1
|
Haap‐Hoff A, Freeley M, Dempsey E, Dunican D, Bennett E, Triglia D, Skubis‐Zegadlo J, Mitchell Davies A, Kelleher D, Long A. RNAi library screening reveals Gβ1, Casein Kinase 2 and ICAP-1 as novel regulators of LFA-1-mediated T cell polarity and migration. Immunol Cell Biol 2025; 103:73-92. [PMID: 39607284 PMCID: PMC11688611 DOI: 10.1111/imcb.12838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/28/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024]
Abstract
The αLβ2 integrin LFA-1 plays a key role in T-cell adhesion to the endothelial vasculature and migration into both secondary lymphoid organs and peripheral tissues via interactions with its target protein ICAM-1, but the pathways that regulate LFA-1-mediated T-cell polarity and migration are not fully understood. In this study we screened two RNAi libraries targeting G protein-coupled receptors (GPCR)/GPCR-associated proteins and kinases in a HuT 78 T cell line model of LFA-1-stimulated T-cell migration. Based on staining of the actin cytoskeleton, multiple parameters to measure cell morphology were used to assess the contribution of 1109 genes to LFA-1-mediated T-cell polarity and migration. These RNAi screens identified a number of both novel and previously identified genes that either increased or decreased the polarity and migratory capacity of these cells. Following multiparametric analysis, hierarchical clustering and pathway analysis, three of these genes were characterized in further detail using primary human T cells, revealing novel roles for the heterotrimeric G protein subunit Gβ1 and Casein Kinase 2 in LFA-1-mediated T-cell polarity and migration in vitro. Our studies also highlighted a new role for ICAP-1, an adaptor protein previously described to be associated with β1 integrins, in β2 integrin LFA-1-directed migration in T cells. Knockdown of ICAP-1 expression in primary T cells revealed a role in cell polarity, cell velocity and transmigration towards SDF-1 for this adaptor protein. This study therefore uncovers new roles for GPCR/GPCR-associated proteins and kinases in T-cell migration and provides potential novel targets for modulation of the T-cell immune response.
Collapse
Affiliation(s)
- Antje Haap‐Hoff
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Michael Freeley
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
- Present address:
School of BiotechnologyDublin City UniversityDublinIreland
| | - Eugene Dempsey
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Dara Dunican
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Emily Bennett
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Denise Triglia
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Joanna Skubis‐Zegadlo
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Anthony Mitchell Davies
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| | - Dermot Kelleher
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
- Present address:
Faculty of MedicineUniversity of British ColumbiaVancouverBCCanada
| | - Aideen Long
- Trinity Translational Medicine Institute & Department of Clinical Medicine, Trinity College DublinSt James's HospitalDublinRepublic of Ireland
| |
Collapse
|
2
|
Glading A. KRIT1 in vascular biology and beyond. Biosci Rep 2024; 44:BSR20231675. [PMID: 38980708 PMCID: PMC11263069 DOI: 10.1042/bsr20231675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 06/21/2024] [Accepted: 07/09/2024] [Indexed: 07/10/2024] Open
Abstract
KRIT1 is a 75 kDa scaffolding protein which regulates endothelial cell phenotype by limiting the response to inflammatory stimuli and maintaining a quiescent and stable endothelial barrier. Loss-of-function mutations in KRIT1 lead to the development of cerebral cavernous malformations (CCM), a disease marked by the formation of abnormal blood vessels which exhibit a loss of barrier function, increased endothelial proliferation, and altered gene expression. While many advances have been made in our understanding of how KRIT1, and the functionally related proteins CCM2 and PDCD10, contribute to the regulation of blood vessels and the vascular barrier, some important open questions remain. In addition, KRIT1 is widely expressed and KRIT1 and the other CCM proteins have been shown to play important roles in non-endothelial cell types and tissues, which may or may not be related to their role as pathogenic originators of CCM. In this review, we discuss some of the unsettled questions regarding the role of KRIT1 in vascular physiology and discuss recent advances that suggest this ubiquitously expressed protein may have a role beyond the endothelial cell.
Collapse
Affiliation(s)
- Angela J. Glading
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY, U.S.A
| |
Collapse
|
3
|
Sidibé A, Mykuliak VV, Zhang P, Hytönen VP, Wu J, Wehrle-Haller B. Acetyl-NPKY of integrin-β1 binds KINDLIN2 to control endothelial cell proliferation and junctional integrity. iScience 2024; 27:110129. [PMID: 38904068 PMCID: PMC11187247 DOI: 10.1016/j.isci.2024.110129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 02/09/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Integrin-dependent crosstalk between cell-matrix adhesions and cell-cell junctions is critical for controlling endothelial permeability and proliferation in cancer and inflammatory diseases but remains poorly understood. Here, we investigated how acetylation of the distal NPKY-motif of Integrin-β1 influences endothelial cell physiology and barrier function. Expression of an acetylation-mimetic β1-K794Q-GFP mutant led to the accumulation of immature cell-matrix adhesions accompanied by a transcriptomic reprograming of endothelial cells, involving genes associated with cell adhesion, proliferation, polarity, and barrier function. β1-K794Q-GFP induced constitutive MAPK signaling, junctional impairment, proliferation, and reduced contact inhibition at confluence. Structural analysis of Integrin-β1 interaction with KINDLIN2, biochemical pulldown assay, and binding energy determination by using molecular dynamics simulation showed that acetylation of K794 and the K794Q-mutant increased KINDLIN2 binding affinity to the Integrin-β1. Thus, enhanced recruitment of KINDLIN2 to Lysine-acetylated Integrin-β1 and resulting modulation of barrier function, offers new therapeutic possibilities for controlling vascular permeability and disease conditions.
