1
|
Bisen S, Gogoi P, Sharma A, Mukhopadhyay CS, Singh NK. A Disintegrin and Metalloproteinase 10 Regulates Ephrin B2-Mediated Endothelial Cell Sprouting and Ischemic Retinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00114-2. [PMID: 40252970 DOI: 10.1016/j.ajpath.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 03/14/2025] [Accepted: 03/21/2025] [Indexed: 04/21/2025]
Abstract
Retinal neovascularization is the leading cause of visual impairment in diabetic retinopathy, retinopathy of prematurity, and age-related macular degeneration. The extracellular matrix breakdown by metalloproteinase leads to vascular complications in various proliferative retinopathies. A disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) is involved in physiological angiogenesis. However, limited information exists regarding the role of ADAM10 in proliferative retinopathies. In this study, the levels of active ADAM10 were significantly up-regulated in the ischemic retina, and down-regulation or inactivation of ADAM10 significantly inhibited the proliferation, sprouting, migration, and tube formation of human retinal microvascular endothelial cell. Furthermore, the endothelial cell (EC)-specific deletion of ADAM10 (ADAM10iΔEC) significantly attenuated vascular leakage, edema, endothelial cell sprouting, and retinal neovascularization in ischemic retinas of mice exposed to oxygen-induced retinopathy. In experiments investigating the mechanisms through which ADAM10 regulated pathologic angiogenesis, ADAM10 regulated ephrin B2 (EfnB2) expression in endothelial cells. Down-regulation of EfnB2 expression influenced human retinal microvascular endothelial cell proliferation, migration, sprouting, and tube formation. In addition, a significant up-regulation of EfnB2 expression in the ischemic retina was detected. EC-specific depletion of ADAM10 significantly reduced EfnB2 expression, suggesting its involvement in ADAM10-regulated retinal neovascularization. The findings demonstrate how EC-specific ADAM10 regulates pathologic retinal neovascularization in the ischemic retina, indicating its significance as a potential therapeutic target for proliferative retinopathies.
Collapse
Affiliation(s)
- Shivantika Bisen
- Integrative Biosciences Center, Wayne State University, Detroit, Michigan; Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan
| | - Purnima Gogoi
- Integrative Biosciences Center, Wayne State University, Detroit, Michigan; Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan; Department of Medical Genetics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin
| | - Anamika Sharma
- Integrative Biosciences Center, Wayne State University, Detroit, Michigan; Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan
| | - Chandra S Mukhopadhyay
- Department of Bioinformatics, College of Animal Biotechnology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| | - Nikhlesh K Singh
- Integrative Biosciences Center, Wayne State University, Detroit, Michigan; Department of Ophthalmology, Visual and Anatomical Sciences, School of Medicine, Wayne State University, Detroit, Michigan.
| |
Collapse
|
2
|
Badouel C, Audouard C, Davy A. Heterogeneity in the size of the apical surface of cortical progenitors. Dev Dyn 2023; 252:363-376. [PMID: 36153792 DOI: 10.1002/dvdy.539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The apical surface (AS) of epithelial cells is highly specialized; it is important for morphogenetic processes that are essential to shape organs and tissues and it plays a role in morphogen and growth factor signaling. Apical progenitors in the mammalian neocortex are pseudoepithelial cells whose apical surface lines the ventricle. Whether changes in their apical surface sizes are important for cortical morphogenesis and/or other aspects of neocortex development has not been thoroughly addressed. RESULTS Here we show that apical progenitors are heterogeneous with respect to their apical surface area. In Efnb1 mutants, the size of the apical surface is modified and this correlates with discrete alterations of tissue organization without impacting apical progenitors proliferation. CONCLUSIONS Altogether, our data reveal heterogeneity in apical progenitors AS area in the developing neocortex and shows a role for Ephrin B1 in controlling AS size. Our study also indicates that changes in AS size do not have strong repercussion on apical progenitor behavior.
Collapse
Affiliation(s)
- Caroline Badouel
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Christophe Audouard
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, Université de Toulouse, Toulouse, France
| | - Alice Davy
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), CNRS, UPS, Université de Toulouse, Toulouse, France
| |
Collapse
|
3
|
Yoon J, Sun J, Lee M, Hwang YS, Daar IO. Wnt4 and ephrinB2 instruct apical constriction via Dishevelled and non-canonical signaling. Nat Commun 2023; 14:337. [PMID: 36670115 PMCID: PMC9860048 DOI: 10.1038/s41467-023-35991-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 01/11/2023] [Indexed: 01/22/2023] Open
Abstract
Apical constriction is a cell shape change critical to vertebrate neural tube closure, and the contractile force required for this process is generated by actin-myosin networks. The signaling cue that instructs this process has remained elusive. Here, we identify Wnt4 and the transmembrane ephrinB2 protein as playing an instructive role in neural tube closure as members of a signaling complex we termed WERDS (Wnt4, EphrinB2, Ror2, Dishevelled (Dsh2), and Shroom3). Disruption of function or interaction among members of the WERDS complex results in defects of apical constriction and neural tube closure. The mechanism of action involves an interaction of ephrinB2 with the Dsh2 scaffold protein that enhances the formation of the WERDS complex, which in turn, activates Rho-associated kinase to induce apical constriction. Moreover, the ephrinB2/Dsh2 interaction promotes non-canonical Wnt signaling and shows how cross-talk between two major signal transduction pathways, Eph/ephrin and Wnt, coordinate morphogenesis of the neural tube.
Collapse
Affiliation(s)
- Jaeho Yoon
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA.
| | - Jian Sun
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Moonsup Lee
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Yoo-Seok Hwang
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | - Ira O Daar
- Cancer & Developmental Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA.
| |
Collapse
|
4
|
Wisniewski DJ, Liyasova MS, Korrapati S, Zhang X, Ratnayake S, Chen Q, Gilbert SF, Catalano A, Voeller D, Meerzaman D, Guha U, Porat-Shliom N, Annunziata CM, Lipkowitz S. Flotillin-2 regulates epidermal growth factor receptor activation, degradation by Cbl-mediated ubiquitination, and cancer growth. J Biol Chem 2023; 299:102766. [PMID: 36470425 PMCID: PMC9823131 DOI: 10.1016/j.jbc.2022.102766] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/08/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) signaling is frequently dysregulated in various cancers. The ubiquitin ligase Casitas B-lineage lymphoma proto-oncogene (Cbl) regulates degradation of activated EGFR through ubiquitination and acts as an adaptor to recruit proteins required for trafficking. Here, we used stable isotope labeling with amino acids in cell culture mass spectrometry to compare Cbl complexes with or without epidermal growth factor (EGF) stimulation. We identified over a hundred novel Cbl interactors, and a secondary siRNA screen found that knockdown of Flotillin-2 (FLOT2) led to increased phosphorylation and degradation of EGFR upon EGF stimulation in HeLa cells. In PC9 and H441 cells, FLOT2 knockdown increased EGF-stimulated EGFR phosphorylation, ubiquitination, and downstream signaling, reversible by EGFR inhibitor erlotinib. CRISPR knockout (KO) of FLOT2 in HeLa cells confirmed EGFR downregulation, increased signaling, and increased dimerization and endosomal trafficking. Furthermore, we determined that FLOT2 interacted with both Cbl and EGFR. EGFR downregulation upon FLOT2 loss was Cbl dependent, as coknockdown of Cbl and Cbl-b restored EGFR levels. In addition, FLOT2 overexpression decreased EGFR signaling and growth. Overexpression of wildtype (WT) FLOT2, but not the soluble G2A FLOT2 mutant, inhibited EGFR phosphorylation upon EGF stimulation in HEK293T cells. FLOT2 loss induced EGFR-dependent proliferation and anchorage-independent growth. Lastly, FLOT2 KO increased tumor formation and tumor volume in nude mice and NSG mice, respectively. Together, these data demonstrated that FLOT2 negatively regulated EGFR activation and dimerization, as well as its subsequent ubiquitination, endosomal trafficking, and degradation, leading to reduced proliferation in vitro and in vivo.
