51
|
Lee Y, Chakraborty S, Meininger CJ, Muthuchamy M. Insulin resistance disrupts cell integrity, mitochondrial function, and inflammatory signaling in lymphatic endothelium. Microcirculation 2018; 25:e12492. [PMID: 30025187 DOI: 10.1111/micc.12492] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 07/09/2018] [Accepted: 07/16/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Lymphatic vessel dysfunction and increased lymph leakage have been directly associated with several metabolic diseases. However, the underlying cellular mechanisms causing lymphatic dysfunction have not been determined. Aberrant insulin signaling affects the metabolic function of cells and consequently impairs tissue function. We hypothesized that insulin resistance in LECs decreases eNOS activity, disrupts barrier integrity increases permeability, and activates mitochondrial dysfunction and pro-inflammatory signaling pathways. METHODS LECs were treated with insulin and/or glucose to determine the mechanisms leading to insulin resistance. RESULTS Acute insulin treatment increased eNOS phosphorylation and NO production in LECs via activation of the PI3K/Akt signaling pathway. Prolonged hyperglycemia and hyperinsulinemia induced insulin resistance in LECs. Insulin-resistant LECs produced less NO due to a decrease in eNOS phosphorylation and showed a significant decrease in impedance across an LEC monolayer that was associated with disruption in the adherence junctional proteins. Additionally, insulin resistance in LECs impaired mitochondrial function by decreasing basal-, maximal-, and ATP-linked OCRs and activated NF-κB nuclear translocation coupled with increased pro-inflammatory signaling. CONCLUSION Our data provide the first evidence that insulin resistance disrupts endothelial barrier integrity, decreases eNOS phosphorylation and mitochondrial function, and activates inflammation in LECs.
Collapse
Affiliation(s)
- Yang Lee
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, Texas
| | - Sanjukta Chakraborty
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, Texas
| | - Cynthia J Meininger
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, Texas
| | - Mariappan Muthuchamy
- Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, Texas
| |
Collapse
|
52
|
Debordeaux F, Chansel-Debordeaux L, Pinaquy JB, Fernandez P, Schulz J. What about αvβ3 integrins in molecular imaging in oncology? Nucl Med Biol 2018; 62-63:31-46. [DOI: 10.1016/j.nucmedbio.2018.04.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/19/2018] [Accepted: 04/30/2018] [Indexed: 10/17/2022]
|
53
|
Halder SK, Kant R, Milner R. Chronic mild hypoxia promotes profound vascular remodeling in spinal cord blood vessels, preferentially in white matter, via an α5β1 integrin-mediated mechanism. Angiogenesis 2018; 21:251-266. [PMID: 29299782 DOI: 10.1007/s10456-017-9593-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 12/26/2017] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) leads to rapid destruction of neuronal tissue, resulting in devastating motor and sensory deficits. This is exacerbated by damage to spinal cord blood vessels and loss of vascular integrity. Thus, approaches that protect existing blood vessels or stimulate the growth of new blood vessels might present a novel approach to minimize loss or promote regeneration of spinal cord tissue following SCI. In light of the remarkable power of chronic mild hypoxia (CMH) to stimulate vascular remodeling in the brain, the goal of this study was to examine how CMH (8% O2 for up to 7 days) affects blood vessel remodeling in the spinal cord. We found that CMH promoted the following: (1) endothelial proliferation and increased vascularity as a result of angiogenesis and arteriogenesis, (2) increased vascular expression of the angiogenic extracellular matrix protein fibronectin as well as concomitant increases in endothelial expression of the fibronectin receptor α5β1 integrin, (3) strongly upregulated endothelial expression of the tight junction proteins claudin-5, ZO-1 and occludin and (4) astrocyte activation. Of note, the vascular remodeling changes induced by CMH were more extensive in white matter. Interestingly, hypoxic-induced vascular remodeling in spinal cord blood vessels was markedly attenuated in mice lacking endothelial α5 integrin expression (α5-EC-KO mice). Taken together, these studies demonstrate the considerable remodeling potential of spinal cord blood vessels and highlight an important angiogenic role for the α5β1 integrin in promoting endothelial proliferation. They also imply that stimulation of the α5β1 integrin or controlled use of mild hypoxia might provide new approaches for promoting angiogenesis and improving vascular integrity in spinal cord blood vessels.
Collapse
Affiliation(s)
- Sebok K Halder
- Department of Molecular Medicine, MEM-132, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Ravi Kant
- Department of Molecular Medicine, MEM-132, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Richard Milner
- Department of Molecular Medicine, MEM-132, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|
54
|
Turner CJ, Badu-Nkansah K, Hynes RO. Endothelium-derived fibronectin regulates neonatal vascular morphogenesis in an autocrine fashion. Angiogenesis 2017; 20:519-531. [PMID: 28667352 PMCID: PMC5660148 DOI: 10.1007/s10456-017-9563-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 06/21/2017] [Indexed: 12/02/2022]
Abstract
Fibronectin containing alternatively spliced EIIIA and EIIIB domains is largely absent from mature quiescent vessels in adults, but is highly expressed around blood vessels during developmental and pathological angiogenesis. The precise functions of fibronectin and its splice variants during developmental angiogenesis however remain unclear due to the presence of cardiac, somitic, mesodermal and neural defects in existing global fibronectin KO mouse models. Using a rare family of surviving EIIIA EIIIB double KO mice, as well as inducible endothelial-specific fibronectin-deficient mutant mice, we show that vascular development in the neonatal retina is regulated in an autocrine manner by endothelium-derived fibronectin, and requires both EIIIA and EIIIB domains and the RGD-binding α5 and αv integrins for its function. Exogenous sources of fibronectin do not fully substitute for the autocrine function of endothelial fibronectin, demonstrating that fibronectins from different sources contribute differentially to specific aspects of angiogenesis.
Collapse
Affiliation(s)
- Christopher J Turner
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, 76-361, Cambridge, MA, 02139, USA
- University of Suffolk, James Hehir Building, University Avenue, Ipswich, Suffolk, IP3 0FS, UK
| | - Kwabena Badu-Nkansah
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, 76-361, Cambridge, MA, 02139, USA
- Duke University Medical Center, 307 Research Drive, Durham, NC, 27710, USA
| | - Richard O Hynes
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Ave, 76-361, Cambridge, MA, 02139, USA.
| |
Collapse
|
55
|
Exploring the Role of RGD-Recognizing Integrins in Cancer. Cancers (Basel) 2017; 9:cancers9090116. [PMID: 28869579 PMCID: PMC5615331 DOI: 10.3390/cancers9090116] [Citation(s) in RCA: 313] [Impact Index Per Article: 39.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/28/2017] [Accepted: 08/31/2017] [Indexed: 12/18/2022] Open
Abstract
Integrins are key regulators of communication between cells and with their microenvironment. Eight members of the integrin superfamily recognize the tripeptide motif Arg-Gly-Asp (RGD) within extracelluar matrix (ECM) proteins. These integrins constitute an important subfamily and play a major role in cancer progression and metastasis via their tumor biological functions. Such transmembrane adhesion and signaling receptors are thus recognized as promising and well accessible targets for novel diagnostic and therapeutic applications for directly attacking cancer cells and their fatal microenvironment. Recently, specific small peptidic and peptidomimetic ligands as well as antibodies binding to distinct integrin subtypes have been developed and synthesized as new drug candidates for cancer treatment. Understanding the distinct functions and interplay of integrin subtypes is a prerequisite for selective intervention in integrin-mediated diseases. Integrin subtype-specific ligands labelled with radioisotopes or fluorescent molecules allows the characterization of the integrin patterns in vivo and later the medical intervention via subtype specific drugs. The coating of nanoparticles, larger proteins, or encapsulating agents by integrin ligands are being explored to guide cytotoxic reagents directly to the cancer cell surface. These ligands are currently under investigation in clinical studies for their efficacy in interference with tumor cell adhesion, migration/invasion, proliferation, signaling, and survival, opening new treatment approaches in personalized medicine.
Collapse
|
56
|
Lu S, Liu S, Wietelmann A, Kojonazarov B, Atzberger A, Tang C, Schermuly RT, Gröne HJ, Offermanns S. Developmental vascular remodeling defects and postnatal kidney failure in mice lacking Gpr116 (Adgrf5) and Eltd1 (Adgrl4). PLoS One 2017; 12:e0183166. [PMID: 28806758 PMCID: PMC5555693 DOI: 10.1371/journal.pone.0183166] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 07/31/2017] [Indexed: 11/18/2022] Open
Abstract
GPR116 (ADGRF5) and ELTD1 (ADGRL4) belong to different subfamilies of the adhesion G-protein-coupled receptor group but are both expressed in endothelial cells. We therefore analyzed their functions in mice lacking these receptors. While loss of GPR116 or ELTD1 alone had no obvious effect on cardiovascular or kidney function, mice lacking both, GPR116 and ELTD1, showed malformations of the aortic arch arteries and the cardiac outflow tract leading to perinatal lethality in about 50% of the mutants. In addition to cardiovascular malformations, surviving mice developed renal thrombotic microangiopathy as well as hemolysis and splenomegaly, and their lifespan was significantly reduced. Loss of GPR116 and ELTD1 specifically in endothelial cells or neural crest-derived cells did not recapitulate any of the phenotypes observed in GPR116-ELTD1 double deficient mice, indicating that loss of GPR116 and ELTD1 expressed by other cells accounts for the observed cardiovascular and renal defects.
Collapse
Affiliation(s)
- Shun Lu
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail: (SL); (SO)
| | - Shuya Liu
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Astrid Wietelmann
- Scientific Service Group Nuclear Magnetic Resonance Imaging, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Baktybek Kojonazarov
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Ann Atzberger
- Flow Cytometry Service Facility, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Cong Tang
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ralph Theo Schermuly
- Department of Internal Medicine, Justus-Liebig-University Giessen, Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Hermann-Josef Gröne
- Department of Cellular and Molecular Pathology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan Offermanns
- Department of Pharmacology, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Medical Faculty, J.W. Goethe University Frankfurt, Frankfurt, Germany
- * E-mail: (SL); (SO)
| |
Collapse
|
57
|
Genova T, Grolez GP, Camillo C, Bernardini M, Bokhobza A, Richard E, Scianna M, Lemonnier L, Valdembri D, Munaron L, Philips MR, Mattot V, Serini G, Prevarskaya N, Gkika D, Pla AF. TRPM8 inhibits endothelial cell migration via a non-channel function by trapping the small GTPase Rap1. J Cell Biol 2017; 216:2107-2130. [PMID: 28550110 PMCID: PMC5496606 DOI: 10.1083/jcb.201506024] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 06/26/2016] [Accepted: 04/12/2017] [Indexed: 01/30/2023] Open
Abstract
Endothelial cell adhesion and migration are critical steps of the angiogenic process, whose dysfunction is associated with tumor growth and metastasis. The TRPM8 channel has recently been proposed to play a protective role in prostate cancer by impairing cell motility. However, the mechanisms by which it could influence vascular behavior are unknown. Here, we reveal a novel non-channel function for TRPM8 that unexpectedly acts as a Rap1 GTPase inhibitor, thereby inhibiting endothelial cell motility, independently of pore function. TRPM8 retains Rap1 intracellularly through direct protein-protein interaction, thus preventing its cytoplasm-plasma membrane trafficking. In turn, this mechanism impairs the activation of a major inside-out signaling pathway that triggers the conformational activation of integrin and, consequently, cell adhesion, migration, in vitro endothelial tube formation, and spheroid sprouting. Our results bring to light a novel, pore-independent molecular mechanism by which endogenous TRPM8 expression inhibits Rap1 GTPase and thus plays a critical role in the behavior of vascular endothelial cells by inhibiting migration.
