Tutuianu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Stefan Szedlacsek
- Institute of Biochemistry of the Romanian Academy, Bucharest, Romania
| | - Alexandrina Burlacu
- Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| |
Collapse
|
48
|
Leligdowicz A, Richard-Greenblatt M, Wright J, Crowley VM, Kain KC. Endothelial Activation: The Ang/Tie Axis in Sepsis. Front Immunol 2018; 9:838. [PMID: 29740443 PMCID: PMC5928262 DOI: 10.3389/fimmu.2018.00838] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 04/05/2018] [Indexed: 12/21/2022] Open
Abstract
Sepsis, a dysregulated host response to infection that causes life-threatening organ dysfunction, is a highly heterogeneous syndrome with no specific treatment. Although sepsis can be caused by a wide variety of pathogenic organisms, endothelial dysfunction leading to vascular leak is a common mechanism of injury that contributes to the morbidity and mortality associated with the syndrome. Perturbations to the angiopoietin (Ang)/Tie2 axis cause endothelial cell activation and contribute to the pathogenesis of sepsis. In this review, we summarize how the Ang/Tie2 pathway is implicated in sepsis and describe its prognostic as well as therapeutic utility in life-threatening infections.
Collapse
Affiliation(s)
- Aleksandra Leligdowicz
- Sandra Rotman Centre for Global Health, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Melissa Richard-Greenblatt
- Sandra Rotman Centre for Global Health, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Julie Wright
- Sandra Rotman Centre for Global Health, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Valerie M Crowley
- Sandra Rotman Centre for Global Health, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Kevin C Kain
- Sandra Rotman Centre for Global Health, University Health Network and University of Toronto, Toronto, ON, Canada
| |
Collapse
|
49
|
Molina-Ortiz P, Orban T, Martin M, Habets A, Dequiedt F, Schurmans S. Rasa3 controls turnover of endothelial cell adhesion and vascular lumen integrity by a Rap1-dependent mechanism. PLoS Genet 2018; 14:e1007195. [PMID: 29381707 PMCID: PMC5806903 DOI: 10.1371/journal.pgen.1007195] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 02/09/2018] [Accepted: 01/09/2018] [Indexed: 11/18/2022] Open
Abstract
Rasa3 is a GTPase activating protein of the GAP1 family which targets R-Ras and Rap1. Although catalytic inactivation or deletion of Rasa3 in mice leads to severe hemorrhages and embryonic lethality, the biological function and cellular location of Rasa3 underlying these defects remains unknown. Here, using a combination of loss of function studies in mouse and zebrafish as well as in vitro cell biology approaches, we identify a key role for Rasa3 in endothelial cells and vascular lumen integrity. Specific ablation of Rasa3 in the mouse endothelium, but not in megakaryocytes and platelets, lead to embryonic bleeding and death at mid-gestation, recapitulating the phenotype observed in full Rasa3 knock-out mice. Reduced plexus/sprouts formation and vascular lumenization defects were observed when Rasa3 was specifically inactivated in mouse endothelial cells at the postnatal or adult stages. Similar results were obtained in zebrafish after decreasing Rasa3 expression. In vitro, depletion of Rasa3 in cultured endothelial cells increased β1 integrin activation and cell adhesion to extracellular matrix components, decreased cell migration and blocked tubulogenesis. During migration, these Rasa3-depleted cells exhibited larger and more mature adhesions resulting from a perturbed dynamics of adhesion assembly and disassembly which significantly increased their life time. These defects were due to a hyperactivation of the Rap1 GTPase and blockade of FAK/Src signaling. Finally, Rasa3-depleted cells showed reduced turnover of VE-cadherin-based adhesions resulting in more stable endothelial cell-cell adhesion and decreased endothelial permeability. Altogether, our results indicate that Rasa3 is a critical regulator of Rap1 in endothelial cells which controls adhesions properties and vascular lumen integrity; its specific endothelial cell inactivation results in occluded blood vessels, hemorrhages and early embryonic death in mouse, mimicking thus the Rasa3-/- mouse phenotype. Because it delivers oxygen and nutriments to every tissue in the body, the vascular system is essential to vertebrate life. Blood vessels consist of a layer of interconnected endothelial cells delineating a luminal space through which blood flows. Formation of vascular lumens is a critical step in vascular development, as vessels should allow unrestricted blood flow while absorbing the pressure from cardiac activity yet retaining flexibility to adapt to homeostatic needs. Our current knowledge of how lumens are established and maintained is still modest and has come essentially from in vitro systems. Here, using a combination of loss of function studies in mouse and zebrafish and in vitro cell biology approaches, we show that Rasa3, a GTPase activating protein of the GAP1 family, controls Rap1 activation, endothelial cell adhesion and migration as well as formation of vascular lumens. We also found that inactivation of Rasa3 specifically in mouse endothelial cells lead to embryonic bleeding and death at mid-gestation, recapitulating the phenotype observed in full Rasa3 knock-out mice.