Collapse
Affiliation(s)
- David J Wisniewski
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Mariya S Liyasova
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Soumya Korrapati
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Xu Zhang
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Shashikala Ratnayake
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, Maryland, USA
| | - Qingrong Chen
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, Maryland, USA
| | - Samuel F Gilbert
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Alexis Catalano
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Donna Voeller
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Daoud Meerzaman
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Rockville, Maryland, USA
| | - Udayan Guha
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Natalie Porat-Shliom
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Christina M Annunziata
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Stanley Lipkowitz
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA.
| |
Collapse
|
5
|
Chandrasekera P, Perfetto M, Lu C, Zhuo M, Bahudhanapati H, Li J, Chen WC, Kulkarni P, Christian L, Liu J, Yien YY, Yu C, Wei S. Metalloprotease ADAM9 cleaves ephrin-B ligands and differentially regulates Wnt and mTOR signaling downstream of Akt kinase in colorectal cancer cells. J Biol Chem 2022; 298:102225. [PMID: 35780836 PMCID: PMC9358476 DOI: 10.1016/j.jbc.2022.102225] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/13/2022] [Accepted: 06/21/2022] [Indexed: 12/02/2022] Open
Abstract
Ephrin-B signaling has been implicated in many normal and pathological processes, including neural crest development and tumor metastasis. We showed previously that proteolysis of ephrin-B ligands by the disintegrin metalloprotease ADAM13 is necessary for canonical Wnt signal activation and neural crest induction in Xenopus, but it was unclear if these mechanisms are conserved in mammals. Here, we report that mammalian ADAM9 cleaves ephrin-B1 and ephrin-B2 and can substitute for Xenopus ADAM13 to induce the neural crest. We found that ADAM9 expression is elevated in human colorectal cancer (CRC) tissues and that knockdown (KD) of ADAM9 inhibits the migration and invasion of SW620 and HCT116 CRC cells by reducing the activity of Akt kinase, which is antagonized by ephrin-Bs. Akt is a signaling node that activates multiple downstream pathways, including the Wnt and mTOR pathways, both of which can promote CRC cell migration/invasion. Surprisingly, we also found that KD of ADAM9 downregulates Wnt signaling but has negligible effects on mTOR signaling in SW620 cells; in contrast, mTOR activity is suppressed while Wnt signaling remains unaffected by ADAM9 KD in HCT116 cells. These results suggest that mammalian ADAM9 cleaves ephrin-Bs to derepress Akt and promote CRC migration and invasion; however, the signaling pathways downstream of Akt are differentially regulated by ADAM9 in different CRC cell lines, reflecting the heterogeneity of CRC cells in responding to manipulations of upstream Akt regulators.
Collapse
Affiliation(s)
| | - Mark Perfetto
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA; Department of Biology, West Virginia University, Morgantown, West Virginia, USA; Pittsburgh Heart, Lung and Blood Vascular Medicine Institute and Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Congyu Lu
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Minghui Zhuo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | | | - Jiejing Li
- Department of Biology, West Virginia University, Morgantown, West Virginia, USA; Department of Clinical Laboratory, The Affiliated Hospital of KMUST, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Wei-Chih Chen
- Department of Biology, West Virginia University, Morgantown, West Virginia, USA
| | - Pallavi Kulkarni
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Laura Christian
- Department of Biology, West Virginia University, Morgantown, West Virginia, USA
| | - Jun Liu
- Department of Biochemistry and Cancer Institute, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | - Yvette Y Yien
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA; Pittsburgh Heart, Lung and Blood Vascular Medicine Institute and Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, China
| | - Shuo Wei
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA.
| |
Collapse
|
6
|
Eph and Ephrin Variants in Malaysian Neural Tube Defect Families. Genes (Basel) 2022; 13:genes13060952. [PMID: 35741713 PMCID: PMC9222557 DOI: 10.3390/genes13060952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/20/2022] [Accepted: 05/23/2022] [Indexed: 02/01/2023] Open
Abstract
Neural tube defects (NTDs) are common birth defects with a complex genetic etiology. Mouse genetic models have indicated a number of candidate genes, of which functional mutations in some have been found in human NTDs, usually in a heterozygous state. This study focuses on Ephs-ephrins as candidate genes of interest owing to growing evidence of the role of this gene family during neural tube closure in mouse models. Eph-ephrin genes were analyzed in 31 Malaysian individuals comprising seven individuals with sporadic spina bifida, 13 parents, one twin-sibling and 10 unrelated controls. Whole exome sequencing analysis and bioinformatic analysis were performed to identify variants in 22 known Eph-ephrin genes. We reported that three out of seven spina bifida probands and three out of thirteen family members carried a variant in either EPHA2 (rs147977279), EPHB6 (rs780569137) or EFNB1 (rs772228172). Analysis of public databases shows that these variants are rare. In exome datasets of the probands and parents of the probands with Eph-ephrin variants, the genotypes of spina bifida-related genes were compared to investigate the probability of the gene–gene interaction in relation to environmental risk factors. We report the presence of Eph-ephrin gene variants that are prevalent in a small cohort of spina bifida patients in Malaysian families.
Collapse
|
7
|
Abstract
The EPH receptor tyrosine kinases and their signaling partners, the EPHRINS, comprise a large class of cell signaling molecules that plays diverse roles in development. As cell membrane-anchored signaling molecules, they regulate cellular organization by modulating the strength of cellular contacts, usually by impacting the actin cytoskeleton or cell adhesion programs. Through these cellular functions, EPH/EPHRIN signaling often regulates tissue shape. Indeed, recent evidence indicates that this signaling family is ancient and associated with the origin of multicellularity. Though extensively studied, our understanding of the signaling mechanisms employed by this large family of signaling proteins remains patchwork, and a truly "canonical" EPH/EPHRIN signal transduction pathway is not known and may not exist. Instead, several foundational evolutionarily conserved mechanisms are overlaid by a myriad of tissue -specific functions, though common themes emerge from these as well. Here, I review recent advances and the related contexts that have provided new understanding of the conserved and varied molecular and cellular mechanisms employed by EPH/EPHRIN signaling during development.
Collapse
Affiliation(s)
- Jeffrey O Bush
- Department of Cell and Tissue Biology, University of California San Francisco, San Francisco, CA, United States; Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, United States; Institute for Human Genetics, University of California San Francisco, San Francisco, CA, United States; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, United States.
| |
Collapse
|
8
|
Abstract
Apical constriction refers to the active, actomyosin-driven process that reduces apical cell surface area in epithelial cells. Apical constriction is utilized in epithelial morphogenesis during embryonic development in multiple contexts, such as gastrulation, neural tube closure, and organogenesis. Defects in apical constriction can result in congenital birth defects, yet our understanding of the molecular control of apical constriction is relatively limited. To uncover new genetic regulators of apical constriction and gain mechanistic insight into the cell biology of this process, we need reliable assay systems that allow real-time observation and quantification of apical constriction as it occurs and permit gain- and loss-of-function analyses to explore gene function and interaction during apical constriction. In this chapter, we describe using the early Xenopus embryo as an assay system to investigate molecular mechanisms involved in apical constriction during both gastrulation and neurulation.