Collapse
Affiliation(s)
- Tullio Genova
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy.,Department of Surgical Sciences, C.I.R. Dental School, University of Torino, Torino, Italy
| | - Guillaume P Grolez
- Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Chiara Camillo
- Laboratory of Cell Adhesion Dynamics, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia, Istituto di Ricovero e Cura a Carattere Scientifico, Department of Oncology, University of Torino School of Medicine, Candiolo, Italy
| | - Michela Bernardini
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy.,Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Alexandre Bokhobza
- Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Elodie Richard
- BICeL Campus Lille1, FR3688 FRABio, Université de Lille, Villeneuve d'Ascq, France
| | - Marco Scianna
- Department of Mathematical Sciences, Politecnico di Torino, Torino, Italy
| | - Loic Lemonnier
- Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Donatella Valdembri
- Laboratory of Cell Adhesion Dynamics, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia, Istituto di Ricovero e Cura a Carattere Scientifico, Department of Oncology, University of Torino School of Medicine, Candiolo, Italy
| | - Luca Munaron
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy.,Nanostructured Interfaces and Surfaces Centre of Excellence, University of Torino, Torino, Italy
| | - Mark R Philips
- Cancer Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY
| | - Virginie Mattot
- Centre National de la Recherche Scientifique, Institut Pasteur de Lille, UMR 8161 - Mechanisms of Tumorigenesis and Target Therapies, Universite de Lille, Lille, France
| | - Guido Serini
- Laboratory of Cell Adhesion Dynamics, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia, Istituto di Ricovero e Cura a Carattere Scientifico, Department of Oncology, University of Torino School of Medicine, Candiolo, Italy
| | - Natalia Prevarskaya
- Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Dimitra Gkika
- Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| | - Alessandra Fiorio Pla
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy .,Nanostructured Interfaces and Surfaces Centre of Excellence, University of Torino, Torino, Italy.,Laboratoire de Physiologie cellulaire, Institut National de la Santé et de la Recherche Médicale U1003, Laboratory of Excellence, Ion Channels Science and Therapeutics, Université de Lille, Villeneuve d'Ascq, France
| |
Collapse
|
58
|
Kwan BH, Zhu EF, Tzeng A, Sugito HR, Eltahir AA, Ma B, Delaney MK, Murphy PA, Kauke MJ, Angelini A, Momin N, Mehta NK, Maragh AM, Hynes RO, Dranoff G, Cochran JR, Wittrup KD. Integrin-targeted cancer immunotherapy elicits protective adaptive immune responses. J Exp Med 2017; 214:1679-1690. [PMID: 28473400 PMCID: PMC5460993 DOI: 10.1084/jem.20160831] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 09/25/2016] [Accepted: 03/23/2017] [Indexed: 01/02/2023] Open
Abstract
Integrin targeting for cancer has primarily focused on antagonizing integrin function, which has been clinically ineffective to date. In this study, Kwan et al. repurpose integrins as a beacon for recruiting immune effector functions to bolster current cancer immunotherapy approaches. Certain RGD-binding integrins are required for cell adhesion, migration, and proliferation and are overexpressed in most tumors, making them attractive therapeutic targets. However, multiple integrin antagonist drug candidates have failed to show efficacy in cancer clinical trials. In this work, we instead exploit these integrins as a target for antibody Fc effector functions in the context of cancer immunotherapy. By combining administration of an engineered mouse serum albumin/IL-2 fusion with an Fc fusion to an integrin-binding peptide (2.5F-Fc), significant survival improvements are achieved in three syngeneic mouse tumor models, including complete responses with protective immunity. Functional integrin antagonism does not contribute significantly to efficacy; rather, this therapy recruits both an innate and adaptive immune response, as deficiencies in either arm result in reduced tumor control. Administration of this integrin-targeted immunotherapy together with an anti–PD-1 antibody further improves responses and predominantly results in cures. Overall, this well-tolerated therapy achieves tumor specificity by redirecting inflammation to a functional target fundamental to tumorigenic processes but expressed at significantly lower levels in healthy tissues, and it shows promise for translation.
Collapse
Affiliation(s)
- Byron H Kwan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Eric F Zhu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Alice Tzeng
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Harun R Sugito
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Ahmed A Eltahir
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Botong Ma
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Mary K Delaney
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Patrick A Murphy
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Monique J Kauke
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Alessandro Angelini
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Noor Momin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Naveen K Mehta
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Alecia M Maragh
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139.,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Richard O Hynes
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Glenn Dranoff
- Novartis Institutes for BioMedical Research, Cambridge, MA 02139
| | - Jennifer R Cochran
- Department of Bioengineering, Stanford University, Stanford, CA 94305.,Department of Chemical Engineering, Stanford University, Stanford, CA 94305
| | - K Dane Wittrup
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 .,Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139
| |
Collapse
|
59
|
Gundry C, Marco S, Rainero E, Miller B, Dornier E, Mitchell L, Caswell PT, Campbell AD, Hogeweg A, Sansom OJ, Morton JP, Norman JC. Phosphorylation of Rab-coupling protein by LMTK3 controls Rab14-dependent EphA2 trafficking to promote cell:cell repulsion. Nat Commun 2017; 8:14646. [PMID: 28294115 PMCID: PMC5355957 DOI: 10.1038/ncomms14646] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/18/2017] [Indexed: 12/21/2022] Open
Abstract
The Rab GTPase effector, Rab-coupling protein (RCP) is known to promote invasive behaviour in vitro by controlling integrin and receptor tyrosine kinase (RTK) trafficking, but how RCP influences metastasis in vivo is unclear. Here we identify an RTK of the Eph family, EphA2, to be a cargo of an RCP-regulated endocytic pathway which controls cell:cell repulsion and metastasis in vivo. Phosphorylation of RCP at Ser435 by Lemur tyrosine kinase-3 (LMTK3) and of EphA2 at Ser897 by Akt are both necessary to promote Rab14-dependent (and Rab11-independent) trafficking of EphA2 which generates cell:cell repulsion events that drive tumour cells apart. Genetic disruption of RCP or EphA2 opposes cell:cell repulsion and metastasis in an autochthonous mouse model of pancreatic adenocarcinoma-whereas conditional knockout of another RCP cargo, α5 integrin, does not suppress pancreatic cancer metastasis-indicating a role for RCP-dependent trafficking of an Eph receptor to drive tumour dissemination in vivo.
Collapse
Affiliation(s)
- Christine Gundry
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Sergi Marco
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Elena Rainero
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Bryan Miller
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Emmanuel Dornier
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Louise Mitchell
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Patrick T. Caswell
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
- Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Manchester M13 9PT, UK
| | - Andrew D. Campbell
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Anna Hogeweg
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Owen J. Sansom
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Jennifer P. Morton
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Jim C. Norman
- CRUK Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| |
Collapse
|
60
|
Dagher Z, Gerhardinger C, Vaz J, Goodridge M, Tecilazich F, Lorenzi M. The Increased Transforming Growth Factor-β Signaling Induced by Diabetes Protects Retinal Vessels. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:627-638. [PMID: 28162229 PMCID: PMC5397667 DOI: 10.1016/j.ajpath.2016.11.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 11/08/2016] [Accepted: 11/15/2016] [Indexed: 12/19/2022]
Abstract
The roles of transforming growth factor (TGF)-β in extracellular matrix production and vascular remodeling, coupled with increased TGF-β expression and signaling in diabetes, suggest TGF-β as an important contributor to the microangiopathy of diabetic retinopathy and nephropathy. To investigate whether increased TGF-β signaling could be a therapeutic target for preventing retinopathy, we used a pharmacologic approach (SM16, a selective inhibitor of the type 1 TGF-β receptor activin receptor-like kinase 5, orally active) to inhibit the increased, but not the basal, Tgf-β signaling in retinal vessels of diabetic rats. At the level of vascular gene expression, 3.5 months' diabetes induced minimal changes. Diabetes + SM16 for 3 weeks caused widespread changes in gene expression poised to enhance vascular inflammation, thrombosis, leakage, and wall instability; these changes were not observed in control rats given SM16. The synergy of diabetes and SM16 in altering gene expression was not observed in the lung. At the level of vascular network morphology, 7 months' diabetes induced no detectable changes. Diabetes + SM16 for 3 weeks caused instead distorted morphology and decreased density. Thus, in diabetes, retinal vessels become dependent on a small increase in TGF-β signaling via activin receptor-like kinase 5 to maintain early integrity. The increased TGF-β signaling may protect against rapid retinopathy progression and should not be a target of inhibitory interventions.
Collapse
Affiliation(s)
- Zeina Dagher
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Chiara Gerhardinger
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Joseph Vaz
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Michael Goodridge
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts
| | - Francesco Tecilazich
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts
| | - Mara Lorenzi
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Boston, Massachusetts; Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
61
|
Integrin signaling in atherosclerosis. Cell Mol Life Sci 2017; 74:2263-2282. [PMID: 28246700 DOI: 10.1007/s00018-017-2490-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/24/2017] [Accepted: 02/15/2017] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic lipid-driven inflammatory disease affecting large arteries, represents the primary cause of cardiovascular disease in the world. The local remodeling of the vessel intima during atherosclerosis involves the modulation of vascular cell phenotype, alteration of cell migration and proliferation, and propagation of local extracellular matrix remodeling. All of these responses represent targets of the integrin family of cell adhesion receptors. As such, alterations in integrin signaling affect multiple aspects of atherosclerosis, from the earliest induction of inflammation to the development of advanced fibrotic plaques. Integrin signaling has been shown to regulate endothelial phenotype, facilitate leukocyte homing, affect leukocyte function, and drive smooth muscle fibroproliferative remodeling. In addition, integrin signaling in platelets contributes to the thrombotic complications that typically drive the clinical manifestation of cardiovascular disease. In this review, we examine the current literature on integrin regulation of atherosclerotic plaque development and the suitability of integrins as potential therapeutic targets to limit cardiovascular disease and its complications.
Collapse
|
62
|
Kapp TG, Rechenmacher F, Neubauer S, Maltsev OV, Cavalcanti-Adam EA, Zarka R, Reuning U, Notni J, Wester HJ, Mas-Moruno C, Spatz J, Geiger B, Kessler H. A Comprehensive Evaluation of the Activity and Selectivity Profile of Ligands for RGD-binding Integrins. Sci Rep 2017; 7:39805. [PMID: 28074920 PMCID: PMC5225454 DOI: 10.1038/srep39805] [Citation(s) in RCA: 429] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/25/2016] [Indexed: 12/12/2022] Open
Abstract
Integrins, a diverse class of heterodimeric cell surface receptors, are key regulators of cell structure and behaviour, affecting cell morphology, proliferation, survival and differentiation. Consequently, mutations in specific integrins, or their deregulated expression, are associated with a variety of diseases. In the last decades, many integrin-specific ligands have been developed and used for modulation of integrin function in medical as well as biophysical studies. The IC50-values reported for these ligands strongly vary and are measured using different cell-based and cell-free systems. A systematic comparison of these values is of high importance for selecting the optimal ligands for given applications. In this study, we evaluate a wide range of ligands for their binding affinity towards the RGD-binding integrins αvβ3, αvβ5, αvβ6, αvβ8, α5β1, αIIbβ3, using homogenous ELISA-like solid phase binding assay.