Collapse
Affiliation(s)
- Adama Sidibé
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Vasyl V. Mykuliak
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Pingfeng Zhang
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Vesa P. Hytönen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
- Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland
| | - Jinhua Wu
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| |
Collapse
|
4
|
Maldonado H, Leyton L. CSK-mediated signalling by integrins in cancer. Front Cell Dev Biol 2023; 11:1214787. [PMID: 37519303 PMCID: PMC10382208 DOI: 10.3389/fcell.2023.1214787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
Cancer progression and metastasis are processes heavily controlled by the integrin receptor family. Integrins are cell adhesion molecules that constitute the central components of mechanosensing complexes called focal adhesions, which connect the extracellular environment with the cell interior. Focal adhesions act as key players in cancer progression by regulating biological processes, such as cell migration, invasion, proliferation, and survival. Src family kinases (SFKs) can interplay with integrins and their downstream effectors. SFKs also integrate extracellular cues sensed by integrins and growth factor receptors (GFR), transducing them to coordinate metastasis and cell survival in cancer. The non-receptor tyrosine kinase CSK is a well-known SFK member that suppresses SFK activity by phosphorylating its specific negative regulatory loop (C-terminal Y527 residue). Consequently, CSK may play a pivotal role in tumour progression and suppression by inhibiting SFK oncogenic effects in several cancer types. Remarkably, CSK can localise near focal adhesions when SFKs are activated and even interact with focal adhesion components, such as phosphorylated FAK and Paxillin, among others, suggesting that CSK may regulate focal adhesion dynamics and structure. Even though SFK oncogenic signalling has been extensively described before, the specific role of CSK and its crosstalk with integrins in cancer progression, for example, in mechanosensing, remain veiled. Here, we review how CSK, by regulating SFKs, can regulate integrin signalling, and focus on recent discoveries of mechanotransduction. We additionally examine the cross talk of integrins and GFR as well as the membrane availability of these receptors in cancer. We also explore new pharmaceutical approaches to these signalling pathways and analyse them as future therapeutic targets.
Collapse
Affiliation(s)
- Horacio Maldonado
- Receptor Dynamics in Cancer Laboratory, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
5
|
Ahmad SMS, Nazar H, Rahman MM, Rusyniak RS, Ouhtit A. ITGB1BP1, a Novel Transcriptional Target of CD44-Downstream Signaling Promoting Cancer Cell Invasion. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:373-380. [PMID: 37252376 PMCID: PMC10225144 DOI: 10.2147/bctt.s404565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/25/2023] [Indexed: 05/31/2023]
Abstract
Breast cancer (BC) is the most common malignancy worldwide and has a poor prognosis, because it begins in the breast and disseminates to lymph nodes and distant organs. While invading, BC cells acquire aggressive characteristics from the tumor microenvironment through several mechanisms. Thus, understanding the mechanisms underlying the process of BC cell invasion can pave the way towards the development of targeted therapeutics focused on metastasis. We have previously reported that the activation of CD44 receptor with its major ligand hyaluronan (HA) promotes BC metastasis to the liver in vivo. Next, a gene expression profiling microarray analysis was conducted to identify and validate CD44-downstream transcriptional targets mediating its pro-metastatic function from RNA samples collected from Tet CD44-induced versus control MCF7-B5 cells. We have already validated a number of novel CD44-target genes and published their underlying signaling pathways in promoting BC cell invasion. From the same microarray analysis, Integrin subunit beta 1 binding protein 1 (ITGB1BP1) was also identified as a potential CD44-target gene that was upregulated (2-fold) upon HA activation of CD44. This report will review the lines of evidence collected from the literature to support our hypothesis, and further discuss the possible mechanisms linking HA activation of CD44 to its novel potential transcriptional target ITGB1BP1.
Collapse
Affiliation(s)
- Salma M S Ahmad
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Hanan Nazar
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Md Mizanur Rahman
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Radoslaw Stefan Rusyniak
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| | - Allal Ouhtit
- Biological Sciences Program, Department of Biological and Environmental Sciences, College of Arts and Science, Qatar University, Doha, Qatar
| |
Collapse
|
6
|
Perrelli A, Ferraris C, Berni E, Glading AJ, Retta SF. KRIT1: A Traffic Warden at the Busy Crossroads Between Redox Signaling and the Pathogenesis of Cerebral Cavernous Malformation Disease. Antioxid Redox Signal 2023; 38:496-528. [PMID: 36047808 PMCID: PMC10039281 DOI: 10.1089/ars.2021.0263] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 12/18/2022]
Abstract
Significance: KRIT1 (Krev interaction trapped 1) is a scaffolding protein that plays a critical role in vascular morphogenesis and homeostasis. Its loss-of-function has been unequivocally associated with the pathogenesis of Cerebral Cavernous Malformation (CCM), a major cerebrovascular disease of genetic origin characterized by defective endothelial cell-cell adhesion and ensuing structural alterations and hyperpermeability in brain capillaries. KRIT1 contributes to the maintenance of endothelial barrier function by stabilizing the integrity of adherens junctions and inhibiting the formation of actin stress fibers. Recent Advances: Among the multiple regulatory mechanisms proposed so far, significant evidence accumulated over the past decade has clearly shown that the role of KRIT1 in the stability of endothelial barriers, including the blood-brain barrier, is largely based on its involvement in the complex machinery governing cellular redox homeostasis and responses to oxidative stress and inflammation. KRIT1 loss-of-function has, indeed, been demonstrated to cause an impairment of major redox-sensitive mechanisms involved in spatiotemporal regulation of cell adhesion and signaling, which ultimately leads to decreased cell-cell junction stability and enhanced sensitivity to oxidative stress and inflammation. Critical Issues: This review explores the redox mechanisms that influence endothelial cell adhesion and barrier function, focusing on the role of KRIT1 in such mechanisms. We propose that this supports a novel model wherein redox signaling forms the common link between the various pathogenetic mechanisms and therapeutic approaches hitherto associated with CCM disease. Future Directions: A comprehensive characterization of the role of KRIT1 in redox control of endothelial barrier physiology and defense against oxy-inflammatory insults will provide valuable insights into the development of precision medicine strategies. Antioxid. Redox Signal. 38, 496-528.