Collapse
Affiliation(s)
- Austin T Baldwin
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | - Ivan K Popov
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - John B Wallingford
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA.
| | - Chenbei Chang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
9
|
Yu Z, Li W, Lan J, Hayakawa K, Ji X, Lo EH, Wang X. EphrinB2-EphB2 signaling for dendrite protection after neuronal ischemia in vivo and oxygen-glucose deprivation in vitro. J Cereb Blood Flow Metab 2021; 41:1744-1755. [PMID: 33325764 PMCID: PMC8221775 DOI: 10.1177/0271678x20973119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
In order to rescue neuronal function, neuroprotection should be required not only for the neuron soma but also the dendrites. Here, we propose the hypothesis that ephrin-B2-EphB2 signaling may be involved in dendritic degeneration after ischemic injury. A mouse model of focal cerebral ischemia with middle cerebral artery occlusion (MCAO) method was used for EphB2 signaling test in vivo. Primary cortical neuron culture and oxygen-glucose deprivation were used to assess EphB2 signaling in vitro. siRNA and soluble ephrin-B2 ectodomain were used to block ephrin-B2-Ephb2 signaling. In the mouse model of focal cerebral ischemia and in neurons subjected to oxygen-glucose deprivation, clustering of ephrin-B2 with its receptor EphB2 was detected. Phosphorylation of EphB2 suggested activation of this signaling pathway. RNA silencing of EphB2 prevented neuronal death and preserved dendritic length. To assess therapeutic potential, we compared the soluble EphB2 ectodomain with the NMDA antagonist MK801 in neurons after oxygen-glucose deprivation. Both agents equally reduced lactate dehydrogenase release as a general marker of neurotoxicity. However, only soluble EphB2 ectodomain protected the dendrites. These findings provide a proof of concept that ephrin-B2-EphB2 signaling may represent a novel therapeutic target to protect both the neuron soma as well as dendrites against ischemic injury.
Collapse
Affiliation(s)
- Zhanyang Yu
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Wenlu Li
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Jing Lan
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.,Cerebrovascular Research Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Kazuhide Hayakawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China.,Cerebrovascular Research Institute, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Xiaoying Wang
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA.,Clinical Neuroscience Research Center, Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
10
|
Tosetti F, Alessio M, Poggi A, Zocchi MR. ADAM10 Site-Dependent Biology: Keeping Control of a Pervasive Protease. Int J Mol Sci 2021; 22:4969. [PMID: 34067041 PMCID: PMC8124674 DOI: 10.3390/ijms22094969] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/04/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
Enzymes, once considered static molecular machines acting in defined spatial patterns and sites of action, move to different intra- and extracellular locations, changing their function. This topological regulation revealed a close cross-talk between proteases and signaling events involving post-translational modifications, membrane tyrosine kinase receptors and G-protein coupled receptors, motor proteins shuttling cargos in intracellular vesicles, and small-molecule messengers. Here, we highlight recent advances in our knowledge of regulation and function of A Disintegrin And Metalloproteinase (ADAM) endopeptidases at specific subcellular sites, or in multimolecular complexes, with a special focus on ADAM10, and tumor necrosis factor-α convertase (TACE/ADAM17), since these two enzymes belong to the same family, share selected substrates and bioactivity. We will discuss some examples of ADAM10 activity modulated by changing partners and subcellular compartmentalization, with the underlying hypothesis that restraining protease activity by spatial segregation is a complex and powerful regulatory tool.
Collapse
Affiliation(s)
- Francesca Tosetti
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico S. Martino Largo R. Benzi 10, 16132 Genoa, Italy;
| | - Massimo Alessio
- Proteome Biochemistry, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| | - Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico S. Martino Largo R. Benzi 10, 16132 Genoa, Italy;
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy;
| |
Collapse
|
11
|
Yoon J, Garo J, Lee M, Sun J, Hwang YS, Daar IO. Rab11fip5 regulates telencephalon development via ephrinB1 recycling. Development 2021; 148:dev196527. [PMID: 33462110 PMCID: PMC7875491 DOI: 10.1242/dev.196527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/25/2020] [Indexed: 12/12/2022]
Abstract
Rab11 family-interacting protein 5 (Rab11fip5) is an adaptor protein that binds to the small GTPase Rab11, which has an important function in endosome recycling and trafficking of cellular proteins to the plasma membrane. Rab11fip5 is involved in many cellular processes, such as cytoskeleton rearrangement, iron uptake and exocytosis in neuroendocrine cells, and is also known as a candidate gene for autism-spectrum disorder. However, the role of Rab11fip5 during early embryonic development is not clearly understood. In this study, we identified Rab11fip5 as a protein that interacts with ephrinB1, a transmembrane ligand for Eph receptors. The PDZ binding motif in ephrinB1 and the Rab-binding domain in Rab11fip5 are necessary for their interaction in a complex. EphrinB1 and Rab11fip5 display overlapping expression in the telencephalon of developing amphibian embryos. The loss of Rab11fip5 function causes a reduction in telencephalon size and a decrease in the expression level of ephrinB1. Moreover, morpholino oligonucleotide-mediated knockdown of Rab11fip5 decreases cell proliferation in the telencephalon. The overexpression of ephrinB1 rescues these defects, suggesting that ephrinB1 recycling by the Rab11/Rab11fip5 complex is crucial for proper telencephalon development.
Collapse
Affiliation(s)
- Jaeho Yoon
- Cancer and Developmental Biology Laboratory (CDBL), Center for Cancer Research (CCR) - Frederick, National Cancer Institute, Frederick, MD 21702, USA
| | - Jerlin Garo
- Cancer and Developmental Biology Laboratory (CDBL), Center for Cancer Research (CCR) - Frederick, National Cancer Institute, Frederick, MD 21702, USA
| | - Moonsup Lee
- Cancer and Developmental Biology Laboratory (CDBL), Center for Cancer Research (CCR) - Frederick, National Cancer Institute, Frederick, MD 21702, USA
| | - Jian Sun
- Cancer and Developmental Biology Laboratory (CDBL), Center for Cancer Research (CCR) - Frederick, National Cancer Institute, Frederick, MD 21702, USA
| | - Yoo-Seok Hwang
- Cancer and Developmental Biology Laboratory (CDBL), Center for Cancer Research (CCR) - Frederick, National Cancer Institute, Frederick, MD 21702, USA
| | - Ira O Daar
- Cancer and Developmental Biology Laboratory (CDBL), Center for Cancer Research (CCR) - Frederick, National Cancer Institute, Frederick, MD 21702, USA
| |
Collapse
|
12
|
Mueller AC, Piper M, Goodspeed A, Bhuvane S, Williams JS, Bhatia S, Phan AV, Van Court B, Zolman KL, Peña B, Oweida AJ, Zakem S, Meguid C, Knitz MW, Darragh L, Bickett TE, Gadwa J, Mestroni L, Taylor MRG, Jordan KR, Dempsey P, Lucia MS, McCarter MD, Chiaro MD, Messersmith WA, Schulick RD, Goodman KA, Gough MJ, Greene CS, Costello JC, Neto AG, Lagares D, Hansen KC, Van Bokhoven A, Karam SD. Induction of ADAM10 by Radiation Therapy Drives Fibrosis, Resistance, and Epithelial-to-Mesenchyal Transition in Pancreatic Cancer. Cancer Res 2021; 81:3255-3269. [PMID: 33526513 DOI: 10.1158/0008-5472.can-20-3892] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/18/2020] [Accepted: 01/27/2021] [Indexed: 02/07/2023]
Abstract
Stromal fibrosis activates prosurvival and proepithelial-to-mesenchymal transition (EMT) pathways in pancreatic ductal adenocarcinoma (PDAC). In patient tumors treated with neoadjuvant stereotactic body radiation therapy (SBRT), we found upregulation of fibrosis, extracellular matrix (ECM), and EMT gene signatures, which can drive therapeutic resistance and tumor invasion. Molecular, functional, and translational analysis identified two cell-surface proteins, a disintegrin and metalloprotease 10 (ADAM10) and ephrinB2, as drivers of fibrosis and tumor progression after radiation therapy (RT). RT resulted in increased ADAM10 expression in tumor cells, leading to cleavage of ephrinB2, which was also detected in plasma. Pharmacologic or genetic targeting of ADAM10 decreased RT-induced fibrosis and tissue tension, tumor cell migration, and invasion, sensitizing orthotopic tumors to radiation killing and prolonging mouse survival. Inhibition of ADAM10 and genetic ablation of ephrinB2 in fibroblasts reduced the metastatic potential of tumor cells after RT. Stimulation of tumor cells with ephrinB2 FC protein reversed the reduction in tumor cell invasion with ADAM10 ablation. These findings represent a model of PDAC adaptation that explains resistance and metastasis after RT and identifies a targetable pathway to enhance RT efficacy. SIGNIFICANCE: Targeting a previously unidentified adaptive resistance mechanism to radiation therapy in PDAC tumors in combination with radiation therapy could increase survival of the 40% of PDAC patients with locally advanced disease.See related commentary by Garcia Garcia et al., p. 3158 GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/12/3255/F1.large.jpg.