Collapse
Affiliation(s)
- Tobias G Kapp
- Institute for Advanced Study and Center for Integrated Protein Science, Department of Chemistry, Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
| | - Florian Rechenmacher
- Institute for Advanced Study and Center for Integrated Protein Science, Department of Chemistry, Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
| | - Stefanie Neubauer
- Institute for Advanced Study and Center for Integrated Protein Science, Department of Chemistry, Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
| | - Oleg V Maltsev
- Institute for Advanced Study and Center for Integrated Protein Science, Department of Chemistry, Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
| | - Elisabetta A Cavalcanti-Adam
- Max-Planck-Institute for Medical Research, Department of Biointerface Science and Technology, Heidelberg, Postal address: Heisenbergstr. 3, 70 569 Stuttgart, Germany
| | - Revital Zarka
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics &Gynecology, Technische Universität München, Ismaninger Strasse 22, 81675 Munich, Germany
| | - Johannes Notni
- Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany
| | - Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering, and Centre for Research in NanoEngineering (CRNE), Technical University of Catalonia, 08028-Barcelona, Spain
| | - Joachim Spatz
- Max-Planck-Institute for Medical Research, Department of Biointerface Science and Technology, Heidelberg, Postal address: Heisenbergstr. 3, 70 569 Stuttgart, Germany
| | - Benjamin Geiger
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Horst Kessler
- Institute for Advanced Study and Center for Integrated Protein Science, Department of Chemistry, Technische Universität München, Lichtenbergstr. 4, 85747 Garching, Germany
| |
Collapse
|
63
|
Roberts J, de Hoog L, Bix GJ. Mice deficient in endothelial α5 integrin are profoundly resistant to experimental ischemic stroke. J Cereb Blood Flow Metab 2017; 37:85-96. [PMID: 26661237 PMCID: PMC5363730 DOI: 10.1177/0271678x15616979] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/27/2015] [Accepted: 09/27/2015] [Indexed: 11/16/2022]
Abstract
Stroke is a disease in dire need of better therapies. We have previously shown that a fragment of the extracellular matrix proteoglycan, perlecan, has beneficial effects following cerebral ischemia via the α5β1 integrin receptor. We now report that endothelial cell selective α5 integrin deficient mice (α5 KO) are profoundly resistant to ischemic infarct after transient middle cerebral artery occlusion. Specifically, α5 KOs had little to no infarct 2-3 days post-stroke, whereas controls had an increase in mean infarct volume over the same time period as expected. Functional outcome is also improved in the α5 KOs compared with controls. Importantly, no differences in cerebrovascular anatomy or collateral blood flow were noted that could account for this difference in ischemic injury. Rather, we demonstrate that α5 KOs have increased blood-brain barrier integrity (increased expression of claudin-5, and absent brain parenchymal IgG extravasation) after stroke compared with controls, which could explain their resistance to ischemic injury. Additionally, inhibition of α5 integrin in vitro leads to decreased permeability of brain endothelial cells following oxygen-glucose deprivation. Together, these findings indicate endothelial cell α5 integrin plays an important role in stroke outcome and blood-brain barrier integrity, suggesting that α5 integrin could be a novel therapeutic target for stroke.
Collapse
Affiliation(s)
- Jill Roberts
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA.,Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY, USA
| | - Leon de Hoog
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| | - Gregory J Bix
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA .,Department of Anatomy and Neurobiology, University of Kentucky, Lexington, KY, USA.,Department of Neurology, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
64
|
Di Russo J, Hannocks MJ, Luik AL, Song J, Zhang X, Yousif L, Aspite G, Hallmann R, Sorokin L. Vascular laminins in physiology and pathology. Matrix Biol 2017; 57-58:140-148. [DOI: 10.1016/j.matbio.2016.06.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/28/2016] [Indexed: 12/11/2022]
|
65
|
Pashuck ET, Duchet BJR, Hansel CS, Maynard SA, Chow LW, Stevens MM. Controlled Sub-Nanometer Epitope Spacing in a Three-Dimensional Self-Assembled Peptide Hydrogel. ACS NANO 2016; 10:11096-11104. [PMID: 28024362 DOI: 10.1021/acsnano.6b05975] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Cells in the body use a variety of mechanisms to ensure the specificity and efficacy of signal transduction. One way that this is achieved is through tight spatial control over the position of different proteins, signaling sequences, and biomolecules within and around cells. For instance, the extracellular matrix protein fibronectin presents RGDS and PHSRN sequences that synergistically bind the α5β1 integrin when separated by 3.2 nm but are unable to bind when this distance is >5.5 nm.1 Building biomaterials to controllably space different epitopes with subnanometer accuracy in a three-dimensional (3D) hydrogel is challenging. Here, we synthesized peptides that self-assemble into nanofiber hydrogels utilizing the β-sheet motif, which has a known regular spacing along the peptide backbone. By modifying specific locations along the peptide, we are able to controllably space different epitopes with subnanometer accuracy at distances from 0.7 nm to over 6 nm, which is within the size range of many protein clusters. Endothelial cells encapsulated within hydrogels displaying RGDS and PHSRN in the native 3.2 nm spacing showed a significant upregulation in the expression of the alpha 5 integrin subunit compared to those in hydrogels with a 6.2 nm spacing, demonstrating the physiological relevance of the spacing. Furthermore, after 24 h the cells in hydrogels with the 3.2 nm spacing appeared to be more spread with increased staining for the α5β1 integrin. This self-assembling peptide system can controllably space multiple epitopes with subnanometer accuracy, demonstrating an exciting platform to study the effects of ligand density and location on cells within a synthetic 3D environment.
Collapse
Affiliation(s)
- E Thomas Pashuck
- Department of Materials, ‡Department of Bioengineering, §Institute of Biomedical Engineering, and ⊥Department of Chemistry, Imperial College London , London SW7 2AZ, United Kingdom
| | - Benoît J R Duchet
- Department of Materials, ‡Department of Bioengineering, §Institute of Biomedical Engineering, and ⊥Department of Chemistry, Imperial College London , London SW7 2AZ, United Kingdom
| | - Catherine S Hansel
- Department of Materials, ‡Department of Bioengineering, §Institute of Biomedical Engineering, and ⊥Department of Chemistry, Imperial College London , London SW7 2AZ, United Kingdom
| | - Stephanie A Maynard
- Department of Materials, ‡Department of Bioengineering, §Institute of Biomedical Engineering, and ⊥Department of Chemistry, Imperial College London , London SW7 2AZ, United Kingdom
| | - Lesley W Chow
- Department of Materials, ‡Department of Bioengineering, §Institute of Biomedical Engineering, and ⊥Department of Chemistry, Imperial College London , London SW7 2AZ, United Kingdom
| | - Molly M Stevens
- Department of Materials, ‡Department of Bioengineering, §Institute of Biomedical Engineering, and ⊥Department of Chemistry, Imperial College London , London SW7 2AZ, United Kingdom
| |
Collapse
|
66
|
Di Russo J, Luik AL, Yousif L, Budny S, Oberleithner H, Hofschröer V, Klingauf J, van Bavel E, Bakker EN, Hellstrand P, Bhattachariya A, Albinsson S, Pincet F, Hallmann R, Sorokin LM. Endothelial basement membrane laminin 511 is essential for shear stress response. EMBO J 2016; 36:183-201. [PMID: 27940654 PMCID: PMC5239996 DOI: 10.15252/embj.201694756] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 11/09/2022] Open
Abstract
Shear detection and mechanotransduction by arterial endothelium requires junctional complexes containing PECAM-1 and VE-cadherin, as well as firm anchorage to the underlying basement membrane. While considerable information is available for junctional complexes in these processes, gained largely from in vitro studies, little is known about the contribution of the endothelial basement membrane. Using resistance artery explants, we show that the integral endothelial basement membrane component, laminin 511 (laminin α5), is central to shear detection and mechanotransduction and its elimination at this site results in ablation of dilation in response to increased shear stress. Loss of endothelial laminin 511 correlates with reduced cortical stiffness of arterial endothelium in vivo, smaller integrin β1-positive/vinculin-positive focal adhesions, and reduced junctional association of actin-myosin II In vitro assays reveal that β1 integrin-mediated interaction with laminin 511 results in high strengths of adhesion, which promotes p120 catenin association with VE-cadherin, stabilizing it at cell junctions and increasing cell-cell adhesion strength. This highlights the importance of endothelial laminin 511 in shear response in the physiologically relevant context of resistance arteries.
Collapse
Affiliation(s)
- Jacopo Di Russo
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Anna-Liisa Luik
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Lema Yousif
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Sigmund Budny
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Hans Oberleithner
- Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany.,Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Verena Hofschröer
- Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany.,Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Juergen Klingauf
- Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany.,Institute of Medical Physics, University of Muenster, Muenster, Germany
| | - Ed van Bavel
- Biomedical Engineering and Physics, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Erik Ntp Bakker
- Biomedical Engineering and Physics, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Per Hellstrand
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | | - Frederic Pincet
- Laboratoire de Physique Statistique, École Normale Superieure - PSL Research University, Paris, France.,CNRS UMR8550, Sorbonne Universités - UPMC Univ Paris 06, Université Paris, Paris, France
| | - Rupert Hallmann
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany.,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| | - Lydia M Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Muenster, Germany .,Cells-in-Motion Cluster of Excellence, University of Muenster, Muenster, Germany
| |
Collapse
|
67
|
Ghatak S, Niland S, Schulz JN, Wang F, Eble JA, Leitges M, Mauch C, Krieg T, Zigrino P, Eckes B. Role of Integrins α1β1 and α2β1 in Wound and Tumor Angiogenesis in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:3011-3027. [DOI: 10.1016/j.ajpath.2016.06.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 06/01/2016] [Accepted: 06/30/2016] [Indexed: 12/11/2022]
|
68
|
Gomez Perdiguero E, Liabotis-Fontugne A, Durand M, Faye C, Ricard-Blum S, Simonutti M, Augustin S, Robb BM, Paques M, Valenzuela DM, Murphy AJ, Yancopoulos GD, Thurston G, Galaup A, Monnot C, Germain S. ANGPTL4-αvβ3 interaction counteracts hypoxia-induced vascular permeability by modulating Src signalling downstream of vascular endothelial growth factor receptor 2. J Pathol 2016; 240:461-471. [PMID: 27577973 DOI: 10.1002/path.4805] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 08/22/2016] [Accepted: 08/26/2016] [Indexed: 01/13/2023]
Abstract
Dynamic control of endothelial cell junctions is essential for vascular homeostasis and angiogenesis. We recently provided genetic evidence that ANGPTL4 is a key regulator of vascular integrity both during developmental and in hypoxia-induced pathological conditions. The purpose of the present study was to decipher the molecular mechanisms through which ANGPTL4 regulates vascular integrity. Using surface plasmon resonance and proximity ligation assays, we show that ANGPTL4 binds integrin αvβ3. In vitro and in vivo functional assays with Angptl4-deficient mice demonstrate that ANGPTL4-αvβ3 interaction is necessary to mediate ANGPTL4 vasoprotective effects. Mechanistically, ANGPTL4-αvβ3 interaction enhances Src recruitment to integrin αvβ3 and inhibits Src signalling downstream of vascular endothelial growth factor receptor 2 (VEFGR2), thereby repressing hypoxia-induced breakdown of VEGFR2-VE-cadherin and VEGFR2-αvβ3 complexes. We further demonstrate that intravitreal injection of recombinant human ANGPTL4 limits vascular permeability and leads to increased adherens junction and tight junction integrity. These findings identify a novel mechanism by which ANGPTL4 counteracts hypoxia-driven vascular permeability through integrin αvβ3 binding, modulation of VEGFR2-Src kinase signalling, and endothelial junction stabilization. We further demonstrate that Angptl4-deficient mice show increased vascular leakage in vivo in a model of laser-induced choroidal neovascularization, indicating that this newly identified ANGPTL4-αvβ3 axis might be a target for pharmaceutical intervention in pathological conditions. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Elisa Gomez Perdiguero
- Centre for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France.,Ecole Doctorale 394: Physiologie, Physiopathologie et Thérapeutique, Université Pierre et Marie Curie, Paris, France
| | - Athanasia Liabotis-Fontugne
- Centre for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France.,Ecole Doctorale 394: Physiologie, Physiopathologie et Thérapeutique, Université Pierre et Marie Curie, Paris, France
| | - Mélanie Durand
- Centre for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Clément Faye
- UMR 5086 CNRS Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Sylvie Ricard-Blum
- UMR 5086 CNRS Université Lyon 1, Institut de Biologie et Chimie des Protéines, Lyon, France
| | - Manuel Simonutti
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Sébastien Augustin
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Bryan M Robb
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, CNRS, Institut de la Vision, Paris, France
| | - Michel Paques
- Sorbonne Universités, UPMC Univ. Paris 06, INSERM, CNRS, Institut de la Vision, Paris, France.,Centre Hospitalier National d'Ophtalmologie des Quinze-Vingts, INSERM-DHOS CIC 503, Paris, France
| | | | | | | | | | - Ariane Galaup
- Centre for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Catherine Monnot
- Centre for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| | - Stéphane Germain
- Centre for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, PSL Research University, Paris, France
| |
Collapse
|
69
|
Dalton AC, Shlamkovitch T, Papo N, Barton WA. Constitutive Association of Tie1 and Tie2 with Endothelial Integrins is Functionally Modulated by Angiopoietin-1 and Fibronectin. PLoS One 2016; 11:e0163732. [PMID: 27695111 PMCID: PMC5047623 DOI: 10.1371/journal.pone.0163732] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/13/2016] [Indexed: 12/29/2022] Open
Abstract
Functional cross-talk between Tie2 and Integrin signaling pathways is essential to coordinate endothelial cell adhesion and migration in response to the extracellular matrix, yet the mechanisms behind this phenomenon are unclear. Here, we examine the possibility that receptor cross-talk is driven through uncharacterized Tie-integrin interactions on the endothelial surface. Using a live cell FRET-based proximity assay, we monitor Tie-integrin receptor recognition and demonstrate that both Tie1 and Tie2 readily associate with integrins α5ß1 and αVß3 through their respective ectodomains. Although not required, Tie2-integrin association is significantly enhanced in the presence of the extracellular component and integrin ligand fibronectin. In vitro binding assays with purified components reveal that Tie-integrin recognition is direct, and further demonstrate that the receptor binding domain of the Tie2 ligand Ang-1, but not the receptor binding domain of Ang-2, can independently associate with α5ß1 or αVß3. Finally, we reveal that cooperative Tie/integrin interactions selectively stimulate ERK/MAPK signaling in the presence of both Ang-1 and fibronectin, suggesting a molecular mechanism to sensitize Tie2 to extracellular matrix. We provide a mechanistic model highlighting the role of receptor localization and association in regulating distinct signaling cascades and in turn, the angiogenic switch.