Collapse
Affiliation(s)
- Andrea Perrelli
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Chiara Ferraris
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Elisa Berni
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| | - Angela J. Glading
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Saverio Francesco Retta
- Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
- CCM Italia Research Network, National Coordination Center at the Department of Clinical and Biological Sciences, University of Torino, Torino, Italy
| |
Collapse
|
7
|
Wen L, Lyu Q, Ley K, Goult BT. Structural Basis of β2 Integrin Inside—Out Activation. Cells 2022; 11:cells11193039. [PMID: 36231001 PMCID: PMC9564206 DOI: 10.3390/cells11193039] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
β2 integrins are expressed on all leukocytes. Precise regulation of the β2 integrin is critical for leukocyte adhesion and trafficking. In neutrophils, β2 integrins participate in slow rolling. When activated by inside–out signaling, fully activated β2 integrins mediate rapid leukocyte arrest and adhesion. The two activation pathways, starting with selectin ligand engagement and chemokine receptor ligation, respectively, converge on phosphoinositide 3-kinase, talin-1, kindlin-3 and Rap1. Here, we focus on recent structural insights into autoinhibited talin-1 and autoinhibited trimeric kindlin-3. When activated, both talin-1 and kindlin-3 can bind the β2 cytoplasmic tail at separate but adjacent sites. We discuss possible pathways for talin-1 and kindlin-3 activation, recruitment to the plasma membrane, and their role in integrin activation. We propose new models of the final steps of integrin activation involving the complex of talin-1, kindlin-3, integrin and the plasma membrane.
Collapse
Affiliation(s)
- Lai Wen
- Department of Pharmacology, Center for Molecular and Cellular Signaling in the Cardiovascular System, Reno School of Medicine, University of Nevada, Reno, NV 89577, USA
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Qingkang Lyu
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Klaus Ley
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Immunology Center of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Benjamin T. Goult
- School of Biosciences, University of Kent, Canterbury CT2 7NJ, UK
- Correspondence: ; Tel.: +44-(0)1227-816-142
| |
Collapse
|
8
|
Sevilla-Movilla S, Fuentes P, Rodríguez-García Y, Arellano-Sánchez N, Krenn PW, de Val SI, Montero-Herradón S, García-Ceca J, Burdiel-Herencia V, Gardeta SR, Aguilera-Montilla N, Barrio-Alonso C, Crainiciuc G, Bouvard D, García-Pardo A, Zapata AG, Hidalgo A, Fässler R, Carrasco YR, Toribio ML, Teixidó J. ICAP-1 loss impairs CD8 + thymocyte development and leads to reduced marginal zone B cells in mice. Eur J Immunol 2022; 52:1228-1242. [PMID: 35491946 PMCID: PMC9543158 DOI: 10.1002/eji.202149560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 03/15/2022] [Accepted: 04/29/2022] [Indexed: 11/12/2022]
Abstract
ICAP‐1 regulates β1‐integrin activation and cell adhesion. Here, we used ICAP‐1‐null mice to study ICAP‐1 potential involvement during immune cell development and function. Integrin α4β1‐dependent adhesion was comparable between ICAP‐1‐null and control thymocytes, but lack of ICAP‐1 caused a defective single‐positive (SP) CD8+ cell generation, thus, unveiling an ICAP‐1 involvement in SP thymocyte development. ICAP‐1 bears a nuclear localization signal and we found it displayed a strong nuclear distribution in thymocytes. Interestingly, there was a direct correlation between the lack of ICAP‐1 and reduced levels in SP CD8+ thymocytes of Runx3, a transcription factor required for CD8+ thymocyte generation. In the spleen, ICAP‐1 was found evenly distributed between cytoplasm and nuclear fractions, and ICAP‐1–/– spleen T and B cells displayed upregulation of α4β1‐mediated adhesion, indicating that ICAP‐1 negatively controls their attachment. Furthermore, CD3+‐ and CD19+‐selected spleen cells from ICAP‐1‐null mice showed reduced proliferation in response to T‐ and B‐cell stimuli, respectively. Finally, loss of ICAP‐1 caused a remarkable decrease in marginal zone B‐ cell frequencies and a moderate increase in follicular B cells. Together, these data unravel an ICAP‐1 involvement in the generation of SP CD8+ thymocytes and in the control of marginal zone B‐cell numbers.
Collapse
Affiliation(s)
- Silvia Sevilla-Movilla
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Patricia Fuentes
- Development and Function of the Immune System Unit, Centro de Biología Molecular Severo Ochoa, CSIC, Universidad Autónoma de Madrid, Madrid, Spain
| | - Yaiza Rodríguez-García
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Nohemi Arellano-Sánchez
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Peter W Krenn
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany.,Present address: Paris-Lodron Universität Salzburg, Austria
| | - Soledad Isern de Val
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Sara Montero-Herradón
- Department of Cell Biology; Faculty of Biology, Complutense University of Madrid, Madrid, 28040.,Spain and Health Research Institute, Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
| | - Javier García-Ceca
- Department of Cell Biology; Faculty of Biology, Complutense University of Madrid, Madrid, 28040.,Spain and Health Research Institute, Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
| | - Valeria Burdiel-Herencia
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Sofía R Gardeta
- Department on Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, 28049, Spain
| | - Noemí Aguilera-Montilla
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Celia Barrio-Alonso
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain.,Present address: Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Georgiana Crainiciuc
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029, Spain.,Institute for Cardiovascular Prevention, Ludwig-Maximilians University, Munich, 80336, Germany
| | - Daniel Bouvard
- Centre de Recherche en Biologie Cellulaire de Montpellier, Montpellier, France
| | - Angeles García-Pardo
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| | - Agustin G Zapata
- Department of Cell Biology; Faculty of Biology, Complutense University of Madrid, Madrid, 28040.,Spain and Health Research Institute, Hospital 12 de Octubre (imas12), Madrid, 28041, Spain
| | - Andrés Hidalgo
- Area of Developmental and Cell Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029, Spain.,Institute for Cardiovascular Prevention, Ludwig-Maximilians University, Munich, 80336, Germany
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Yolanda R Carrasco
- Department on Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, 28049, Spain
| | - Maria L Toribio
- Development and Function of the Immune System Unit, Centro de Biología Molecular Severo Ochoa, CSIC, Universidad Autónoma de Madrid, Madrid, Spain
| | - Joaquin Teixidó
- Department of Molecular Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Madrid, Spain
| |
Collapse