Collapse
Affiliation(s)
- Adam C Mueller
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.,Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Miles Piper
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Andrew Goodspeed
- Department of Pharmacology, University of Colorado Comprehensive Cancer Center, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Shiv Bhuvane
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Jason S Williams
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Shilpa Bhatia
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Andy V Phan
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Benjamin Van Court
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Kathryn L Zolman
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Brisa Peña
- Department of Cardiology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Ayman J Oweida
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.,Département de médecine nucléaire et radiobiologie, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Sara Zakem
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Cheryl Meguid
- Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Michael W Knitz
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Laurel Darragh
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Thomas E Bickett
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Jacob Gadwa
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Luisa Mestroni
- Department of Cardiology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Matthew R G Taylor
- Department of Cardiology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Kimberly R Jordan
- Human Immune Monitoring Shared Resource, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Peter Dempsey
- Department of Gastroenterology, Hepatology and Nutrition, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - M Scott Lucia
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Martin D McCarter
- Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Marco Del Chiaro
- Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Wells A Messersmith
- Department of Medical Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Richard D Schulick
- Department of Surgery, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Karyn A Goodman
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.,Department of Radiation Oncology, Mount Sinai Hospital, New York, New York
| | | | - Casey S Greene
- Center for Health Artificial Intelligence, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - James C Costello
- Department of Pharmacology, University of Colorado Comprehensive Cancer Center, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Antonio Galveo Neto
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - David Lagares
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Charlestown, Massachusetts
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Adrie Van Bokhoven
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado, Anschutz Medical Campus, Aurora, Colorado.
| |
Collapse
|
13
|
Hodeify R, Dib M, Alcantara-Adap E, Courjaret R, Nader N, Reyes CZ, Hammad AS, Hubrack S, Yu F, Machaca K. The carboxy terminal coiled-coil modulates Orai1 internalization during meiosis. Sci Rep 2021; 11:2290. [PMID: 33504898 PMCID: PMC7840751 DOI: 10.1038/s41598-021-82048-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/30/2020] [Indexed: 12/19/2022] Open
Abstract
Regulation of Ca2+ signaling is critical for the progression of cell division, especially during meiosis to prepare the egg for fertilization. The primary Ca2+ influx pathway in oocytes is Store-Operated Ca2+ Entry (SOCE). SOCE is tightly regulated during meiosis, including internalization of the SOCE channel, Orai1. Orai1 is a four-pass membrane protein with cytosolic N- and C-termini. Orai1 internalization requires a caveolin binding motif (CBM) in the N-terminus as well as the C-terminal cytosolic domain. However, the molecular determinant for Orai1 endocytosis in the C-terminus are not known. Here we show that the Orai1 C-terminus modulates Orai1 endocytosis during meiosis through a structural motif that is based on the strength of the C-terminal intersubunit coiled coil (CC) domains. Deletion mutants show that a minimal C-terminal sequence after transmembrane domain 4 (residues 260–275) supports Orai1 internalization. We refer to this region as the C-terminus Internalization Handle (CIH). Access to CIH however is dependent on the strength of the intersubunit CC. Mutants that increase the stability of the coiled coil prevent internalization independent of specific mutation. We further used human and Xenopus Orai isoforms with different propensity to form C-terminal CC and show a strong correlation between the strength of the CC and Orai internalization. Furthermore, Orai1 internalization does not depend on clathrin, flotillin or PIP2. Collectively these results argue that Orai1 internalization requires both the N-terminal CBM and C-terminal CIH where access to CIH is controlled by the strength of intersubunit C-terminal CC.
Collapse
Affiliation(s)
- Rawad Hodeify
- Department of Physiology and Biophysics, Ca2+ signaling Group, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar.,Department of Biotechnology, American University of Ras Al Khaimah, Ras al Khaimah, UAE
| | - Maya Dib
- Department of Physiology and Biophysics, Ca2+ signaling Group, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Ethel Alcantara-Adap
- Department of Physiology and Biophysics, Ca2+ signaling Group, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Raphael Courjaret
- Department of Physiology and Biophysics, Ca2+ signaling Group, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Nancy Nader
- Department of Physiology and Biophysics, Ca2+ signaling Group, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Cleo Z Reyes
- Department of Physiology and Biophysics, Ca2+ signaling Group, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar.,Lehigh Valley Health Network, Allentown, PA, USA
| | - Ayat S Hammad
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Satanay Hubrack
- Department of Physiology and Biophysics, Ca2+ signaling Group, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar.,Sidra Medicine, Doha, Qatar
| | - Fang Yu
- Department of Physiology and Biophysics, Ca2+ signaling Group, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Khaled Machaca
- Department of Physiology and Biophysics, Ca2+ signaling Group, Weill Cornell Medicine Qatar, Education City, Qatar Foundation, Doha, Qatar.
| |
Collapse
|
14
|
Wang Q, Liu Y, Zhao Y, Sun LZ, Wang LX, Han M, Mi FL. [Research progress on the expression and function of erythropoietin-producing hepatomocellular receptors and their receptor-interacting proteins in oral-related diseases]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2020; 38:218-223. [PMID: 32314898 DOI: 10.7518/hxkq.2020.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Erythropoietin-producing hepatomocellular receptors and their receptor-interacting proteins (Eph/ephrin) can participate in the regulation of growth and development and promote the development of diseases through short-distance signal transduction between cells. To study the mechanism of Eph/ephrin and oral-related diseases, we provided a new theoretical basis and a strategy for the treatment of oral diseases. The Eph/ephrin pathway has been used to regulate oral diseases, especially in periodontal disease prevention, orthodontic bone reconstruction, and biological treatment of oral tumors. This paper reviews the research progress of Eph/ephrin pathway in oral-related diseases.
Collapse
Affiliation(s)
- Qi Wang
- Dept. of Stomatology, North Sichuan Medical College, Nanchong 637000, China
| | - Yan Liu
- Dept. of Prosthodontics, the Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Yun Zhao
- Dept. of Stomatology, North Sichuan Medical College, Nanchong 637000, China
| | - Li-Zhong Sun
- Dept. of Stomatology, North Sichuan Medical College, Nanchong 637000, China
| | - Lin-Xuan Wang
- Dept. of Stomatology, North Sichuan Medical College, Nanchong 637000, China
| | - Mei Han
- Dept. of Stomatology, North Sichuan Medical College, Nanchong 637000, China
| | - Fang-Lin Mi
- Dept. of Stomatology, North Sichuan Medical College, Nanchong 637000, China
| |
Collapse
|
15
|
Deng F, Miller J. A review on protein markers of exosome from different bio-resources and the antibodies used for characterization. J Histotechnol 2019; 42:226-239. [PMID: 31432761 DOI: 10.1080/01478885.2019.1646984] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Exosomes are small membrane vesicles (ranging from 30 nm to 150 nm), secreted by different cell types upon fusion of multivesicular bodies (MVB) to the cell plasma membrane under a variety of normal and pathological conditions. Through transferring their cargos such as proteins, lipids and nucleic acids from donor cells to recipient cells, exosomes play a crucial role in cell-to-cell communication. Due to their presence in most body fluids (such as blood, breast milk, saliva, urine, bile, pancreatic juice, cerebrospinal and peritoneal fluids), and their role in carrying bioactive molecules from the cells of origin, exosomes have attracted great interest in their diagnostic and prognostic value for various diseases and therapeutic approaches. Although a large body of literature has documented the importance of exosomes over the past decade, there is no article systematically summarizing protein markers of exosome from different resources and the antibodies that are suited to characterize exosomes. In this review, we briefly summarize the exosome marker proteins, exosomal biomarkers for different diseases, and the antibodies suitable for different bio-resources exosomes characterization.