Collapse
Affiliation(s)
- Annamarie C. Dalton
- Virginia Commonwealth University, Department of Biochemistry and Molecular Biology, Richmond, Virginia, 23298, United States of America
| | - Tomer Shlamkovitch
- Ben-Gurion University of the Negev, Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Beer-Sheva, 8410501, Israel
| | - Niv Papo
- Ben-Gurion University of the Negev, Department of Biotechnology Engineering and the National Institute of Biotechnology in the Negev, Beer-Sheva, 8410501, Israel
| | - William A. Barton
- Virginia Commonwealth University, Department of Biochemistry and Molecular Biology, Richmond, Virginia, 23298, United States of America
- * E-mail:
| |
Collapse
|
70
|
Candela ME, Wang C, Gunawardena AT, Zhang K, Cantley L, Yasuhara R, Usami Y, Francois N, Iwamoto M, van der Flier A, Zhang Y, Qin L, Han L, Enomoto-Iwamoto M. Alpha 5 Integrin Mediates Osteoarthritic Changes in Mouse Knee Joints. PLoS One 2016; 11:e0156783. [PMID: 27280771 PMCID: PMC4900574 DOI: 10.1371/journal.pone.0156783] [Citation(s) in RCA: 16] [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: 02/14/2016] [Accepted: 05/19/2016] [Indexed: 01/26/2023] Open
Abstract
Osteoarthritis (OA) is one of most common skeletal disorders and can affect synovial joints such as knee and ankle joints. α5 integrin, a major fibronectin receptor, is expressed in articular cartilage and has been demonstrated to play roles in synovial joint development and in the regulation of chondrocyte survival and matrix degradation in articular cartilage. We hypothesized that α5 integrin signaling is involved in pathogenesis of OA. To test this, we generated compound mice that conditionally ablate α5 integrin in the synovial joints using the Gdf5Cre system. The compound mice were born normally and had an overall appearance similar to the control mice. However, when the mutant mice received the OA surgery, they showed stronger resistance to osteoarthritic changes than the control. Specifically the mutant knee joints presented lower levels of cartilage matrix and structure loss and synovial changes and showed stronger biomechanical properties than the control knee joints. These findings indicate that α5 integrin may not be essential for synovial joint development but play a causative role in induction of osteoarthritic changes.
Collapse
Affiliation(s)
- Maria Elena Candela
- Department of Surgery, Division of Orthopaedic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Chao Wang
- School of Biomedical Engineering Science, and Health Systems, Drexel University, Philadelphia, PA, United States of America
| | - Aruni T. Gunawardena
- Department of Surgery, Division of Orthopaedic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Kairui Zhang
- Department of Surgery, Division of Orthopaedic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- Department of Orthopaedics and Traumatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Leslie Cantley
- Department of Surgery, Division of Orthopaedic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Rika Yasuhara
- Division of Pathology, Department of Oral Diagnosis Science, School of Dentistry, Showa University, Tokyo, Japan
| | - Yu Usami
- Department of Surgery, Division of Orthopaedic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Noelle Francois
- Department of Surgery, Division of Orthopaedic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Masahiro Iwamoto
- Department of Surgery, Division of Orthopaedic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Arjan van der Flier
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Yejia Zhang
- Department of Physical Medicine and Rehabilitation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Ling Qin
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Lin Han
- School of Biomedical Engineering Science, and Health Systems, Drexel University, Philadelphia, PA, United States of America
| | - Motomi Enomoto-Iwamoto
- Department of Surgery, Division of Orthopaedic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|
71
|
Mas-Moruno C, Fraioli R, Rechenmacher F, Neubauer S, Kapp TG, Kessler H. αvβ3- or α5β1-Integrin-Selective Peptidomimetics for Surface Coating. Angew Chem Int Ed Engl 2016; 55:7048-67. [PMID: 27258759 DOI: 10.1002/anie.201509782] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Indexed: 12/21/2022]
Abstract
Engineering biomaterials with integrin-binding activity is a very powerful approach to promote cell adhesion, modulate cell behavior, and induce specific biological responses at the surface level. The aim of this Review is to illustrate the evolution of surface-coating molecules in this field: from peptides and proteins with relatively low integrin-binding activity and receptor selectivity to highly active and selective peptidomimetic ligands. In particular, we will bring into focus the difficult challenge of achieving selectivity between the two closely related integrin subtypes αvβ3 and α5β1. The functionalization of surfaces with such peptidomimetics opens the way for a new generation of highly specific cell-instructive surfaces to dissect the biological role of integrin subtypes and for application in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering and Centre for Research in NanoEngineering, Universitat Politècnica de Catalunya (UPC), Diagonal 647, 08028, Barcelona, Spain.
| | - Roberta Fraioli
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering and Centre for Research in NanoEngineering, Universitat Politècnica de Catalunya (UPC), Diagonal 647, 08028, Barcelona, Spain
| | - Florian Rechenmacher
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Stefanie Neubauer
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Tobias G Kapp
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany
| | - Horst Kessler
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM), Technische Universität München, Lichtenbergstrasse 4, 85748, Garching, Germany.
| |
Collapse
|
72
|
Mas-Moruno C, Fraioli R, Rechenmacher F, Neubauer S, Kapp TG, Kessler H. αvβ3- oder α5β1-Integrin-selektive Peptidmimetika für die Oberflächenbeschichtung. Angew Chem Int Ed Engl 2016. [DOI: 10.1002/ange.201509782] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Carlos Mas-Moruno
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering and Centre for Research in NanoEngineering; Universitat Politècnica de Catalunya (UPC); Diagonal 647 08028 Barcelona Spanien
| | - Roberta Fraioli
- Biomaterials, Biomechanics and Tissue Engineering Group, Department of Materials Science and Metallurgical Engineering and Centre for Research in NanoEngineering; Universitat Politècnica de Catalunya (UPC); Diagonal 647 08028 Barcelona Spanien
| | - Florian Rechenmacher
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM); Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| | - Stefanie Neubauer
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM); Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| | - Tobias G. Kapp
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM); Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| | - Horst Kessler
- Institute for Advanced Study at the Department Chemie und Center of Integrated Protein Science München (CIPSM); Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| |
Collapse
|
73
|
Wang WQ, Wang FH, Qin WX, Liu HY, Lu B, Chung C, Zhu J, Gu Q, Shi W, Wen C, Wu F, Zhang K, Sun XD. Joint Antiangiogenic Effect of ATN-161 and Anti-VEGF Antibody in a Rat Model of Early Wet Age-Related Macular Degeneration. Mol Pharm 2016; 13:2881-90. [PMID: 27089240 DOI: 10.1021/acs.molpharmaceut.6b00056] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The wet form of age-related macular degeneration (AMD) is a leading cause of blindness among elderly Americans and is characterized by abnormal vessel growth, termed choroidal neovascularization (CNV). Integrin α5β1 is a transmembrane receptor that binds matrix macromolecules and proteinases to stimulate angiogenesis. We recently demonstrated that integrin α5β1 plays a critical role in the development of choroidal neovascularization. In this study, we determined the role and underlying mechanisms of integrin α5β1 in angiogenesis in human choroidal endothelial cells and evaluated the antiangiogenic effects of delivering a combination therapy of ATN-161, an integrin α5β1 inhibitor, and an anti-VEGF monoclonal antibody to rats with laser-induced CNV. Vascular endothelial growth factor (VEGF) is a signaling protein that stimulates vasculogenesis and angiogenesis through a pathway that is distinct from the integrin α5β1 signaling pathway. Our results indicate that fibronectin binds to integrin α5β1 and synergizes VEGF-induced angiogenesis via two independent signaling pathways, FN/integrin α5β1/FAK/ERK1/2 and FN/integrin α5β1/FAK/AKT. Integrin α5 knockdown by shRNA inhibits endothelial cell migration, tube formation, and proliferation, while ATN-161 only partially decreases integrin α5 function. Treatment with ATN-161 combined with anti-VEGF antibody showed joint effects in attenuating angiogenesis. In summary, our results provide the first evidence for the mechanisms by which integrin α5β1 is involved in ocular pathological neovascularization in vivo, suggesting that dual inhibition of integrin α5β1 and VEGF may be a promising novel therapeutic strategy for CNV in wet AMD.