|
9
|
Muscolino E, Di Stefano AB, Trapani M, Sabatino MA, Giacomazza D, Moschella F, Cordova A, Toia F, Dispenza C. Injectable xyloglucan hydrogels incorporating spheroids of adipose stem cells for bone and cartilage regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112545. [PMID: 34857257 DOI: 10.1016/j.msec.2021.112545] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 10/31/2021] [Accepted: 11/07/2021] [Indexed: 12/12/2022]
Abstract
Cartilage or bone regeneration approaches based on the direct injection of mesenchymal stem cells (MSCs) at the lesion site encounter several challenges, related to uncontrolled cell spreading and differentiation, reduced cell viability and poor engrafting. This work presents a simple and versatile strategy based on the synergic combination of in-situ forming hydrogels and spheroids of adipose stem cells (SASCs) with great potential for minimally invasive regenerative interventions aimed to threat bone and cartilage defects. Aqueous dispersions of partially degalactosylated xyloglucan (dXG) are mixed with SASCs derived from liposuction and either a chondroinductive or an osteoinductive medium. The dispersions rapidly set into hydrogels when temperature is brought to 37 °C. The physico-chemical and mechanical properties of the hydrogels are controlled by polymer concentration. The hydrogels, during 21 day incubation at 37 °C, undergo significant structural rearrangements that support cell proliferation and spreading. In formulations containing 1%w dXG cell viability increases up to 300% for SASCs-derived osteoblasts and up to 1000% for SASCs-derived chondrocytes if compared with control 2D cultures. The successful differentiation into the target cells is supported by the expression of lineage-specific genes. Cell-cell and cell-matrix interactions are also investigated. All formulations resulted injectable, and the incorporated cells are fully viable after injection.
Collapse
Affiliation(s)
- Emanuela Muscolino
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze 6, 90128 Palermo, Italy
| | - Anna Barbara Di Stefano
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Marco Trapani
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Maria Antonietta Sabatino
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze 6, 90128 Palermo, Italy
| | - Daniela Giacomazza
- Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146, Palermo, Italy
| | - Francesco Moschella
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy; Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Adriana Cordova
- BIOPLAST-Laboratory of BIOlogy and Regenerative Medicine-PLASTic Surgery, Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy; Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Francesca Toia
- Dipartimento di Discipline Chirurgiche, Oncologiche e Stomatologiche, Università degli Studi di Palermo, via del Vespro 129, 90127 Palermo, Italy
| | - Clelia Dispenza
- Dipartimento di Ingegneria, Università degli Studi di Palermo, Viale delle Scienze 6, 90128 Palermo, Italy; Istituto di Biofisica, Consiglio Nazionale delle Ricerche, Via U. La Malfa 153, 90146, Palermo, Italy.
| |
Collapse
|
10
|
Abstract
Cerebral cavernous malformations (CCMs) are neurovascular abnormalities characterized by thin, leaky blood vessels resulting in lesions that predispose to haemorrhages, stroke, epilepsy and focal neurological deficits. CCMs arise due to loss-of-function mutations in genes encoding one of three CCM complex proteins, KRIT1, CCM2 or CCM3. These widely expressed, multi-functional adaptor proteins can assemble into a CCM protein complex and (either alone or in complex) modulate signalling pathways that influence cell adhesion, cell contractility, cytoskeletal reorganization and gene expression. Recent advances, including analysis of the structures and interactions of CCM proteins, have allowed substantial progress towards understanding the molecular bases for CCM protein function and how their disruption leads to disease. Here, we review current knowledge of CCM protein signalling with a focus on three pathways which have generated the most interest—the RhoA–ROCK, MEKK3–MEK5–ERK5–KLF2/4 and cell junctional signalling pathways—but also consider ICAP1-β1 integrin and cdc42 signalling. We discuss emerging links between these pathways and the processes that drive disease pathology and highlight important open questions—key among them is the role of subcellular localization in the control of CCM protein activity.
Collapse
Affiliation(s)
- Valerie L Su
- Department of Pharmacology, Yale University School of Medicine, PO Box 208066, 333 Cedar Street, New Haven, CT 06520, USA
| | - David A Calderwood
- Department of Pharmacology, Yale University School of Medicine, PO Box 208066, 333 Cedar Street, New Haven, CT 06520, USA.,Department of Cell Biology, Yale University School of Medicine, PO Box 208066, 333 Cedar Street, New Haven, CT 06520, USA
| |
Collapse
|
11
|
Su VL, Simon B, Draheim KM, Calderwood DA. Serine phosphorylation of the small phosphoprotein ICAP1 inhibits its nuclear accumulation. J Biol Chem 2020; 295:3269-3284. [PMID: 32005669 DOI: 10.1074/jbc.ra119.009794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 01/29/2020] [Indexed: 02/06/2023] Open
Abstract
Nuclear accumulation of the small phosphoprotein integrin cytoplasmic domain-associated protein-1 (ICAP1) results in recruitment of its binding partner, Krev/Rap1 interaction trapped-1 (KRIT1), to the nucleus. KRIT1 loss is the most common cause of cerebral cavernous malformation, a neurovascular dysplasia resulting in dilated, thin-walled vessels that tend to rupture, increasing the risk for hemorrhagic stroke. KRIT1's nuclear roles are unknown, but it is known to function as a scaffolding or adaptor protein at cell-cell junctions and in the cytosol, supporting normal blood vessel integrity and development. As ICAP1 controls KRIT1 subcellular localization, presumably influencing KRIT1 function, in this work, we investigated the signals that regulate ICAP1 and, hence, KRIT1 nuclear localization. ICAP1 contains a nuclear localization signal within an unstructured, N-terminal region that is rich in serine and threonine residues, several of which are reportedly phosphorylated. Using quantitative microscopy, we revealed that phosphorylation-mimicking substitutions at Ser-10, or to a lesser extent at Ser-25, within this N-terminal region inhibit ICAP1 nuclear accumulation. Conversely, phosphorylation-blocking substitutions at these sites enhanced ICAP1 nuclear accumulation. We further demonstrate that p21-activated kinase 4 (PAK4) can phosphorylate ICAP1 at Ser-10 both in vitro and in cultured cells and that active PAK4 inhibits ICAP1 nuclear accumulation in a Ser-10-dependent manner. Finally, we show that ICAP1 phosphorylation controls nuclear localization of the ICAP1-KRIT1 complex. We conclude that serine phosphorylation within the ICAP1 N-terminal region can prevent nuclear ICAP1 accumulation, providing a mechanism that regulates KRIT1 localization and signaling, potentially influencing vascular development.