Collapse
Affiliation(s)
- Fengyan Deng
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, MO, USA
| | - Josh Miller
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, MO, USA
| |
Collapse
|
16
|
Su SA, Xie Y, Zhang Y, Xi Y, Cheng J, Xiang M. Essential roles of EphrinB2 in mammalian heart: from development to diseases. Cell Commun Signal 2019; 17:29. [PMID: 30909943 PMCID: PMC6434800 DOI: 10.1186/s12964-019-0337-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/11/2019] [Indexed: 12/11/2022] Open
Abstract
EphrinB2, a membrane-tethered ligand preferentially binding to its receptor EphB4, is ubiquitously expressed in all mammals. Through the particular bidirectional signaling, EphrinB2 plays a critical role during the development of cardiovascular system, postnatal angiogenesis physiologically and pathologically, and cardiac remodeling after injuries as an emerging role. This review highlights the pivotal involvement of EphrinB2 in heart, from developmental cardiogenesis to pathological cardiac remodeling process. Further potential translational therapies will be discussed in targeting EphrinB2 signaling, to better understand the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Sheng-An Su
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yuhao Zhang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yutao Xi
- Texas Heart Institute, Houston, 77030, USA.
| | - Jie Cheng
- Texas Heart Institute, Houston, 77030, USA
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
17
|
Li X, Dardik A, Guo R, Zhang W, Xiang Y, Li S, Liu D, Shu C. Atorvastatin regulates pericardial patch healing via the microRNA140-ADAM10-ephrinB2 pathway. Am J Transl Res 2018; 10:4054-4064. [PMID: 30662650 PMCID: PMC6325491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 11/12/2018] [Indexed: 06/09/2023]
Abstract
Background: Pericardial patches are frequently used in vascular surgery to close arteriotomies. The early healing of these patches is mediated by attraction of CD34 and ephrinB2-positive cells. Atorvastatin is a commonly used statin drug that promotes healing of cardiovascular injury. We hypothesized that atorvastatin attracts ephrinB2-positive cells by regulating the microRNA140-ADAM10-ephrinB2 pathway during patch healing in the arterial environment. Methods: Pericardial patches were used to close an infra-renal aortic arteriotomy in Wistar rats (male, 200-400 g). Atorvastatin was given to rats at a daily dose of 0 mg, 2.5 mg, 5 mg or 10 mg. Patches were harvested at 1 or 4 weeks and analyzed by histology, immunohistochemistry, immunofluorescence, western blot and qPCR. Result: Animals treated with atorvastatin showed a higher number of infiltrating cells and a thicker patch neointima than the control animals. Furthermore, ADAM10 protein expression decreased (P<0.01) and ephrinB2 expression increased (P<0.01) in time- and atorvastatin dose-dependent manner. Similarly, ADAM10 mRNA expression decreased (P<0.01), while the expression of ephrinB2 mRNA and miR-140 mRNA expression increased (P<0.01; P<0.01) in a time- and dose-dependent manner. Conclusion: Atorvastatin regulates neointimal growth after pericardial patch angioplasty; atorvastatin is associated with infiltration of ephrinB2-positive cells, diminished ADAM10 expression, and increased ephrinB2 and miR-140 expression. These results suggest new mechanisms for regulating neointimal formation after vascular procedures. Clinical relevance: This study may help physicians to know more healing mechanism after pericardial patch angioplasty. Further, it may reveal some mechanism that how atorvastatin play roles in endothelium repair of the cardiovascular system.
Collapse
Affiliation(s)
- Xin Li
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South UniversityChangsha 410011, Hunan, PR China
- Vascular Disease Institute, Central South UniversityChangsha 410011, Hunan, PR China
| | - Alan Dardik
- Department of Surgery and The Interdepartmental Program in Vascular Biology & Therapeutics, Yale University School of MedicineNew Haven, Connecticut, United States of America
| | - Ren Guo
- Department of Pharmacy, The Third Xiangya Hospital, Central South UniversityChangsha 410013, Hunan, P R China
| | - Weichang Zhang
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South UniversityChangsha 410011, Hunan, PR China
- Vascular Disease Institute, Central South UniversityChangsha 410011, Hunan, PR China
| | - Yuanyuan Xiang
- Department of Pharmacy, The Third Xiangya Hospital, Central South UniversityChangsha 410013, Hunan, P R China
- Department of Scientific Research, Hospital of Somatological, Sun Yat-sen UniversityGuangzhou 510055, Guangdong, PR China
| | - Shuang Li
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South UniversityChangsha 410011, Hunan, PR China
- Vascular Disease Institute, Central South UniversityChangsha 410011, Hunan, PR China
| | - Dingxiao Liu
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South UniversityChangsha 410011, Hunan, PR China
- Vascular Disease Institute, Central South UniversityChangsha 410011, Hunan, PR China
| | - Chang Shu
- Department of Vascular Surgery, The Second Xiangya Hospital, Central South UniversityChangsha 410011, Hunan, PR China
- Vascular Disease Institute, Central South UniversityChangsha 410011, Hunan, PR China
- State Key Laboratory of Cardiovascular Disease, Center of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing 10037, PR China
| |
Collapse
|
18
|
Saha N, Robev D, Mason EO, Himanen JP, Nikolov DB. Therapeutic potential of targeting the Eph/ephrin signaling complex. Int J Biochem Cell Biol 2018; 105:123-133. [PMID: 30343150 DOI: 10.1016/j.biocel.2018.10.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/09/2018] [Accepted: 10/16/2018] [Indexed: 12/27/2022]
Abstract
The Eph-ephrin signaling pathway mediates developmental processes and the proper functioning of the adult human body. This distinctive bidirectional signaling pathway includes a canonical downstream signal cascade inside the Eph-bearing cells, as well as a reverse signaling in the ephrin-bearing cells. The signaling is terminated by ADAM metalloproteinase cleavage, internalization, and degradation of the Eph/ephrin complexes. Consequently, the Eph-ephrin-ADAM signaling cascade has emerged as a key target with immense therapeutic potential particularly in the context of cancer. An interesting twist was brought forth by the emergence of ephrins as the entry receptors for the pathological Henipaviruses, which has spurred new studies to target the viral entry. The availability of high-resolution structures of the multi-modular Eph receptors in complexes with ephrins and other binding partners, such as peptides, small molecule inhibitors and antibodies, offers a wealth of information for the structure-guided development of therapeutic intervention. Furthermore, genomic data mining of Eph mutants involved in cancer provides information for targeted drug development. In this review we summarize the distinct avenues for targeting the Eph-ephrin signaling pathway, including its termination by ADAM proteinases. We highlight the latest developments in Eph-related pharmacology in the context of Eph-ephrin-ADAM-based antibodies and small molecules. Finally, the future prospects of genomics- and proteomics-based medicine are discussed.