Collapse
Affiliation(s)
- Wen-Qiu Wang
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China.,Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Feng-Hua Wang
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China
| | - Wen-Xin Qin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai, 200032, China
| | - Hai-Yun Liu
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China
| | - Bing Lu
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China
| | - Christopher Chung
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Jie Zhu
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Qing Gu
- Shanghai Key Laboratory of Fundus Disease and Eye Research Institute, Shanghai JiaoTong University , Shanghai 200080, China
| | - William Shi
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States.,Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Sichuan 610041, China
| | - Cindy Wen
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States.,Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Sichuan 610041, China
| | - Frances Wu
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States
| | - Kang Zhang
- Department of Ophthalmology, Biomaterial and Tissue Engineering Center, Institute of Engineering in Medicine and Institute for Genomic Medicine, University of California, San Diego , La Jolla, California 92093, United States.,Molecular Medicine Research Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University , Sichuan 610041, China.,Veterans Administration Healthcare System , San Diego, California 92161, United States
| | - Xiao-Dong Sun
- Department of Ophthalmology, Shanghai First People's Hospital, School of Medicine, Shanghai JiaoTong University , Shanghai, 20080, China.,Shanghai Key Laboratory of Fundus Disease and Eye Research Institute, Shanghai JiaoTong University , Shanghai 200080, China
| |
Collapse
|
74
|
Rossi E, Smadja DM, Boscolo E, Langa C, Arevalo MA, Pericacho M, Gamella-Pozuelo L, Kauskot A, Botella LM, Gaussem P, Bischoff J, Lopez-Novoa JM, Bernabeu C. Endoglin regulates mural cell adhesion in the circulatory system. Cell Mol Life Sci 2016; 73:1715-39. [PMID: 26646071 PMCID: PMC4805714 DOI: 10.1007/s00018-015-2099-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/19/2015] [Accepted: 11/23/2015] [Indexed: 02/06/2023]
Abstract
The circulatory system is walled off by different cell types, including vascular mural cells and podocytes. The interaction and interplay between endothelial cells (ECs) and mural cells, such as vascular smooth muscle cells or pericytes, play a pivotal role in vascular biology. Endoglin is an RGD-containing counter-receptor for β1 integrins and is highly expressed by ECs during angiogenesis. We find that the adhesion between vascular ECs and mural cells is enhanced by integrin activators and inhibited upon suppression of membrane endoglin or β1-integrin, as well as by addition of soluble endoglin (SolEng), anti-integrin α5β1 antibody or an RGD peptide. Analysis of different endoglin mutants, allowed the mapping of the endoglin RGD motif as involved in the adhesion process. In Eng (+/-) mice, a model for hereditary hemorrhagic telangectasia type 1, endoglin haploinsufficiency induces a pericyte-dependent increase in vascular permeability. Also, transgenic mice overexpressing SolEng, an animal model for preeclampsia, show podocyturia, suggesting that SolEng is responsible for podocytes detachment from glomerular capillaries. These results suggest a critical role for endoglin in integrin-mediated adhesion of mural cells and provide a better understanding on the mechanisms of vessel maturation in normal physiology as well as in pathologies such as preeclampsia or hereditary hemorrhagic telangiectasia.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cell Adhesion/physiology
- Cell Line, Tumor
- Disease Models, Animal
- Endoglin
- Endothelium, Vascular/metabolism
- Female
- Human Umbilical Vein Endothelial Cells/metabolism
- Humans
- Integrin beta1/genetics
- Jurkat Cells
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Nude
- Mice, Transgenic
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Neovascularization, Pathologic/metabolism
- Pericytes/metabolism
- Podocytes/metabolism
- Pre-Eclampsia/genetics
- Pre-Eclampsia/pathology
- Pregnancy
- Protein Binding
- RNA Interference
- RNA, Small Interfering
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Retina/metabolism
- Telangiectasia, Hereditary Hemorrhagic/genetics
- Telangiectasia, Hereditary Hemorrhagic/pathology
Collapse
Affiliation(s)
- Elisa Rossi
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
- Paris Descartes University, Sorbonne Paris Cite, Paris, France
- Hematology Department, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - David M Smadja
- Hematology Department, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
- Faculté de Pharmacie, Inserm UMR-S1140, Paris, France
| | - Elisa Boscolo
- Department of Surgery, Harvard Medical School, Children's Hospital, Boston, MA, 02115, USA
| | - Carmen Langa
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
| | - Miguel A Arevalo
- Departamento de Anatomía e Histología Humanas, Facultad de Medicina, Universidad de Salamanca, 37007, Salamanca, Spain
- Instituto de Investigaciones Biomédicas de Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Miguel Pericacho
- Instituto de Investigaciones Biomédicas de Salamanca (IBSAL), 37007, Salamanca, Spain
- Departamento de Fisiología y Farmacología, Unidad de Fisiopatología Renal y Cardiovascular, Universidad de Salamanca, 37007, Salamanca, Spain
| | - Luis Gamella-Pozuelo
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
- Departamento de Fisiología y Farmacología, Unidad de Fisiopatología Renal y Cardiovascular, Universidad de Salamanca, 37007, Salamanca, Spain
| | - Alexandre Kauskot
- Inserm UMR-S1176, Le Kremlin Bicêtre, Paris, France
- Université Paris Sud, Le Kremlin Bicêtre, Paris, France
| | - Luisa M Botella
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain
| | - Pascale Gaussem
- Hematology Department, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
- Faculté de Pharmacie, Inserm UMR-S1140, Paris, France
| | - Joyce Bischoff
- Department of Surgery, Harvard Medical School, Children's Hospital, Boston, MA, 02115, USA
| | - José M Lopez-Novoa
- Instituto de Investigaciones Biomédicas de Salamanca (IBSAL), 37007, Salamanca, Spain
- Departamento de Fisiología y Farmacología, Unidad de Fisiopatología Renal y Cardiovascular, Universidad de Salamanca, 37007, Salamanca, Spain
| | - Carmelo Bernabeu
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), c/Ramiro de Maeztu 9, 28040, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 28040, Madrid, Spain.
| |
Collapse
|
75
|
Liang Q, Ju Y, Chen Y, Wang W, Li J, Zhang L, Xu H, Wood RW, Schwarz EM, Boyce BF, Wang Y, Xing L. Lymphatic endothelial cells efferent to inflamed joints produce iNOS and inhibit lymphatic vessel contraction and drainage in TNF-induced arthritis in mice. Arthritis Res Ther 2016; 18:62. [PMID: 26970913 PMCID: PMC4789262 DOI: 10.1186/s13075-016-0963-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 02/26/2016] [Indexed: 12/11/2022] Open
Abstract
Background In this study, we sought to determine the cellular source of inducible nitric oxide synthase (iNOS) induced in lymphatic endothelial cells (LECs) in response to tumor necrosis factor (TNF), the effects of iNOS on lymphatic smooth muscle cell (LSMC) function and on the development of arthritis in TNF-transgenic (TNF-Tg) mice, and whether iNOS inhibitors improve lymphatic function and reduce joint destruction in inflammatory erosive arthritis. Methods We used quantitative polymerase chain reactions, immunohistochemistry, histology, and near-infrared imaging to examine (1) iNOS expression in podoplanin + LECs and lymphatic vessels from wild-type (WT) and TNF-Tg mice, (2) iNOS induction by TNF in WT LECs, (3) the effects of iNOS inhibitors on expression of functional muscle genes in LSMCs, and (4) the effects of iNOS inhibitors on lymphatic vessel contraction and drainage, as well as the severity of arthritis, in TNF-Tg mice. Results LECs from TNF-Tg mice had eight fold higher iNOS messenger RNA levels than WT cells, and iNOS expression was confirmed immunohistochemically in podoplanin + LECs in lymphatic vessels from inflamed joints. TNF (0.1 ng/ml) increased iNOS levels 40-fold in LECs. LSMCs cocultured with LECs pretreated with TNF had reduced expression of functional muscle genes. This reduction was prevented by ferulic acid, which blocked nitric oxide production. Local injection of L-N6-(1-iminoethyl)lysine 5-tetrazole-amide into inflamed paws of TNF-Tg mice resulted in recovery of lymphatic vessel contractions and drainage. Treatment of TNF-Tg mice with ferulic acid reduced synovial inflammation as well as cartilage and bone erosion, and it also restored lymphatic contraction and drainage. Conclusions iNOS is produced primarily by LECs in lymphatic vessel efferent from inflamed joints of TNF-Tg mice in response to TNF and inhibits LSMC contraction and lymph drainage. Ferulic acid represents a potential new therapy to restore lymphatic function and thus improve inflammatory arthritis by inhibiting local production of nitric oxide by LSMCs. Electronic supplementary material The online version of this article (doi:10.1186/s13075-016-0963-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qianqian Liang
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Yawen Ju
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Yan Chen
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Wensheng Wang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Jinlong Li
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Li Zhang
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China
| | - Hao Xu
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Ronald W Wood
- Departments of Obstetrics and Gynecology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Edward M Schwarz
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.,Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA
| | - Yongjun Wang
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai, 200032, China. .,Institute of Spine, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai, 200032, China.
| | - Lianping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA. .,Center for Musculoskeletal Research, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA. .,Departments of Obstetrics and Gynecology, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, 14642, USA.
| |
Collapse
|
76
|
Liang QQ, Shi Q, Wood RW, Xing LP, Wang YJ. Peri-articular lymphatic system and "Bi" theory of Chinese medicine in the pathogenesis and treatment of arthritis. Chin J Integr Med 2015; 21:648-55. [PMID: 26432788 DOI: 10.1007/s11655-015-2305-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Indexed: 12/11/2022]
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) are the two most common joint diseases, and they have characterization of synovial inflammation and cartilage destruction, associated with the accumulation of numerous catabolic mediators and inflammatory cells in the synovial space and surrounding soft tissues. How these factors are cleared and if the "clearance" process contributes to pathogenesis of arthritis are not known. Recently, we found the existence of the peri-articular lymphatic system in mouse joints. The blockade of lymphangiogenesis and lymphatic draining function accelerates while stimulation of lymphatic function attenuates the severity of joint tissue lesions in mouse models of RA and OA. More importantly, we noticed the similarity between the dysfunction of lymphatic drainage in arthritic joints and "Bi" theory of Chinese medicine (CM), and demonstrated that several Bi disease-treated herbal drugs directly affect the function of lymphatic endothelial cells. Here we review the advances about the interactions between joint inflammation and changes in the peri-articular lymphatic system and discuss our view of linking "Bi" theory of CM to lymphatic dysfunction in arthritis.
Collapse
Affiliation(s)
- Qian-Qian Liang
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Qi Shi
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China
| | - Ronald W Wood
- Department of Obstetrics and Gynecology, Urology, and Neurobiology and Anatomy, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Lian-Ping Xing
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, 14642, USA. .,Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| | - Yong-Jun Wang
- Department of Orthopaedics, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, China.
| |
Collapse
|
77
|
Chen D, Wang X, Liang D, Gordon J, Mittal A, Manley N, Degenhardt K, Astrof S. Fibronectin signals through integrin α5β1 to regulate cardiovascular development in a cell type-specific manner. Dev Biol 2015; 407:195-210. [PMID: 26434918 DOI: 10.1016/j.ydbio.2015.09.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 09/09/2015] [Accepted: 09/12/2015] [Indexed: 01/23/2023]
Abstract
Fibronectin (Fn1) is an evolutionarily conserved extracellular matrix glycoprotein essential for embryonic development. Global deletion of Fn1 leads to mid-gestation lethality from cardiovascular defects. However, severe morphogenetic defects that occur early in embryogenesis in these embryos precluded assigning a direct role for Fn1 in cardiovascular development. We noticed that Fn1 is expressed in strikingly non-uniform patterns during mouse embryogenesis, and that its expression is particularly enriched in the pharyngeal region corresponding with the pharyngeal arches 3, 4, and 6. This region bears a special importance for the developing cardiovascular system, and we hypothesized that the localized enrichment of Fn1 in the pharyngeal region may be essential for cardiovascular morphogenesis. To test this hypothesis, we ablated Fn1 using the Isl1(Cre) knock-in strain of mice. Deletion of Fn1 using the Isl1(Cre) strain resulted in defective formation of the 4th pharyngeal arch arteries (PAAs), aberrant development of the cardiac outflow tract (OFT), and ventricular septum defects. To determine the cell types responding to Fn1 signaling during cardiovascular development, we deleted a major Fn1 receptor, integrin α5 using the Isl1(Cre) strain, and observed the same spectrum of abnormalities seen in the Fn1 conditional mutants. Additional conditional mutagenesis studies designed to ablate integrin α5 in distinct cell types within the Isl1(+) tissues and their derivatives, suggested that the expression of integrin α5 in the pharyngeal arch mesoderm, endothelium, surface ectoderm and the neural crest were not required for PAA formation. Our studies suggest that an (as yet unknown) integrin α5-dependent signal extrinsic to the pharyngeal endothelium mediates the formation of the 4th PAAs.
Collapse
Affiliation(s)
- Dongying Chen
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA; Cell and Developmental Biology graduate program, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Xia Wang
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Dong Liang
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Julie Gordon
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Ashok Mittal
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Nancy Manley
- Department of Genetics, University of Georgia, Athens, GA 30602, USA
| | - Karl Degenhardt
- Children's Hospital of Pennsylvania, University of Pennsylvania, Philadelphia, PA 19107, USA
| | - Sophie Astrof
- Sidney Kimmel Medical College of Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA; Cell and Developmental Biology graduate program, Sidney Kimmel Medical College of Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
78
|
Guasch J, Conings B, Neubauer S, Rechenmacher F, Ende K, Rolli CG, Kappel C, Schaufler V, Micoulet A, Kessler H, Boyen HG, Cavalcanti-Adam EA, Spatz JP. Segregation versus colocalization: orthogonally functionalized binary micropatterned substrates regulate the molecular distribution in focal adhesions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2015; 27:3737-3747. [PMID: 25981929 DOI: 10.1002/adma.201500900] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 04/05/2015] [Indexed: 06/04/2023]
Abstract
Orthogonally functionalized binary micropatterned substrates are produced using a novel protocol. The use of adequate peptido-mimetics enables an unprecedented segregation of purified αvβ3 and α5β1 integrins in adjacent microislands and evidences the preference of U2OS cells to colocalize such receptors. Moreover, this tendency can be altered by varying the geometry and composition of the micropatterns.