Collapse
Affiliation(s)
- Valerie L Su
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Bertrand Simon
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Kyle M Draheim
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520
| | - David A Calderwood
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut 06520; Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520.
| |
Collapse
|
12
|
Serebriiskii IG, Elmekawy M, Golemis EA. Identification of the KRIT1 Protein by LexA-Based Yeast Two-Hybrid System. Methods Mol Biol 2020; 2152:269-289. [PMID: 32524559 DOI: 10.1007/978-1-0716-0640-7_20] [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] [Indexed: 06/11/2023]
Abstract
Cerebral cavernous malformation (CCM) is a vascular malformation of the central nervous system that is associated with leaky capillaries, and a predisposition to serious clinical conditions including intracerebral hemorrhage and seizures. Germline or sporadic mutations in the CCM1/KRIT1 gene are responsible for the majority of cases of CCM. In this article, we describe the original characterization of the CCM1/KRIT1 gene. This cloning was done through the use of a variant of the yeast two-hybrid screen known as the interaction trap, using the RAS-family GTPase KREV1/RAP1A as a bait. The partial clone of KRIT1 (Krev1 Interaction Trapped) initially identified was extended through 5'RACE and computational analysis to obtain a full-length cDNA, then used in a sequential screen to define the integrin-associated ICAP1 protein as a KRIT1 partner protein. We discuss how these interactions are relevant to the current understanding of KRIT1/CCM1 biology, and provide a protocol for library screening with the Interaction Trap.
Collapse
Affiliation(s)
- Ilya G Serebriiskii
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia.
| | - Mohamed Elmekawy
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
- Moscow Institute of Physics and Technology, Moscow Region, Russia
| | - Erica A Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
13
|
Regulation of cell adhesion: a collaborative effort of integrins, their ligands, cytoplasmic actors, and phosphorylation. Q Rev Biophys 2019; 52:e10. [PMID: 31709962 DOI: 10.1017/s0033583519000088] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Integrins are large heterodimeric type 1 membrane proteins expressed in all nucleated mammalian cells. Eighteen α-chains and eight β-chains can combine to form 24 different integrins. They are cell adhesion proteins, which bind to a large variety of cellular and extracellular ligands. Integrins are required for cell migration, hemostasis, translocation of cells out from the blood stream and further movement into tissues, but also for the immune response and tissue morphogenesis. Importantly, integrins are not usually active as such, but need activation to become adhesive. Integrins are activated by outside-in activation through integrin ligand binding, or by inside-out activation through intracellular signaling. An important question is how integrin activity is regulated, and this topic has recently drawn much attention. Changes in integrin affinity for ligand binding are due to allosteric structural alterations, but equally important are avidity changes due to integrin clustering in the plane of the plasma membrane. Recent studies have partially solved how integrin cell surface structures change during activation. The integrin cytoplasmic domains are relatively short, but by interacting with a variety of cytoplasmic proteins in a regulated manner, the integrins acquire a number of properties important not only for cell adhesion and movement, but also for cellular signaling. Recent work has shown that specific integrin phosphorylations play pivotal roles in the regulation of integrin activity. Our purpose in this review is to integrate the present knowledge to enable an understanding of how cell adhesion is dynamically regulated.
Collapse
|
14
|
Abstract
Integrins are heterodimeric cell surface receptors ensuring the mechanical connection between cells and the extracellular matrix. In addition to the anchorage of cells to the extracellular matrix, these receptors have critical functions in intracellular signaling, but are also taking center stage in many physiological and pathological conditions. In this review, we provide some historical, structural, and physiological notes so that the diverse functions of these receptors can be appreciated and put into the context of the emerging field of mechanobiology. We propose that the exciting journey of the exploration of these receptors will continue for at least another new generation of researchers.
Collapse
Affiliation(s)
- Michael Bachmann
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| | - Sampo Kukkurainen
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| | - Vesa P Hytönen
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire , Geneva , Switzerland ; and Faculty of Medicine and Health Technology, Tampere University, and Fimlab Laboratories , Tampere , Finland
| |
Collapse
|
15
|
Chakraborty S, Banerjee S, Raina M, Haldar S. Force-Directed “Mechanointeractome” of Talin–Integrin. Biochemistry 2019; 58:4677-4695. [DOI: 10.1021/acs.biochem.9b00442] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Soham Chakraborty
- Department of Biological Sciences, Ashoka University, Sonepat, Haryana 131029, India
| | - Souradeep Banerjee
- Department of Biological Sciences, Ashoka University, Sonepat, Haryana 131029, India
| | - Manasven Raina
- Department of Biological Sciences, Ashoka University, Sonepat, Haryana 131029, India
| | - Shubhasis Haldar
- Department of Biological Sciences, Ashoka University, Sonepat, Haryana 131029, India
| |
Collapse
|
16
|
Wang Y, Zhang X, Tian J, Shan J, Hu Y, Zhai Y, Guo J. Talin promotes integrin activation accompanied by generation of tension in talin and an increase in osmotic pressure in neurite outgrowth. FASEB J 2019; 33:6311-6326. [PMID: 30768370 DOI: 10.1096/fj.201801949rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Neuronal polarization depends on the interaction of intracellular chemical and mechanical activities in which the cytoplasmic protein, talin, plays a pivotal role during neurite growth. To better understand the mechanism underlying talin function in neuronal polarization, we overexpressed several truncated forms of talin and found that the presence of the rod domain within the overexpressed talin is required for its positive effect on neurite elongation because the neurite number only increased when the talin head region was overexpressed. The tension in the talin rod was recognized using a Förster resonance energy transfer-based tension probe. Nerve growth factor treatment resulted in inward tension of talin elicited by microfilament force and outward osmotic pressure. By contrast, the glial scar-inhibitor aggrecan weakened these forces, suggesting that interactions between inward pull forces in the talin rod and outward osmotic pressure participate in neuronal polarization. Integrin activation is also involved in up-regulation of talin tension and osmotic pressure. Aggrecan stimuli resulted in up-regulation of docking protein 1 (DOK1), leading to the down-regulation of integrin activity and attenuation of the intracellular mechanical force. Our study suggests interactions between the intracellular inward tension in talin and the outward osmotic pressure as the effective channel for promoting neurite outgrowth, which can be up-regulated by integrin activation and down-regulated by DOK1.-Wang, Y., Zhang, X., Tian, J., Shan, J., Hu, Y., Zhai, Y., Guo, J. Talin promotes integrin activation accompanied by generation of tension in talin and an increase in osmotic pressure in neurite outgrowth.