Collapse
Affiliation(s)
- Nayanendu Saha
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States
| | - Dorothea Robev
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States
| | - Emilia O Mason
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States
| | - Juha P Himanen
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States.
| | - Dimitar B Nikolov
- Sloan-Kettering Institute for Cancer Research, Structural Biology Program, 1275 York Avenue, New York, NY 10065, United States
| |
Collapse
|
19
|
Scilabra SD, Pigoni M, Pravatá V, Schätzl T, Müller SA, Troeberg L, Lichtenthaler SF. Increased TIMP-3 expression alters the cellular secretome through dual inhibition of the metalloprotease ADAM10 and ligand-binding of the LRP-1 receptor. Sci Rep 2018; 8:14697. [PMID: 30279425 PMCID: PMC6168507 DOI: 10.1038/s41598-018-32910-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 08/06/2018] [Indexed: 01/21/2023] Open
Abstract
The tissue inhibitor of metalloproteinases-3 (TIMP-3) is a major regulator of extracellular matrix turnover and protein shedding by inhibiting different classes of metalloproteinases, including disintegrin metalloproteinases (ADAMs). Tissue bioavailability of TIMP-3 is regulated by the endocytic receptor low-density-lipoprotein receptor-related protein-1 (LRP-1). TIMP-3 plays protective roles in disease. Thus, different approaches have been developed aiming to increase TIMP-3 bioavailability, yet overall effects of increased TIMP-3 in vivo have not been investigated. Herein, by using unbiased mass-spectrometry we demonstrate that TIMP-3-overexpression in HEK293 cells has a dual effect on shedding of transmembrane proteins and turnover of soluble proteins. Several membrane proteins showing reduced shedding are known as ADAM10 substrates, suggesting that exogenous TIMP-3 preferentially inhibits ADAM10 in HEK293 cells. Additionally identified shed membrane proteins may be novel ADAM10 substrate candidates. TIMP-3-overexpression also increased extracellular levels of several soluble proteins, including TIMP-1, MIF and SPARC. Levels of these proteins similarly increased upon LRP-1 inactivation, suggesting that TIMP-3 increases soluble protein levels by competing for their binding to LRP-1 and their subsequent internalization. In conclusion, our study reveals that increased levels of TIMP-3 induce substantial modifications in the cellular secretome and that TIMP-3-based therapies may potentially provoke undesired, dysregulated functions of ADAM10 and LRP-1.
Collapse
Affiliation(s)
- Simone D Scilabra
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Strasse 17, 81377, Munich, Germany. .,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, 81675, Munich, Germany.
| | - Martina Pigoni
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Strasse 17, 81377, Munich, Germany
| | - Veronica Pravatá
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Strasse 17, 81377, Munich, Germany
| | - Tobias Schätzl
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Strasse 17, 81377, Munich, Germany
| | - Stephan A Müller
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Strasse 17, 81377, Munich, Germany
| | - Linda Troeberg
- Kennedy Institute of Rheumatology, University of Oxford, Roosevelt Drive, Oxford, OX3 7FY, UK
| | - Stefan F Lichtenthaler
- German Center for Neurodegenerative Diseases (DZNE), Feodor-Lynen Strasse 17, 81377, Munich, Germany.,Neuroproteomics, School of Medicine, Klinikum rechts der Isar, Technische Universität München, 81675, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.,Institute for Advanced Study, Technische Universität München, Munich, Germany
| |
Collapse
|
20
|
TBC1d24-ephrinB2 interaction regulates contact inhibition of locomotion in neural crest cell migration. Nat Commun 2018; 9:3491. [PMID: 30154457 PMCID: PMC6113226 DOI: 10.1038/s41467-018-05924-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 08/02/2018] [Indexed: 11/08/2022] Open
Abstract
Although Eph-ephrin signalling has been implicated in the migration of cranial neural crest (CNC) cells, it is still unclear how ephrinB transduces signals regulating this event. We provide evidence that TBC1d24, a putative Rab35-GTPase activating protein (Rab35 GAP), complexes with ephrinB2 via the scaffold Dishevelled (Dsh) and mediates a signal affecting contact inhibition of locomotion (CIL) in CNC cells. Moreover, we found that, in migrating CNC, the interaction between ephrinB2 and TBC1d24 negatively regulates E-cadherin recycling in these cells via Rab35. Upon engagement of the cognate Eph receptor, ephrinB2 is tyrosine phosphorylated, which disrupts the ephrinB2/Dsh/TBC1d24 complex. The dissolution of this complex leads to increasing E-cadherin levels at the plasma membrane, resulting in loss of CIL and disrupted CNC migration. Our results indicate that TBC1d24 is a critical player in ephrinB2 control of CNC cell migration via CIL.
Collapse
|
21
|
Tae N, Lee S, Kim O, Park J, Na S, Lee JH. Syntenin promotes VEGF-induced VEGFR2 endocytosis and angiogenesis by increasing ephrin-B2 function in endothelial cells. Oncotarget 2018; 8:38886-38901. [PMID: 28418925 PMCID: PMC5503580 DOI: 10.18632/oncotarget.16452] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 02/27/2017] [Indexed: 12/21/2022] Open
Abstract
Syntenin, a tandem PDZ-domain-containing scaffold protein, is involved in the regulation of diverse biological functions, including protein trafficking, exosome biogenesis, and cancer metastasis. Here, we present the first study to explore the significance of syntenin in endothelial cells. Syntenin knockdown in human umbilical vein endothelial cells (HUVECs) impaired vascular endothelial growth factor (VEGF)-mediated proliferation, migration, invasion, vascular permeability, and nitric oxide (NO) production. Syntenin knockdown also suppressed expression of the VEGFR2 target genes VEGF, MMP2, and Nurr77 as well as VEGF-induced angiogenesis in vitro and in vivo. And it decreased cell-surface levels of ephrin-B2. Biochemical analyses revealed that syntenin exists in complex with VEGFR2 and ephrin-B2. Syntenin knockdown abolished the association between VEGFR2 and ephrin-B2, suggesting syntenin functions as a scaffold protein facilitating their association in HUVECs. Consistent with these observations, knocking down syntenin or ephrin-B2 abolished VEGF-induced endocytosis and VEGFR2 phosphorylation and activation of its downstream signaling molecules. Treatment with MG132, a proteasome inhibitor, rescued the downregulation of ephrin-B2 and VEGFR2 signaling induced by syntenin knockdown. These findings demonstrate that syntenin promotes VEGF signaling and, through its PDZ-dependent interaction with ephrin-B2, enhances VEGF-mediated VEGFR2 endocytosis and subsequent downstream signaling and angiogenesis in endothelial cells.
Collapse
Affiliation(s)
- Nara Tae
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do 243-41, Republic of Korea
| | - Suhyun Lee
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do 243-41, Republic of Korea
| | - Okwha Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do 243-41, Republic of Korea
| | - Juhee Park
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do 243-41, Republic of Korea
| | - Sunghun Na
- Department of Obstetrics and Gynecology, Kangwon National University Hospital, school of Medicine, Kangwon National University, Chuncheon, Gangwon-Do 243-41, Republic of Korea
| | - Jeong-Hyung Lee
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-Do 243-41, Republic of Korea
| |
Collapse
|
22
|
ADAM10-mediated ephrin-B2 shedding promotes myofibroblast activation and organ fibrosis. Nat Med 2017; 23:1405-1415. [PMID: 29058717 PMCID: PMC5720906 DOI: 10.1038/nm.4419] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 09/11/2017] [Indexed: 12/13/2022]
Abstract
Maladaptive wound healing responses to chronic tissue injury result in organ fibrosis. Fibrosis, which entails excessive extracellular matrix (ECM) deposition and tissue remodeling by activated myofibroblasts, leads to loss of proper tissue architecture and organ function; however, the molecular mediators of myofibroblast activation have yet to be fully identified. Here we identify soluble ephrin-B2 (sEphrin-B2) as a new profibrotic mediator in lung and skin fibrosis. We provide molecular, functional and translational evidence that the ectodomain of membrane-bound ephrin-B2 is shed from fibroblasts into the alveolar airspace after lung injury. Shedding of sEphrin-B2 promotes fibroblast chemotaxis and activation via EphB3 and/or EphB4 receptor signaling. We found that mice lacking ephrin-B2 in fibroblasts are protected from skin and lung fibrosis and that a disintegrin and metalloproteinase 10 (ADAM10) is the major ephrin-B2 sheddase in fibroblasts. ADAM10 expression is increased by transforming growth factor (TGF)-β1, and ADAM10-mediated sEphrin-B2 generation is required for TGF-β1-induced myofibroblast activation. Pharmacological inhibition of ADAM10 reduces sEphrin-B2 levels in bronchoalveolar lavage and prevents lung fibrosis in mice. Consistent with the mouse data, ADAM10-sEphrin-B2 signaling is upregulated in fibroblasts from human subjects with idiopathic pulmonary fibrosis. These results uncover a new molecular mechanism of tissue fibrogenesis and identify sEphrin-B2, its receptors EphB3 and EphB4 and ADAM10 as potential therapeutic targets in the treatment of fibrotic diseases.