Collapse
Affiliation(s)
- Patricia Molina-Ortiz
- Laboratory of Functional Genetics, GIGA-Molecular Biology of Disease, University of Liège, Liège, Belgium
| | - Tanguy Orban
- Laboratory of Protein signaling and Interactions Signalisation, GIGA-Molecular Biology of Diseases, University of Liège, Liège, Belgium
| | - Maud Martin
- Laboratory of Functional Genetics, GIGA-Molecular Biology of Disease, University of Liège, Liège, Belgium
- Laboratory of Protein signaling and Interactions Signalisation, GIGA-Molecular Biology of Diseases, University of Liège, Liège, Belgium
| | - Audrey Habets
- Laboratory of Protein signaling and Interactions Signalisation, GIGA-Molecular Biology of Diseases, University of Liège, Liège, Belgium
| | - Franck Dequiedt
- Laboratory of Protein signaling and Interactions Signalisation, GIGA-Molecular Biology of Diseases, University of Liège, Liège, Belgium
| | - Stéphane Schurmans
- Laboratory of Functional Genetics, GIGA-Molecular Biology of Disease, University of Liège, Liège, Belgium
- * E-mail:
| |
Collapse
|
50
|
Morini MF, Giampietro C, Corada M, Pisati F, Lavarone E, Cunha SI, Conze LL, O'Reilly N, Joshi D, Kjaer S, George R, Nye E, Ma A, Jin J, Mitter R, Lupia M, Cavallaro U, Pasini D, Calado DP, Dejana E, Taddei A. VE-Cadherin-Mediated Epigenetic Regulation of Endothelial Gene Expression. Circ Res 2018; 122:231-245. [PMID: 29233846 PMCID: PMC5771688 DOI: 10.1161/circresaha.117.312392] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 11/30/2017] [Accepted: 12/11/2016] [Indexed: 01/15/2023]
Abstract
RATIONALE The mechanistic foundation of vascular maturation is still largely unknown. Several human pathologies are characterized by deregulated angiogenesis and unstable blood vessels. Solid tumors, for instance, get their nourishment from newly formed structurally abnormal vessels which present wide and irregular interendothelial junctions. Expression and clustering of the main endothelial-specific adherens junction protein, VEC (vascular endothelial cadherin), upregulate genes with key roles in endothelial differentiation and stability. OBJECTIVE We aim at understanding the molecular mechanisms through which VEC triggers the expression of a set of genes involved in endothelial differentiation and vascular stabilization. METHODS AND RESULTS We compared a VEC-null cell line with the same line reconstituted with VEC wild-type cDNA. VEC expression and clustering upregulated endothelial-specific genes with key roles in vascular stabilization including claudin-5, vascular endothelial-protein tyrosine phosphatase (VE-PTP), and von Willebrand factor (vWf). Mechanistically, VEC exerts this effect by inhibiting polycomb protein activity on the specific gene promoters. This is achieved by preventing nuclear translocation of FoxO1 (Forkhead box protein O1) and β-catenin, which contribute to PRC2 (polycomb repressive complex-2) binding to promoter regions of claudin-5, VE-PTP, and vWf. VEC/β-catenin complex also sequesters a core subunit of PRC2 (Ezh2 [enhancer of zeste homolog 2]) at the cell membrane, preventing its nuclear translocation. Inhibition of Ezh2/VEC association increases Ezh2 recruitment to claudin-5, VE-PTP, and vWf promoters, causing gene downregulation. RNA sequencing comparison of VEC-null and VEC-positive cells suggested a more general role of VEC in activating endothelial genes and triggering a vascular stability-related gene expression program. In pathological angiogenesis of human ovarian carcinomas, reduced VEC expression paralleled decreased levels of claudin-5 and VE-PTP. CONCLUSIONS These data extend the knowledge of polycomb-mediated regulation of gene expression to endothelial cell differentiation and vessel maturation. The identified mechanism opens novel therapeutic opportunities to modulate endothelial gene expression and induce vascular normalization through pharmacological inhibition of the polycomb-mediated repression system.
Collapse
Affiliation(s)
- Marco F Morini
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Costanza Giampietro
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Monica Corada
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Federica Pisati
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Elisa Lavarone
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Sara I Cunha
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Lei L Conze
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Nicola O'Reilly
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Dhira Joshi
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Svend Kjaer
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Roger George
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Emma Nye
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Anqi Ma
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Jian Jin
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Richard Mitter
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Michela Lupia
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Ugo Cavallaro
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Diego Pasini
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Dinis P Calado
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.)
| | - Elisabetta Dejana
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.).
| | - Andrea Taddei
- From the IFOM, FIRC Institute of Molecular Oncology, Milan, Italy (M.F.M., C.G., M.C., F.P., E.D., A.T.); Department of Biomedicine, University of Basel, Switzerland (M.F.M.); Laboratory of Thermodynamics in Emerging Technologies, Department of Mechanical and Process Engineering, ETH Zurich, Switzerland (C.G.); Cogentech, Milan, Italy (F.P.); Department of Experimental Oncology (E.L., D.P.) and Unit of Gynecological Oncology Research (M.L., U.C.), European Institute of Oncology, Milan, Italy; Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (S.I.C., L.L.C., E.D.); Peptide Chemistry (N.O., D.J.), Structural Biology (S.K., R.G.), Experimental Histopathology (E.N.), Bioinformatics & Biostatistics Department (R.M.), and Immunity and Cancer Laboratory (D.P.C., A.T.), The Francis Crick Institute, London, United Kingdom; Center for Chemical Biology and Drug Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY (A.M., J.J.); and Department of Oncology and Hemato-Oncology, University of Milan, Italy (E.D.).
| |
Collapse
|