Collapse
Affiliation(s)
- Judith Guasch
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Department of Biophysical Chemistry, University of Heidelberg INF 253, Heidelberg, D-69120, Germany
| | - Bert Conings
- Institute for Materials Research, Wetenschapspark 1, Diepenbeek, B-3590, Belgium
| | - Stefanie Neubauer
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Institute for Advanced Study (IAS) and Center of Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München, Lichtenbergstr. 4, Garching, D-85747, Germany
| | - Florian Rechenmacher
- Institute for Advanced Study (IAS) and Center of Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München, Lichtenbergstr. 4, Garching, D-85747, Germany
| | - Karen Ende
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
| | - Claudio G Rolli
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Department of Biophysical Chemistry, University of Heidelberg INF 253, Heidelberg, D-69120, Germany
| | - Christian Kappel
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
| | - Viktoria Schaufler
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Department of Biophysical Chemistry, University of Heidelberg INF 253, Heidelberg, D-69120, Germany
| | - Alexandre Micoulet
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Department of Biophysical Chemistry, University of Heidelberg INF 253, Heidelberg, D-69120, Germany
| | - Horst Kessler
- Institute for Advanced Study (IAS) and Center of Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München, Lichtenbergstr. 4, Garching, D-85747, Germany
| | - Hans-Gerd Boyen
- Institute for Materials Research, Wetenschapspark 1, Diepenbeek, B-3590, Belgium
| | - Elisabetta Ada Cavalcanti-Adam
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Department of Biophysical Chemistry, University of Heidelberg INF 253, Heidelberg, D-69120, Germany
| | - Joachim P Spatz
- Department of New Materials and Biosystems, Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, D-70569, Germany
- Department of Biophysical Chemistry, University of Heidelberg INF 253, Heidelberg, D-69120, Germany
| |
Collapse
|
79
|
Wang Z, Jinnin M, Kobayashi Y, Kudo H, Inoue K, Nakayama W, Honda N, Makino K, Kajihara I, Makino T, Fukushima S, Inagaki Y, Ihn H. Mice overexpressing integrin αv in fibroblasts exhibit dermal thinning of the skin. J Dermatol Sci 2015; 79:268-78. [PMID: 26117269 DOI: 10.1016/j.jdermsci.2015.06.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 04/26/2015] [Accepted: 06/17/2015] [Indexed: 11/19/2022]
Abstract
BACKGROUND Integrins, especially αv integrin (ITGAV), are thought to play central roles in tissue fibrosis and the pathogenesis of scleroderma. So far, skin phenotype of tissue-specific transgenic mice of ITGAV have not been investigated. OBJECTIVE To investigate the role of ITGAV in the skin fibrosis, we engineered transgenic mice that overexpress ITGAV in the fibroblasts under the control of the COL1A2 enhancer promoter. METHODS Protein or RNA expression was evaluated by real-time PCR, immunohistochemistry, immunoblotting and immunoprecipitation. RESULTS Dermal thickness and Masson's trichrome staining were decreased in ITGAV transgenic (Tg) mice compared with wild-type (WT) mice. Protein and mRNA levels of COL1A2, COL3A1, CTGF and integrin β3 were down-regulated in the skin of Tg mice. In addition, the cell proliferation of cultured dermal fibroblasts obtained from Tg mice skin was decreased compared to those of WT mice. FAK phosphorylation was reduced in fibroblasts cultured from Tg mice skin in comparison to WT mice fibroblasts. Integrin β3 siRNA inhibited FAK phosphorylation levels, while FAK inhibitor reduced the expression of collagens and CTGF in mice dermal fibroblasts. CONCLUSIONS The down-regulation of collagen or CTGF by decreased integrin β3 and FAK phosphorylation may cause the dermal thinning in Tg mice. Lower CTGF may also result in reduced growth of Tg mice fibroblasts. Our hypothesis is that the balance between α and β chain of integrins positively or negatively control collagen expression and dermal thickness. This study gave a new insight in the treatment of tissue fibrosis and scleroderma by balancing integrin expression.
Collapse
Affiliation(s)
- Zhongzhi Wang
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556, Japan
| | - Masatoshi Jinnin
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556, Japan.
| | - Yuki Kobayashi
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556, Japan
| | - Hideo Kudo
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556, Japan
| | - Kuniko Inoue
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556, Japan
| | - Wakana Nakayama
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556, Japan
| | - Noritoshi Honda
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556, Japan
| | - Katsunari Makino
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556, Japan
| | - Ikko Kajihara
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556, Japan
| | - Takamitsu Makino
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556, Japan
| | - Satoshi Fukushima
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556, Japan
| | - Yutaka Inagaki
- Center for Matrix Biology and Medicine, Graduate School of Medicine, Tokai University, 143 Shimo-kasuya, Isehara, Kanagawa 259-1193, Japan
| | - Hironobu Ihn
- Department of Dermatology and Plastic Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Kumamoto 860-8556, Japan
| |
Collapse
|
80
|
Turner CJ, Badu-Nkansah K, Crowley D, van der Flier A, Hynes RO. α5 and αv integrins cooperate to regulate vascular smooth muscle and neural crest functions in vivo. Development 2015; 142:797-808. [PMID: 25670798 DOI: 10.1242/dev.117572] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The RGD-binding α5 and αv integrins have been shown to be key regulators of vascular smooth muscle cell (vSMC) function in vitro. However, their role on vSMCs during vascular development in vivo remains unclear. To address this issue, we have generated mice that lack α5, αv or both α5 and αv integrins on their vSMCs, using the SM22α-Cre transgenic mouse line. To our surprise, neither α5 nor αv mutants displayed any obvious vascular defects during embryonic development. By contrast, mice lacking both α5 and αv integrins developed interrupted aortic arches, large brachiocephalic/carotid artery aneurysms and cardiac septation defects, but developed extensive and apparently normal vasculature in the skin. Cardiovascular defects were also found, along with cleft palates and ectopically located thymi, in Wnt1-Cre α5/αv mutants, suggesting that α5 and αv cooperate on neural crest-derived cells to control the remodelling of the pharyngeal arches and the septation of the heart and outflow tract. Analysis of cultured α5/αv-deficient vSMCs suggests that this is achieved, at least in part, through proper assembly of RGD-containing extracellular matrix proteins and the correct incorporation and activation of latent TGF-β.
Collapse
Affiliation(s)
- Christopher J Turner
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kwabena Badu-Nkansah
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Denise Crowley
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arjan van der Flier
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Richard O Hynes
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
81
|
Actin-tethered junctional complexes in angiogenesis and lymphangiogenesis in association with vascular endothelial growth factor. BIOMED RESEARCH INTERNATIONAL 2015; 2015:314178. [PMID: 25883953 PMCID: PMC4389985 DOI: 10.1155/2015/314178] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/23/2014] [Accepted: 10/31/2014] [Indexed: 12/21/2022]
Abstract
Vasculature is present in all tissues and therefore is indispensable for development, biology, and pathology of multicellular organisms. Endothelial cells guarantee proper function of the vessels and are the original component in angiogenesis. Morphogenesis of the vascular system utilizes processes like cell adhesion, motility, proliferation, and survival that are closely related to the dynamics of actin filaments and actin-tethered adhesion complexes. Here we review involvement of actin cytoskeleton-associated junctional molecules of endothelial cells in angiogenesis and lymphangiogenesis. Particularly, we focus on F-actin binding protein afadin, an adaptor protein involved in broad range of signaling mechanisms. Afadin mediates the pathways of vascular endothelial growth factor- (VEGF-) and sphingosine 1-phosphate-triggered angiogenesis and is essential for embryonic development of lymph vessels in mice. We propose that targeting actin-tethered junctional molecules, including afadin, may present a new approach to angiogenic therapy that in combination with today used medications like VEGF inhibitors will benefit against development of pathological angiogenesis.
Collapse
|
82
|
Murphy PA, Begum S, Hynes RO. Tumor angiogenesis in the absence of fibronectin or its cognate integrin receptors. PLoS One 2015; 10:e0120872. [PMID: 25807551 PMCID: PMC4373772 DOI: 10.1371/journal.pone.0120872] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 02/10/2015] [Indexed: 11/18/2022] Open
Abstract
Binding of α5β1 and αvβ3/β5 integrin receptors on the endothelium to their fibronectin substrate in the extracellular matrix has been targeted as a possible means of blocking tumor angiogenesis and tumor growth. However, clinical trials of blocking antibodies and peptides have been disappointing despite promising preclinical results, leading to questions about the mechanism of the inhibitors and the reasons for their failure. Here, using tissue-specific and inducible genetics to delete the α5 and αv receptors in the endothelium or their fibronectin substrate, either in the endothelium or globally, we show that both are dispensable for tumor growth, in transplanted tumors as well as spontaneous and angiogenesis-dependent RIP-Tag-driven pancreatic adenocarcinomas. In the nearly complete absence of fibronectin, no differences in vascular density or the deposition of basement membrane laminins, ColIV, Nid1, Nid2, or the TGFβ binding matrix proteins, fibrillin-1 and -2, could be observed. Our results reveal that fibronectin and the endothelial fibronectin receptor subunits, α5 and αv, are dispensable for tumor angiogenesis, suggesting that the inhibition of angiogenesis induced by antibodies or small molecules may occur through a dominant negative effect, rather than a simple functional block.
Collapse
Affiliation(s)
- Patrick A. Murphy
- Howard Hughes Medical Institute, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Shahinoor Begum
- Howard Hughes Medical Institute, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States of America
| | - Richard O. Hynes
- Howard Hughes Medical Institute, David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States of America
- * E-mail:
| |
Collapse
|
83
|
Integrin β1 controls VE-cadherin localization and blood vessel stability. Nat Commun 2015; 6:6429. [PMID: 25752958 DOI: 10.1038/ncomms7429] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 01/28/2015] [Indexed: 01/19/2023] Open
Abstract
Angiogenic blood vessel growth requires several distinct but integrated cellular activities. Endothelial cell sprouting and proliferation lead to the expansion of the vasculature and give rise to a highly branched, immature plexus, which is subsequently reorganized into a mature and stable network. Although it is known that integrin-mediated cell-matrix interactions are indispensable for embryonic angiogenesis, little is known about the function of integrins in different steps of vascular morphogenesis. Here, by investigating the integrin β1-subunit with inducible and endothelial-specific gene targeting in the postnatal mouse retina, we show that β1 integrin promotes endothelial sprouting but is a negative regulator of proliferation. In maturing vessels, integrin β1 is indispensable for proper localization of VE-cadherin and thereby cell-cell junction integrity. The sum of our findings establishes that integrin β1 has critical functions in the growing and maturing vasculature, and is required for the formation of stable, non-leaky blood vessels.
Collapse
|
84
|
Zhu Y, Soderblom C, Trojanowsky M, Lee DH, Lee JK. Fibronectin Matrix Assembly after Spinal Cord Injury. J Neurotrauma 2015; 32:1158-67. [PMID: 25492623 DOI: 10.1089/neu.2014.3703] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
After spinal cord injury (SCI), a fibrotic scar forms at the injury site that is best characterized by the accumulation of perivascular fibroblasts and deposition of the extracellular matrix protein fibronectin. While fibronectin is a growth-permissive substrate for axons, the fibrotic scar is inhibitory to axon regeneration. The mechanism behind how fibronectin contributes to the inhibitory environment and how the fibronectin matrix is assembled in the fibrotic scar is unknown. By deleting fibronectin in myeloid cells, we demonstrate that fibroblasts are most likely the major source of fibronectin in the fibrotic scar. In addition, we demonstrate that fibronectin is initially present in a soluble form and is assembled into a matrix at 7 d post-SCI. Assembly of the fibronectin matrix may be mediated by the canonical fibronectin receptor, integrin α5β1, which is primarily expressed by activated macrophages/microglia in the fibrotic scar. Despite the pronounced cavitation after rat SCI, fibrotic scar also is observed in a rat SCI model, which is considered to be more similar to human pathology. Taken together, our study provides insight into the mechanism of fibrotic scar formation after spinal cord injury.