Collapse
|
23
|
Hwang YS, Daar IO. A frog's view of EphrinB signaling. Genesis 2017; 55. [PMID: 28095646 DOI: 10.1002/dvg.23002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 11/17/2016] [Accepted: 11/17/2016] [Indexed: 12/20/2022]
Abstract
Cell-cell and cell-substrate adhesion are essential to the proper formation and maintenance of tissue patterns during development, and deregulation of these processes can lead to invasion and metastasis of cancer cells. Cell surface adhesion and signaling molecules are key players in both normal development and cancer progression. One set of cell surface proteins, the Eph receptor tyrosine kinases and their membrane-bound ligands, ephrins, are significant regulators of these processes. During embryonic development, the Eph/ephrin signaling system is involved in cell-cell contact events that result in cell sorting and boundary formation between receptor and ligand bearing cells. When migrating cells that display the membrane bound ligands or receptors come in contact with cells bearing the cognate partner, the response may be adhesion or repulsion, ultimately leading to the proper positioning of these cells. During cancer progression, the signaling between these receptor/ligand pairs is often deregulated, leading to increased invasion and metastasis. To gain mechanistic insight into the pathways that mediate Eph receptor and ephrin signaling we have relied upon a very tractable system, the frog Xenopus. This model system has proven to be extremely versatile, and represents a relatively quick and manipulable system to explore signaling events and the in vivo processes affected by these signals.
Collapse
Affiliation(s)
- Yoo-Seok Hwang
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, Maryland, 21702
| | - Ira O Daar
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, Maryland, 21702
| |
Collapse
|
24
|
Morris EAR, Bodin S, Delaval B, Comunale F, Georget V, Costa ML, Lutfalla G, Gauthier-Rouvière C. Flotillins control zebrafish epiboly through their role in cadherin-mediated cell-cell adhesion. Biol Cell 2017; 109:210-221. [DOI: 10.1111/boc.201700001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 02/14/2017] [Accepted: 02/15/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Eduardo A. Rios Morris
- CRBM, CNRS, UMR 5237, Université de Montpellier; Equipe Labellisée Ligue contre le Cancer; Montpellier 34293 France
- MRI-CRBM Imaging facility, CNRS, UMR 5237; Université de Montpellier; Montpellier 34293 France
- Laboratório de Diferenciação Muscular e Citoesqueleto; Departamento de Histologia e Embriologia, Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ 21949-590 Brazil
| | - Stéphane Bodin
- CRBM, CNRS, UMR 5237, Université de Montpellier; Equipe Labellisée Ligue contre le Cancer; Montpellier 34293 France
| | - Bénédicte Delaval
- CRBM, CNRS, UMR 5237, Université de Montpellier; Equipe Labellisée Ligue contre le Cancer; Montpellier 34293 France
| | - Franck Comunale
- CRBM, CNRS, UMR 5237, Université de Montpellier; Equipe Labellisée Ligue contre le Cancer; Montpellier 34293 France
| | - Virginie Georget
- CRBM, CNRS, UMR 5237, Université de Montpellier; Equipe Labellisée Ligue contre le Cancer; Montpellier 34293 France
- MRI-CRBM Imaging facility, CNRS, UMR 5237; Université de Montpellier; Montpellier 34293 France
| | - Manoel L. Costa
- Laboratório de Diferenciação Muscular e Citoesqueleto; Departamento de Histologia e Embriologia, Instituto de Ciências Biomédicas; Universidade Federal do Rio de Janeiro; Rio de Janeiro RJ 21949-590 Brazil
| | - Georges Lutfalla
- Laboratoire de Dynamique des Interactions Membranaires Normales et Pathologiques; Université de Montpellier II et I; CNRS, UMR 5235 Montpellier 34095 France
| | - Cécile Gauthier-Rouvière
- CRBM, CNRS, UMR 5237, Université de Montpellier; Equipe Labellisée Ligue contre le Cancer; Montpellier 34293 France
| |
Collapse
|
25
|
Mechanisms of ephrin-Eph signalling in development, physiology and disease. Nat Rev Mol Cell Biol 2016; 17:240-56. [PMID: 26790531 DOI: 10.1038/nrm.2015.16] [Citation(s) in RCA: 461] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Eph receptor Tyr kinases and their membrane-tethered ligands, the ephrins, elicit short-distance cell-cell signalling and thus regulate many developmental processes at the interface between pattern formation and morphogenesis, including cell sorting and positioning, and the formation of segmented structures and ordered neural maps. Their roles extend into adulthood, when ephrin-Eph signalling regulates neuronal plasticity, homeostatic events and disease processes. Recently, new insights have been gained into the mechanisms of ephrin-Eph signalling in different cell types, and into the physiological importance of ephrin-Eph in different organs and in disease, raising questions for future research directions.
Collapse
|
26
|
The alpha secretase ADAM10: A metalloprotease with multiple functions in the brain. Prog Neurobiol 2015; 135:1-20. [PMID: 26522965 DOI: 10.1016/j.pneurobio.2015.10.003] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/23/2015] [Accepted: 10/26/2015] [Indexed: 01/07/2023]
Abstract
Proteins belonging to the 'A Disintegrin And Metalloproteinase' (ADAM) family are membrane-anchored proteases that are able to cleave the extracellular domains of several membrane-bound proteins in a process known as 'ectodomain shedding'. In the central nervous system, ADAM10 has attracted the most attention, since it was described as the amyloid precursor protein α-secretase over ten years ago. Despite the excitement over the potential of ADAM10 as a novel drug target in Alzheimer disease, the physiological functions of ADAM10 in the brain are not yet well understood. This is largely because of the embryonic lethality of ADAM10-deficient mice, which results from the loss of cleavage and signaling of the Notch receptor, another ADAM10 substrate. However, the recent generation of conditional ADAM10-deficient mice and the identification of further ADAM10 substrates in the brain has revealed surprisingly numerous and fundamental functions of ADAM10 in the development of the embryonic brain and also in the homeostasis of adult neuronal networks. Mechanistically, ADAM10 controls these functions by utilizing unique postsynaptic substrates in the central nervous system, in particular synaptic cell adhesion molecules, such as neuroligin-1, N-cadherin, NCAM, Ephrin A2 and A5. Consequently, a dysregulation of ADAM10 activity is linked to psychiatric and neurological diseases, such as epilepsy, fragile X syndrome and Huntington disease. This review highlights the recent progress in understanding the substrates and function as well as the regulation and cell biology of ADAM10 in the central nervous system and discusses the value of ADAM10 as a drug target in brain diseases.