Collapse
Affiliation(s)
- Yifan Wang
- State Key Laboratory Cultivation Base for Traditional Chinese Medicine (TCM) Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory of Drug Targets and Drugs for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaolong Zhang
- State Key Laboratory Cultivation Base for Traditional Chinese Medicine (TCM) Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory of Drug Targets and Drugs for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jilai Tian
- State Key Laboratory Cultivation Base for Traditional Chinese Medicine (TCM) Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory of Drug Targets and Drugs for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jinjun Shan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yunfeng Hu
- State Key Laboratory Cultivation Base for Traditional Chinese Medicine (TCM) Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory of Drug Targets and Drugs for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yiqian Zhai
- State Key Laboratory Cultivation Base for Traditional Chinese Medicine (TCM) Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory of Drug Targets and Drugs for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jun Guo
- State Key Laboratory Cultivation Base for Traditional Chinese Medicine (TCM) Quality and Efficacy, School of Medicine and Life Science, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory of Drug Targets and Drugs for Degenerative Disease, Nanjing University of Chinese Medicine, Nanjing, China
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
17
|
|
18
|
Armas Melián K, Rodríguez Pons D, Díaz Romero R, Givica Pérez A, Centeno Haro M, Fernandez JJ. Giant cerebral cavernomas malformations. Chirurgia (Bucur) 2019. [DOI: 10.23736/s0394-9508.18.04816-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
Wei R, Kaneko T, Liu X, Liu H, Li L, Voss C, Liu E, He N, Li SSC. Interactome Mapping Uncovers a General Role for Numb in Protein Kinase Regulation. Mol Cell Proteomics 2018; 17:2216-2228. [PMID: 29217616 PMCID: PMC6210222 DOI: 10.1074/mcp.ra117.000114] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 12/04/2017] [Indexed: 12/24/2022] Open
Abstract
Cellular functions are frequently regulated by protein-protein interactions involving the binding of a modular domain in one protein to a specific peptide sequence in another. This mechanism may be explored to identify binding partners for proteins harboring a peptide-recognition domain. Here we report a proteomic strategy combining peptide and protein microarray screening with biochemical and cellular assays to identify modular domain-mediated protein-protein interactions in a systematic manner. We applied this strategy to Numb, a multi-functional protein containing a phosphotyrosine-binding (PTB) domain. Through the screening of a protein microarray, we identified >100 protein kinases, including both Tyr and Ser/Thr kinases, that could potentially interact with the Numb PTB domain, suggesting a general role for Numb in regulating kinase function. The putative interactions between Numb and several tyrosine kinases were subsequently validated by GST pull-down and/or co-immunoprecipitation assays. Furthermore, using the Oriented Peptide Array Library approach, we defined the specificity of the Numb PTB domain which, in turn, allowed us to predict binding partners for Numb at the genome level. The combination of the protein microarray screening with computer-aided prediction produced the most expansive interactome for Numb to date, implicating Numb in regulating phosphorylation signaling through protein kinases and phosphatases. Not only does the data generated from this study provide an important resource for hypothesis-driven research to further define the function of Numb, the proteomic strategy described herein may be employed to uncover the interactome for other peptide-recognition domains whose consensus motifs are known or can be determined.