Collapse
Affiliation(s)
- Yunjiao Zhu
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami School of Medicine , Miami, Florida
| | - Cynthia Soderblom
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami School of Medicine , Miami, Florida
| | - Michelle Trojanowsky
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami School of Medicine , Miami, Florida
| | - Do-Hun Lee
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami School of Medicine , Miami, Florida
| | - Jae K Lee
- Department of Neurological Surgery, Miami Project to Cure Paralysis, University of Miami School of Medicine , Miami, Florida
| |
Collapse
|
85
|
Kim JE, Lee JM, Chung BG. Microwell arrays for uniform-sized embryoid body-mediated endothelial cell differentiation. Biomed Microdevices 2015; 16:559-66. [PMID: 24652615 DOI: 10.1007/s10544-014-9858-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Embryonic stem (ES) cell is of great interest cell source in regenerating tissue constructs. We hypothesized that the interaction of cell-extracellular matrices (ECMs) would enable the control of ES cell differentiation pathway. We fabricated the hydrogel microwell array system to regulate uniform-sized embryoid bodies (EBs) and replate into various ECM components (e.g., gelatin, collagen I, fibronectin, laminin, and Matrigel). We demonstrated that collagen I and laminin largely induced ES cell-derived endothelial cell differentiation compared to gelatin. We also characterized ECMs-dependent endothelial cell differentiation by evaluating the endothelial gene expression, showing that Flk1 endothelial gene was highly expressed on collagen I. We also demonstrated the effect of the integrin on uniform-sized EBs-derived endothelial cell differentiation, showing that integrin α1 was largely expressed on laminin. Therefore, the cell-ECM interaction could be potentially powerful for controlling the uniform-sized EBs-derived endothelial cell differentiation.
Collapse
Affiliation(s)
- Ji-eun Kim
- Department of Bionano Technology, Hanyang University, Ansan, Korea
| | | | | |
Collapse
|
86
|
Koivisto L, Heino J, Häkkinen L, Larjava H. Integrins in Wound Healing. Adv Wound Care (New Rochelle) 2014; 3:762-783. [PMID: 25493210 DOI: 10.1089/wound.2013.0436] [Citation(s) in RCA: 155] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Indexed: 01/06/2023] Open
Abstract
Significance: Regulation of cell adhesions during tissue repair is fundamentally important for cell migration, proliferation, and protein production. All cells interact with extracellular matrix proteins with cell surface integrin receptors that convey signals from the environment into the nucleus, regulating gene expression and cell behavior. Integrins also interact with a variety of other proteins, such as growth factors, their receptors, and proteolytic enzymes. Re-epithelialization and granulation tissue formation are crucially dependent on the temporospatial function of multiple integrins. This review explains how integrins function in wound repair. Recent Advances: Certain integrins can activate latent transforming growth factor beta-1 (TGF-β1) that modulates wound inflammation and granulation tissue formation. Dysregulation of TGF-β1 function is associated with scarring and fibrotic disorders. Therefore, these integrins represent targets for therapeutic intervention in fibrosis. Critical Issues: Integrins have multifaceted functions and extensive crosstalk with other cell surface receptors and molecules. Moreover, in aberrant healing, integrins may assume different functions, further increasing the complexity of their functionality. Discovering and understanding the role that integrins play in wound healing provides an opportunity to identify the mechanisms for medical conditions, such as excessive scarring, chronic wounds, and even cancer. Future Directions: Integrin functions in acute and chronic wounds should be further addressed in models better mimicking human wounds. Application of any products in acute or chronic wounds will potentially alter integrin functions that need to be carefully considered in the design.
Collapse
Affiliation(s)
- Leeni Koivisto
- Laboratory of Periodontal Biology, Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Jyrki Heino
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Lari Häkkinen
- Laboratory of Periodontal Biology, Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Hannu Larjava
- Laboratory of Periodontal Biology, Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| |
Collapse
|
87
|
Abstract
For nearly two decades now, the RGD (Arg-Gly-Asp)-binding αvβ3-integrin has been a focus of anti-angiogenic drug design. These inhibitors are well-tolerated, but have shown only limited success in patients. Over the years, studies in β3-integrin-knockout mice have shed some light on possible explanations for disappointing clinical outcomes. However, studying angiogenesis in β3-integrin-knockout mice is a blunt tool to investigate β3-integrin's role in pathological angiogenesis. Since establishing our laboratory at University of East Anglia (UEA), we have adopted more refined models of genetically manipulating the expression of the β3-integrin subunit. The present review will highlight some of our findings from these models and describe how data from them have forced us to rethink how targeting αvβ3-integrin expression affects tumour angiogenesis and cancer progression. Revisiting the fundamental biology behind how this integrin regulates tumour growth and angiogenesis, we believe, is the key not only to understanding how angiogenesis is normally co-ordinated, but also in success with drugs directed against it.
Collapse
|
88
|
Michaelis UR. Mechanisms of endothelial cell migration. Cell Mol Life Sci 2014; 71:4131-48. [PMID: 25038776 PMCID: PMC11113960 DOI: 10.1007/s00018-014-1678-0] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/23/2014] [Accepted: 07/07/2014] [Indexed: 01/13/2023]
Abstract
Cell migration plays a central role in a variety of physiological and pathological processes during our whole life. Cellular movement is a complex, tightly regulated multistep process. Although the principle mechanisms of migration follow a defined general motility cycle, the cell type and the context of moving influences the detailed mode of migration. Endothelial cells migrate during vasculogenesis and angiogenesis but also in a damaged vessel to restore vessel integrity. Depending on the situation they migrate individually, in chains or sheets and complex signaling, intercellular signals as well as environmental cues modulate the process. Here, the different modes of cell migration, the peculiarities of endothelial cell migration and specific guidance molecules controlling this process will be reviewed.
Collapse
Affiliation(s)
- U Ruth Michaelis
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität, Theodor-Stern-Kai 7, 60590, Frankfurt am Main, Germany,
| |
Collapse
|
89
|
Liang D, Wang X, Mittal A, Dhiman S, Hou SY, Degenhardt K, Astrof S. Mesodermal expression of integrin α5β1 regulates neural crest development and cardiovascular morphogenesis. Dev Biol 2014; 395:232-44. [PMID: 25242040 DOI: 10.1016/j.ydbio.2014.09.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/10/2014] [Accepted: 09/11/2014] [Indexed: 01/09/2023]
Abstract
Integrin α5-null embryos die in mid-gestation from severe defects in cardiovascular morphogenesis, which stem from defective development of the neural crest, heart and vasculature. To investigate the role of integrin α5β1 in cardiovascular development, we used the Mesp1(Cre) knock-in strain of mice to ablate integrin α5 in the anterior mesoderm, which gives rise to all of the cardiac and many of the vascular and muscle lineages in the anterior portion of the embryo. Surprisingly, we found that mutant embryos displayed numerous defects related to the abnormal development of the neural crest such as cleft palate, ventricular septal defect, abnormal development of hypoglossal nerves, and defective remodeling of the aortic arch arteries. We found that defects in arch artery remodeling stem from the role of mesodermal integrin α5β1 in neural crest proliferation and differentiation into vascular smooth muscle cells, while proliferation of pharyngeal mesoderm and differentiation of mesodermal derivatives into vascular smooth muscle cells was not defective. Taken together our studies demonstrate a requisite role for mesodermal integrin α5β1 in signaling between the mesoderm and the neural crest, thereby regulating neural crest-dependent morphogenesis of essential embryonic structures.
Collapse
Affiliation(s)
- Dong Liang
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Xia Wang
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Ashok Mittal
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Sonam Dhiman
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Shuan-Yu Hou
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA
| | - Karl Degenhardt
- Childrens Hospital of Pennsylvania, University of Pennsylvania, Philadelphia, PA 19107, USA
| | - Sophie Astrof
- Thomas Jefferson University, Department of Medicine, Center for Translational Medicine, 1020 Locust Street, Philadelphia, PA 19107, USA.
| |
Collapse
|
90
|
Pulina M, Liang D, Astrof S. Shape and position of the node and notochord along the bilateral plane of symmetry are regulated by cell-extracellular matrix interactions. Biol Open 2014; 3:583-90. [PMID: 24928429 PMCID: PMC4154294 DOI: 10.1242/bio.20148243] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The node and notochord (and their equivalents in other species) are essential signaling centers, positioned along the plane of bilateral symmetry in developing vertebrate embryos. However, genes and mechanisms regulating morphogenesis of these structures and their placement along the embryonic midline are not well understood. In this work, we provide the first evidence that the position of the node and the notochord along the bilateral plane of symmetry are under genetic control and are regulated by integrin α5β1 and fibronectin in mice. We found that the shape of the node is often inverted in integrin α5-null and fibronectin-null mutants, and that the positioning of node and the notochord is often skewed away from the perceived plane of embryonic bilateral of symmetry. Our studies also show that the shape and position of the notochord are dependent on the shape and embryonic placement of the node. Our studies suggest that fibronectin regulates the shape of the node by affecting apico-basal polarity of the nodal cells. Taken together, our data indicate that cell–extracellular matrix interactions mediated by integrin α5β1 and fibronectin regulate the geometry of the node as well as the placement of the node and notochord along the plane of bilateral symmetry in the mammalian embryo.
Collapse
Affiliation(s)
- Maria Pulina
- Present address: Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, NY 10065, USA
| | - Dong Liang
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA Present address: Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, NY 10065, USA
| | - Sophie Astrof
- Department of Medicine, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA Present address: Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
91
|
Turner CJ, Badu-Nkansah K, Crowley D, van der Flier A, Hynes RO. Integrin-α5β1 is not required for mural cell functions during development of blood vessels but is required for lymphatic-blood vessel separation and lymphovenous valve formation. Dev Biol 2014; 392:381-92. [PMID: 24858485 DOI: 10.1016/j.ydbio.2014.05.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 04/22/2014] [Accepted: 05/08/2014] [Indexed: 11/25/2022]
Abstract
Integrin α5β1 is essential for vascular development but it remains unclear precisely where and how it functions. Here, we report that deletion of the gene encoding the integrin-α5 subunit (Itga5) using the Pdgfrb-Cre transgenic mouse line, leads to oedema, haemorrhage and increased levels of embryonic lethality. Unexpectedly, these defects were not caused by loss of α5 from Pdgfrb-Cre expressing mural cells (pericytes and vascular smooth muscle cells), which wrap around the endothelium and stabilise blood vessels, nor by defects in the heart or great vessels, but were due to abnormal development of the lymphatic vasculature. Reminiscent of the pathologies seen in the human lymphatic malformation, fetal cystic hygroma, α5 mutants display defects both in the separation of their blood and lymphatic vasculature and in the formation of the lymphovenous valves. As a consequence, α5-deficient mice develop dilated, blood-filled lymphatic vessels and lymphatic capillaries that are ectopically covered with smooth muscle cells. Analysis of the expression of Pdgfrb during lymphatic development suggests that these defects probably arise from loss of α5β1 integrin in subsets of specialised Prox1(+)Pdgfrb(+) venous endothelial cells that are essential for the separation of the jugular lymph sac from the cardinal vein and formation of the lymphovenous valve leaflets.
Collapse
Affiliation(s)
- Christopher J Turner
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kwabena Badu-Nkansah
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Denise Crowley
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arjan van der Flier
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Richard O Hynes
- Howard Hughes Medical Institute, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
92
|
Kapp TG, Rechenmacher F, Sobahi TR, Kessler H. Integrin modulators: a patent review. Expert Opin Ther Pat 2014; 23:1273-95. [PMID: 24050747 DOI: 10.1517/13543776.2013.818133] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Integrins are heterodimeric cell surface receptors, which enable adhesion, proliferation, and migration of cells by recognizing binding motifs in extracellular matrix (ECM) proteins. As transmembrane linkers between the cytoskeleton and the ECM, they are able to recruit a huge variety of proteins and to influence signaling pathways bidirectionally, thereby regulating gene expression and cell survival. Hence, integrins play a key role in various physiological as well as pathological processes, which has turned them into an attractive target for pharmaceutical research. AREAS COVERED In this review, the latest therapeutic developments of drug candidates and recently patented integrin ligands are summarized. EXPERT OPINION Integrins have been proven to be valuable therapeutic targets in the treatment of several inflammatory and autoimmune diseases, where leukocyte adhesion processes are regulated by them. Furthermore, they play an important role in pathological angiogenesis and tumor metastasis, being a promising target for cancer therapy.