Collapse
|
27
|
Abdul-Wajid S, Morales-Diaz H, Khairallah SM, Smith WC. T-type Calcium Channel Regulation of Neural Tube Closure and EphrinA/EPHA Expression. Cell Rep 2015; 13:829-839. [PMID: 26489462 DOI: 10.1016/j.celrep.2015.09.035] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 08/06/2015] [Accepted: 09/11/2015] [Indexed: 10/22/2022] Open
Abstract
A major class of human birth defects arise from aberrations during neural tube closure (NTC). We report on a NTC signaling pathway requiring T-type calcium channels (TTCCs) that is conserved between primitive chordates (Ciona) and Xenopus. With loss of TTCCs, there is a failure to seal the anterior neural folds. Accompanying loss of TTCCs is an upregulation of EphrinA effectors. Ephrin signaling is known to be important in NTC, and ephrins can affect both cell adhesion and repulsion. In Ciona, ephrinA-d expression is downregulated at the end of neurulation, whereas, with loss of TTCC, ephrinA-d remains elevated. Accordingly, overexpression of ephrinA-d phenocopied TTCC loss of function, while overexpression of a dominant-negative Ephrin receptor was able to rescue NTC in a Ciona TTCC mutant. We hypothesize that signaling through TTCCs is necessary for proper anterior NTC through downregulation of ephrins, and possibly elimination of a repulsive signal.
Collapse
Affiliation(s)
- Sarah Abdul-Wajid
- Department of Molecular, Cell and Developmental Biology, and Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Heidi Morales-Diaz
- Department of Molecular, Cell and Developmental Biology, and Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Stephanie M Khairallah
- Department of Molecular, Cell and Developmental Biology, and Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - William C Smith
- Department of Molecular, Cell and Developmental Biology, and Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.
| |
Collapse
|
28
|
Abstract
Proteases regulate a myriad of cell functions, both in normal and disease states. In addition to protein turnover, they regulate a range of signaling processes, including those mediated by Eph receptors and their ephrin ligands. A variety of proteases is reported to directly cleave Ephs and/or ephrins under different conditions, to promote receptor and/or ligand shedding, and regulate receptor/ligand internalisation and signaling. They also cleave other adhesion proteins in response to Eph-ephrin interactions, to indirectly facilitate Eph-mediated functions. Proteases thus contribute to Eph/ephrin mediated changes in cell-cell and cell-matrix interactions, in cell morphology and in cell migration and invasion, in a manner which appears to be tightly regulated by, and co-ordinated with, Eph signaling. This review summarizes the current literature describing the function and regulation of protease activities during Eph/ephrin-mediated cell signaling.
Collapse
Affiliation(s)
- Lakmali Atapattu
- a Department of Biochemistry and Molecular Biology ; Monash University , Victoria ; Australia
| | | | | |
Collapse
|
29
|
Lisle JE, Mertens-Walker I, Stephens CR, Stansfield SH, Clements JA, Herington AC, Stephenson SA. Murine, but not human, ephrin-B2 can be efficiently cleaved by the serine protease kallikrein-4: implications for xenograft models of human prostate cancer. Exp Cell Res 2015; 333:136-46. [PMID: 25724897 DOI: 10.1016/j.yexcr.2015.02.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 12/24/2014] [Accepted: 02/14/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND Ephrin-B2 is the sole physiologically-relevant ligand of the receptor tyrosine kinase EphB4, which is over-expressed in many epithelial cancers, including 66% of prostate cancers, and contributes to cancer cell survival, invasion and migration. Crucially, however, the cancer-promoting EphB4 signalling pathways are independent of interaction with its ligand ephrin-B2, as activation of ligand-dependent signalling causes tumour suppression. Ephrin-B2, however, is often found on the surface of endothelial cells of the tumour vasculature, where it can regulate angiogenesis to support tumour growth. Proteolytic cleavage of endothelial cell ephrin-B2 has previously been suggested as one mechanism whereby the interaction between tumour cell-expressed EphB4 and endothelial cell ephrin-B2 is regulated to support both cancer promotion and angiogenesis. METHODS An in silico approach was used to search accessible surfaces of 3D protein models for cleavage sites for the key prostate cancer serine protease, KLK4, and this identified murine ephrin-B2 as a potential KLK4 substrate. Mouse ephrin-B2 was then confirmed as a KLK4 substrate by in vitro incubation of recombinant mouse ephrin-B2 with active recombinant human KLK4. Cleavage products were visualised by SDS-PAGE, silver staining and Western blot and confirmed by N-terminal sequencing. RESULTS At low molar ratios, KLK4 cleaved murine ephrin-B2 but other prostate-specific KLK family members (KLK2 and KLK3/PSA) were less efficient, suggesting cleavage was KLK4-selective. The primary KLK4 cleavage site in murine ephrin-B2 was verified and shown to correspond to one of the in silico predicted sites between extracellular domain residues arginine 178 and asparagine 179. Surprisingly, the highly homologous human ephrin-B2 was poorly cleaved by KLK4 at these low molar ratios, likely due to the 3 amino acid differences at this primary cleavage site. CONCLUSION These data suggest that in in vivo mouse xenograft models, endogenous mouse ephrin-B2, but not human tumour ephrin-B2, may be a downstream target of cancer cell secreted human KLK4. This is a critical consideration when interpreting data from murine explants of human EphB4+/KLK4+ cancer cells, such as prostate cancer cells, where differential effects may be seen in mouse models as opposed to human clinical situations.
Collapse
Affiliation(s)
- J E Lisle
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia
| | - I Mertens-Walker
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia
| | - C R Stephens
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia
| | - S H Stansfield
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia
| | - J A Clements
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia
| | - A C Herington
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia
| | - S-A Stephenson
- Institute of Health and Biomedical Innovation and the Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Translational Research Institute, Woolloongabba 4102, QLD, Australia.
| |
Collapse
|
30
|
Park I, Lee HS. EphB/ephrinB signaling in cell adhesion and migration. Mol Cells 2015; 38:14-9. [PMID: 25475547 PMCID: PMC4314128 DOI: 10.14348/molcells.2015.2116] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 10/14/2014] [Accepted: 10/17/2014] [Indexed: 11/27/2022] Open
Abstract
Eph receptors and their ligands, ephrins, represent the largest group of the receptor tyrosine kinase (RTK) family, and they mediate numerous developmental processes in a variety of organisms. Ephrins are membrane-bound proteins that are mainly divided into two classes: A class ephrins, which are linked to the membrane by a glycosylphosphatidylinositol (GPI) linkage, and B class ephrins, which are transmembrane ligands. Based on their domain structures and affinities for ligand binding, the Eph receptors are also divided into two groups. Trans-dimerization of Eph receptors with their membrane-tethered ligands regulates cell-cell interactions and initiates bidirectional signaling pathways. These pathways are intimately involved in regulating cytoskeleton dynamics, cell migration, and alterations in cellular dynamics and shapes. The EphBs and ephrinBs are specifically localized and modified to promote higher-order clustering and initiate of bidirectional signaling. In this review, we present an in-depth overview of the structure, mechanisms, cell signaling, and functions of EphB/ephrinB in cell adhesion and migration.
Collapse
Affiliation(s)
- Inji Park
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701,
Korea
| | - Hyun-Shik Lee
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701,
Korea
| |
Collapse
|
31
|
Abstract
The erythropoietin-producing hepatocellular carcinoma (Eph) receptor tyrosine kinase family plays important roles in developmental processes, adult tissue homeostasis, and various diseases. Interaction with Eph receptor-interacting protein (ephrin) ligands on the surface of neighboring cells triggers Eph receptor kinase-dependent signaling. The ephrins can also transmit signals, leading to bidirectional cell contact-dependent communication. Moreover, Eph receptors and ephrins can function independently of each other through interplay with other signaling systems. Given their involvement in many pathological conditions ranging from neurological disorders to cancer and viral infections, Eph receptors and ephrins are increasingly recognized as attractive therapeutic targets, and various strategies are being explored to modulate their expression and function. Eph receptor/ephrin upregulation in cancer cells, the angiogenic vasculature, and injured or diseased tissues also offer opportunities for Eph/ephrin-based targeted drug delivery and imaging. Thus, despite the challenges presented by the complex biology of the Eph receptor/ephrin system, exciting possibilities exist for therapies exploiting these molecules.
Collapse
Affiliation(s)
- Antonio Barquilla
- Cancer Center, Sanford-Burnham Medical Research Institute, La Jolla, California 92037; ,
| | | |
Collapse
|