Collapse
Affiliation(s)
- Ran Wei
- From the ‡Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Tomonori Kaneko
- From the ‡Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Xuguang Liu
- From the ‡Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Huadong Liu
- From the ‡Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
- §Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, Shanxi, China
| | - Lei Li
- From the ‡Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
- ¶School of Basic Medical Sciences, Qingdao University, Qingdao 266021, Shangdong, China
- ‖College of Pharmacy, Qingdao University, Qingdao 26601, Shangdong, China
| | - Courtney Voss
- From the ‡Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Eric Liu
- From the ‡Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada
| | - Ningning He
- ¶School of Basic Medical Sciences, Qingdao University, Qingdao 266021, Shangdong, China
- ‖College of Pharmacy, Qingdao University, Qingdao 26601, Shangdong, China
| | - Shawn S-C Li
- From the ‡Department of Biochemistry and the Siebens-Drake Medical Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario N6A 5C1, Canada;
- **Department of Oncology and Child Health Research Institute, Western University
| |
Collapse
|
20
|
Richter DJ, Fozouni P, Eisen MB, King N. Gene family innovation, conservation and loss on the animal stem lineage. eLife 2018; 7:34226. [PMID: 29848444 PMCID: PMC6040629 DOI: 10.7554/elife.34226] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 05/26/2018] [Indexed: 02/06/2023] Open
Abstract
Choanoflagellates, the closest living relatives of animals, can provide unique insights into the changes in gene content that preceded the origin of animals. However, only two choanoflagellate genomes are currently available, providing poor coverage of their diversity. We sequenced transcriptomes of 19 additional choanoflagellate species to produce a comprehensive reconstruction of the gains and losses that shaped the ancestral animal gene repertoire. We identified ~1944 gene families that originated on the animal stem lineage, of which only 39 are conserved across all animals in our study. In addition, ~372 gene families previously thought to be animal-specific, including Notch, Delta, and homologs of the animal Toll-like receptor genes, instead evolved prior to the animal-choanoflagellate divergence. Our findings contribute to an increasingly detailed portrait of the gene families that defined the biology of the Urmetazoan and that may underpin core features of extant animals. All animals, from sea sponges and reef-building corals to elephants and humans, share a single common ancestor that lived over half a billion years ago. This single-celled predecessor evolved the ability to develop into a creature made up of many cells with specialized jobs. Reconstructing the steps in this evolutionary process has been difficult because the earliest animals were soft-bodied and microscopic and did not leave behind fossils that scientists can study. Though their bodies have since disintegrated, many of the instructions for building the first animals live on in genes that were passed on to life forms that still exist. Scientists are trying to retrace those genes back to the first animal by comparing the genomes of living animals with their closest relatives, the choanoflagellates. Choanoflagellates are single-celled, colony-forming organisms that live in waters around the world. Comparisons with choanoflagellates may help scientists identify which genes were necessary to help animals evolve and diversify into so many different species. So far, 1,000 animal and two choanoflagellate genomes have been sequenced. But the gene repertoires of most species of choanoflagellates have yet to be analyzed. Now, Richter et al. have cataloged the genes of 19 more species of choanoflagellates. This added information allowed them to recreate the likely gene set of the first animal and to identify genetic changes that occurred during animal evolution. The analyses showed that modern animals lost about a quarter of the genes present in their last common ancestor with choanoflagellates and gained an equal number of new genes. Richter et al. identified several dozen core animal genes that were gained and subsequently preserved throughout animal evolution. Many of these are necessary so that an embryo can develop properly, but the precise roles of some core genes remain a mystery. Most other genes that emerged in the first animals have been lost in at least one living animal. The study of Richter et al. also showed that some very important genes in animals, including genes essential for early development and genes that help the immune system detect pathogens, predate animals. These key genes trace back to animals’ last common ancestor with choanoflagellates and may have evolved new roles in animals.
Collapse
Affiliation(s)
- Daniel J Richter
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States.,Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR 7144, Adaptation et Diversité en Milieu Marin, Équipe EPEP, Station Biologique de Roscoff, Roscoff, France
| | - Parinaz Fozouni
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States.,Medical Scientist Training Program, Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, United States.,Gladstone Institutes, San Francisco, United States
| | - Michael B Eisen
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| | - Nicole King
- Department of Molecular and Cell Biology, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
21
|
Seetharaman S, Etienne-Manneville S. Integrin diversity brings specificity in mechanotransduction. Biol Cell 2018; 110:49-64. [DOI: 10.1111/boc.201700060] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/08/2018] [Indexed: 12/29/2022]
Affiliation(s)
- Shailaja Seetharaman
- Institut Pasteur Paris CNRS UMR3691; Cell Polarity; Migration and Cancer Unit; Equipe Labellisée Ligue Contre le Cancer; Paris Cedex 15 France
- Université Paris Descartes, Sorbonne Paris Cité; Paris 75006 France
| | - Sandrine Etienne-Manneville
- Institut Pasteur Paris CNRS UMR3691; Cell Polarity; Migration and Cancer Unit; Equipe Labellisée Ligue Contre le Cancer; Paris Cedex 15 France
| |
Collapse
|
22
|
Ringer P, Colo G, Fässler R, Grashoff C. Sensing the mechano-chemical properties of the extracellular matrix. Matrix Biol 2017; 64:6-16. [DOI: 10.1016/j.matbio.2017.03.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 03/29/2017] [Accepted: 03/30/2017] [Indexed: 12/13/2022]
|
23
|
Abstract
During vascular development, endothelial cells (ECs) and neighboring stromal cells interact and communicate through autocrine and paracrine signaling mechanisms involving extracellular matrix (ECM) proteins and their cell surface integrin adhesion receptors. Integrin-mediated adhesion and signaling pathways are crucial for normal vascular development and physiology, and alterations in integrin expression and/or function drive several vascular-related pathologies including thrombosis, autoimmune disorders, and cancer. The purpose of this chapter is to discuss integrin adhesion and signaling pathways important for EC growth, survival, and migration. Integrin-mediated paracrine links between ECs and surrounding stromal cells in the organ microenvironment will also be discussed. Lastly, we will review roles for integrins in vascular pathologies and discuss possible targets for therapeutic intervention.
Collapse
Affiliation(s)
- Paola A Guerrero
- University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Joseph H McCarty
- University of Texas MD Anderson Cancer Center, Houston, TX, United States.
| |
Collapse
|
24
|
Research advances on structure and biological functions of integrins. SPRINGERPLUS 2016; 5:1094. [PMID: 27468395 PMCID: PMC4947080 DOI: 10.1186/s40064-016-2502-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 06/02/2016] [Indexed: 12/18/2022]
Abstract
Integrins are an important family of adhesion molecules that were first discovered two decades ago. Integrins are transmembrane heterodimeric glycoprotein receptors consisting of α and β subunits, and are comprised of an extracellular domain, a transmembrane domain, and a cytoplasmic tail. Therein, integrin cytoplasmic domains may associate directly with numerous cytoskeletal proteins and intracellular signaling molecules, which are crucial for modulating fundamental cell processes and functions including cell adhesion, proliferation, migration, and survival. The purpose of this review is to describe the unique structure of each integrin subunit, primary cytoplasmic association proteins, and transduction signaling pathway of integrins, with an emphasis on their biological functions.