Collapse
Affiliation(s)
- Tobias G Kapp
- Institute for Advanced Study (IAS) and Center for Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München , Lichtenbergstrasse 4, 85747 Garching , Germany
| | | | | | | |
Collapse
|
93
|
Rechenmacher F, Steigerwald K, Laufer B, Neubauer S, Kapp TG, Li L, Mas-Moruno C, Joner M, Kessler H. The Integrin Ligandc(RGDf(NMe)Nal) Reduces Neointimal Hyperplasia in a Polymer-Free Drug-Eluting Stent System. ChemMedChem 2014; 9:1413-8. [DOI: 10.1002/cmdc.201400078] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Indexed: 01/28/2023]
|
94
|
Streijger F, Lee JHT, Duncan GJ, Ng MTL, Assinck P, Bhatnagar T, Plunet WT, Tetzlaff W, Kwon BK. Combinatorial treatment of acute spinal cord injury with ghrelin, ibuprofen, C16, and ketogenic diet does not result in improved histologic or functional outcome. J Neurosci Res 2014; 92:870-83. [PMID: 24658967 DOI: 10.1002/jnr.23372] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/28/2013] [Accepted: 01/20/2014] [Indexed: 11/09/2022]
Abstract
Because of the complex, multifaceted nature of spinal cord injury (SCI), it is widely believed that a combination of approaches will be superior to individual treatments. Therefore, we employed a rat model of cervical SCI to evaluate the combination of four noninvasive treatments that individually have been reported to be effective for acute SCI during clinically relevant therapeutic time windows. These treatments included ghrelin, ibuprofen, C16, and ketogenic diet (KD). These were selected not only because of their previously reported efficacy in SCI models but also for their potentially different mechanisms of action. The administration of ghrelin, ibuprofen, C16, and KD several hours to days postinjury was based on previous observations by others that each treatment had profound effects on the pathophysiology and functional outcome following SCI. Here we showed that, with the exception of a modest improvement in performance on the Montoya staircase test at 8-10 weeks postinjury, the combinatorial treatment with ghrelin, ibuprofen, C16, and KD did not result in any significant improvements in the rearing test, grooming test, or horizontal ladder. Histologic analysis of the spinal cords did not reveal any significant differences in tissue sparing between treatment and control groups. Although single approaches of ghrelin, ibuprofen, C16, and KD have been reported to be beneficial after SCI, our results show that the combination of the four interventions did not confer significant functional or histological improvements in a cervical model of SCI. Possible interactions among the treatments may have negated their beneficial effects, emphasizing the challenges that have to be addressed when considering combinatorial drug therapies for SCI.
Collapse
Affiliation(s)
- F Streijger
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada; Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Sheldrake HM, Patterson LH. Strategies to inhibit tumor associated integrin receptors: rationale for dual and multi-antagonists. J Med Chem 2014; 57:6301-15. [PMID: 24568695 DOI: 10.1021/jm5000547] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The integrins are a family of 24 heterodimeric transmembrane cell surface receptors. Involvement in cell attachment to the extracellular matrix, motility, and proliferation identifies integrins as therapeutic targets in cancer and associated conditions: thrombosis, angiogenesis, and osteoporosis. The most reported strategy for drug development is synthesis of an agent that is highly selective for a single integrin receptor. However, the ability of cancer cells to change their integrin repertoire in response to drug treatment renders this approach vulnerable to the development of resistance and paradoxical promotion of tumor growth. Here, we review progress toward development of antagonists targeting two or more members of the Arg-Gly-Asp (RGD) binding integrins, notably αvβ3, αvβ5, αvβ6, αvβ8, α5β1, and αIIbβ3, as anticancer therapeutics.
Collapse
Affiliation(s)
- Helen M Sheldrake
- Institute of Cancer Therapeutics, University of Bradford , Bradford, BD7 1DP, U.K
| | | |
Collapse
|
96
|
Kostourou V, Papalazarou V. Non-collagenous ECM proteins in blood vessel morphogenesis and cancer. Biochim Biophys Acta Gen Subj 2014; 1840:2403-13. [PMID: 24576673 DOI: 10.1016/j.bbagen.2014.02.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/14/2014] [Accepted: 02/17/2014] [Indexed: 12/12/2022]
Abstract
BACKGROUND The extracellular matrix (ECM) is constituted by diverse composite structures, which determine the specific to each organ, histological architecture and provides cells with biological information, mechanical support and a scaffold for adhesion and migration. The pleiotropic effects of the ECM stem from the dynamic changes in its molecular composition and the ability to remodel in order to effectively regulate biological outcomes. Besides collagens, fibronectin and laminin are two major fiber-forming constituents of various ECM structures. SCOPE OF REVIEW This review will focus on the properties and the biological functions of non-collagenous extracellular matrix especially on laminin and fibronectin that are currently emerging as important regulators of blood vessel formation and function in health and disease. MAJOR CONCLUSIONS The ECM is a fundamental component of the microenvironment of blood vessels, with activities extending beyond providing a vascular scaffold; extremely versatile it directly or indirectly modulates all essential cellular functions crucial for angiogenesis, including cell adhesion, migration, proliferation, differentiation and lumen formation. Specifically, fibronectin and laminins play decisive roles in blood vessel morphogenesis both during embryonic development and in pathological conditions, such as cancer. GENERAL SIGNIFICANCE Emerging evidence demonstrates the importance of ECM function during embryonic development, organ formation and tissue homeostasis. A wealth of data also illustrates the crucial role of the ECM in several human pathophysiological processes, including fibrosis, skeletal diseases, vascular pathologies and cancer. Notably, several ECM components have been identified as potential therapeutic targets for various diseases, including cancer. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Vassiliki Kostourou
- Vascular Adhesion Lab, BSRC Alexander Fleming, 34 Fleming Str., Vari, 166 72 Athens, Greece
| | - Vassilis Papalazarou
- Vascular Adhesion Lab, BSRC Alexander Fleming, 34 Fleming Str., Vari, 166 72 Athens, Greece
| |
Collapse
|
97
|
Ulyanova T, Padilla SM, Papayannopoulou T. Stage-specific functional roles of integrins in murine erythropoiesis. Exp Hematol 2014; 42:404-409.e4. [PMID: 24463276 DOI: 10.1016/j.exphem.2014.01.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 01/14/2014] [Indexed: 10/25/2022]
Abstract
When the erythroid integrins α5β1 and α4β1 were each deleted previously at the stem cell level, they yielded distinct physiologic responses to stress by affecting erythoid expansion and terminal differentiation or only the latter, respectively. To test at what stage of differentiation the integrin effects were exerted, we created mice with α4- or α5-integrin deletions only in erythroid cells and characterized them at homeostasis and after phenylhydrazine-induced hemolytic stress. Unlike our prior data, the phenotype of mice with α5-erythroid deletions was similar to controls, especially after stress. These outcomes seem to reconcile divergent prior views on the role of α5-integrin in erythropoiesis. By contrast, α4 integrins whether deleted early or late have a dominant effect on bone marrow retention of erythroblasts and on terminal erythroid maturation at homeostasis and after stress.
Collapse
Affiliation(s)
- Tatyana Ulyanova
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Steven M Padilla
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Thalia Papayannopoulou
- Division of Hematology, Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
98
|
Zeltz C, Orgel J, Gullberg D. Molecular composition and function of integrin-based collagen glues-introducing COLINBRIs. Biochim Biophys Acta Gen Subj 2013; 1840:2533-48. [PMID: 24361615 DOI: 10.1016/j.bbagen.2013.12.022] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 12/13/2013] [Accepted: 12/14/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND Despite detailed knowledge about the structure and signaling properties of individual collagen receptors, much remains to be learned about how these receptors participate in linking cells to fibrillar collagen matrices in tissues. In addition to collagen-binding integrins, a group of proteins with affinity both for fibrillar collagens and integrins link these two protein families together. We have introduced the name COLINBRI (COLlagen INtegrin BRIdging) for this set of molecules. Whereas collagens are the major building blocks in tissues and defects in these structural proteins have severe consequences for tissue integrity, the mild phenotypes of the integrin type of collagen receptors have raised questions about their importance in tissue biology and pathology. SCOPE OF REVIEW We will discuss the two types of cell linkages to fibrillar collagen (direct- versus indirect COLINBRI-mediated) and discuss how the parallel existence of direct and indirect linkages to collagens may ensure tissue integrity. MAJOR CONCLUSIONS The observed mild phenotypes of mice deficient in collagen-binding integrins and the relatively restricted availability of integrin-binding sequences in mature fibrillar collagen matrices support the existence of indirect collagen-binding mechanisms in parallel with direct collagen binding in vivo. GENERAL SIGNIFICANCE A continued focus on understanding the molecular details of cell adhesion mechanisms to collagens will be important and will benefit our understanding of diseases like tissue- and tumor fibrosis where collagen dynamics are disturbed. This article is part of a Special Issue entitled Matrix-mediated cell behaviour and properties.
Collapse
Affiliation(s)
- Cédric Zeltz
- Department of Biomedicine and Centre for Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway
| | - Joseph Orgel
- Departments of Biology, Physics and Biomedical Engineering, Pritzker Institute of Biomedical Science and Engineering, Illinois Institute of Technology, 3440 S. Dearborn Ave, Chicago, IL 60616, USA
| | - Donald Gullberg
- Department of Biomedicine and Centre for Cancer Biomarkers, Norwegian Centre of Excellence, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway.
| |
Collapse
|
99
|
Marelli UK, Rechenmacher F, Sobahi TRA, Mas-Moruno C, Kessler H. Tumor Targeting via Integrin Ligands. Front Oncol 2013; 3:222. [PMID: 24010121 PMCID: PMC3757457 DOI: 10.3389/fonc.2013.00222] [Citation(s) in RCA: 170] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 08/13/2013] [Indexed: 01/02/2023] Open
Abstract
Selective and targeted delivery of drugs to tumors is a major challenge for an effective cancer therapy and also to overcome the side-effects associated with current treatments. Overexpression of various receptors on tumor cells is a characteristic structural and biochemical aspect of tumors and distinguishes them from physiologically normal cells. This abnormal feature is therefore suitable for selectively directing anticancer molecules to tumors by using ligands that can preferentially recognize such receptors. Several subtypes of integrin receptors that are crucial for cell adhesion, cell signaling, cell viability, and motility have been shown to have an upregulated expression on cancer cells. Thus, ligands that recognize specific integrin subtypes represent excellent candidates to be conjugated to drugs or drug carrier systems and be targeted to tumors. In this regard, integrins recognizing the RGD cell adhesive sequence have been extensively targeted for tumor-specific drug delivery. Here we review key recent examples on the presentation of RGD-based integrin ligands by means of distinct drug-delivery systems, and discuss the prospects of such therapies to specifically target tumor cells.
Collapse
Affiliation(s)
- Udaya Kiran Marelli
- Institute for Advanced Study (IAS) and Center for Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München , Garching , Germany
| | | | | | | | | |
Collapse
|
100
|
Overstreet MG, Gaylo A, Angermann BR, Hughson A, Hyun YM, Lambert K, Acharya M, Billroth-Maclurg AC, Rosenberg AF, Topham DJ, Yagita H, Kim M, Lacy-Hulbert A, Meier-Schellersheim M, Fowell DJ. Inflammation-induced interstitial migration of effector CD4⁺ T cells is dependent on integrin αV. Nat Immunol 2013; 14:949-58. [PMID: 23933892 PMCID: PMC4159184 DOI: 10.1038/ni.2682] [Citation(s) in RCA: 129] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 07/02/2013] [Indexed: 12/14/2022]
Abstract
Leukocytes must traverse inflamed tissues to effectively control local infection. Although motility in dense tissues appears to be integrin-independent actin-myosin based, during inflammation changes to the extracellular matrix (ECM) may necessitate distinct motility requirements. Indeed, we found that T cell interstitial motility was critically dependent on RGD-binding integrins in the inflamed dermis. Inflammation-induced deposition of fibronectin was functionally linked to increased αv integrin expression on effector CD4+ T cells. Using intravital multi-photon imaging, we found that CD4+ T cell motility was dependent on αv expression. Selective αv blockade or knockdown arrested TH1 motility in the inflamed tissue and attenuated local effector function. These data show a context-dependent specificity of lymphocyte movement in inflamed tissues that is essential for protective immunity.
Collapse
Affiliation(s)
- Michael G Overstreet
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester, Rochester, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|