Collapse
|
25
|
Topalovski M, Brekken RA. Matrix control of pancreatic cancer: New insights into fibronectin signaling. Cancer Lett 2015; 381:252-8. [PMID: 26742464 DOI: 10.1016/j.canlet.2015.12.027] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 12/18/2015] [Accepted: 12/19/2015] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a highly metastatic disease that resists most current therapies. A defining characteristic of PDA is an intense fibrotic response that promotes tumor cell invasion and chemoresistance. Efforts to understand the complex relationship between the tumor and its extracellular network and to therapeutically perturb tumor-stroma interactions are ongoing. Fibronectin (FN), a provisional matrix protein abundant in PDA stroma but not normal tissues, supports metastatic spread and chemoresistance of this deadly disease. FN also supports angiogenesis, which is required for even hypovascular tumors such as PDA to develop and progress. Targeting components of the tumor stroma, such as FN, can effectively reduce tumor growth and spread while also enhancing delivery of chemotherapy. Here, we review the molecular mechanisms by which FN drives angiogenesis, metastasis and chemoresistance in PDA. In light of these new findings, we also discuss therapeutic strategies to inhibit FN signaling.
Collapse
Affiliation(s)
- Mary Topalovski
- Hamon Center for Therapeutic Oncology Research and the Division of Surgical Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rolf A Brekken
- Hamon Center for Therapeutic Oncology Research and the Division of Surgical Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA; Departments of Surgery and Pharmacology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
26
|
Martínez-Moreno M, Leiva M, Aguilera-Montilla N, Sevilla-Movilla S, Isern de Val S, Arellano-Sánchez N, Gutiérrez NC, Maldonado R, Martínez-López J, Buño I, García-Marco JA, Sánchez-Mateos P, Hidalgo A, García-Pardo A, Teixidó J. In vivo adhesion of malignant B cells to bone marrow microvasculature is regulated by α4β1 cytoplasmic-binding proteins. Leukemia 2015; 30:861-72. [PMID: 26658839 DOI: 10.1038/leu.2015.332] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 10/23/2015] [Accepted: 11/24/2015] [Indexed: 12/14/2022]
Abstract
Multiple myeloma (MM) and chronic lymphocytic leukemia (CLL) cells must attach to the bone marrow (BM) microvasculature before lodging in the BM microenvironment. Using intravital microscopy (IVM) of the BM calvariae we demonstrate that the α4β1 integrin is required for MM and CLL cell firm arrest onto the BM microvasculature, while endothelial P-selectin and E-selectin mediate cell rolling. Talin, kindlin-3 and ICAP-1 are β1-integrin-binding partners that regulate β1-mediated cell adhesion. We show that talin and kindlin-3 cooperatively stimulate high affinity and strength of α4β1-dependent MM and CLL cell attachment, whereas ICAP-1 negatively regulates this adhesion. A functional connection between talin/kindlin-3 and Rac1 was found to be required for MM cell attachment mediated by α4β1. Importantly, IVM analyses with talin- and kindlin-3-silenced MM cells indicate that these proteins are needed for cell arrest on the BM microvasculature. Instead, MM cell arrest is repressed by ICAP-1. Moreover, MM cells silenced for talin and kindlin-3, and cultured on α4β1 ligands showed higher susceptibility to bortezomib-mediated cell apoptosis. Our results highlight the requirement of α4β1 and selectins for the in vivo attachment of MM and CLL cells to the BM microvasculature, and indicate that talin, kindlin-3 and ICAP-1 differentially control physiological adhesion by regulating α4β1 activity.
Collapse
Affiliation(s)
- M Martínez-Moreno
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - M Leiva
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - N Aguilera-Montilla
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - S Sevilla-Movilla
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - S Isern de Val
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - N Arellano-Sánchez
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - N C Gutiérrez
- Department of Hematology, Hospital Universitario de Salamanca, Salamanca, Spain
| | - R Maldonado
- Section of Hematology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - J Martínez-López
- Section of Hematology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - I Buño
- Section of Hematology, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - J A García-Marco
- Hematology Unit, Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - P Sánchez-Mateos
- Section of Immuno-Oncology, Hospital General Universitario Gregorio Marañón, Complutense University School of Medicine, Madrid, Spain
| | - A Hidalgo
- Area of Cell and Developmental Biology, Fundación Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.,Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany
| | - A García-Pardo
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - J Teixidó
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| |
Collapse
|
27
|
Wei C, Li X, Zhang P, Zhang Y, Liu T, Jiang S, Han F, Zhang Y. Characterization of porcine partially reprogrammed iPSCs from adipose-derived stem cells. Reproduction 2015; 149:485-96. [PMID: 25646510 DOI: 10.1530/rep-14-0410] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Partially reprogrammed induced pluripotent stem cells (PiPSCs) have great potential for investigating reprogramming mechanisms and represent an alternative potential material for making genetically modified animals and regenerative medicine. To date, PiPSCs have scarcely been reported in detail when compared with mice and humans. In this study, we obtained PiPSCs from porcine adipose-derived stem cells (pADSCs) by ectopic expression of human transcription factors (OCT4, SOX2, c-MYC, and KLF4) in feeder-free condition. The morphology and proliferation activity of porcine PiPSCs (pPiPSCs) were similar to those of porcine fully reprogrammed iPSCs (pFiPSCs); furthermore, pPiPSCs expressed higher levels of the typical surface molecules (CD29) found in pADSCs. However, pPiPSCs were negative for key proteins (NANOG) connected with stemness and possessed lower differentiation ability in vivo and in vitro. When differentiation-inhibiting factors were withdrawn, pPiPSCs-derived cells (pPiPSC-DCs) showed similar features to pADSCs in many aspects, including proliferation, differentiation, and immunosuppression. When both types of cells were used to produce cloned embryos, we found that the blastocyst formation rate of 19DC (one of the pPiPSC-DC cell lines)-derived cloned embryos was obviously higher than that of others. The total cell number of 19DC-derived blastocysts was significantly higher than the 30DC (one pFiPSC-DC cell line)-derived blastocysts. In all, through limited differentiation ability, the proliferation activity of pPiPSCs is similar to that of pFiPSCs, and pPiPSCs can retain several of the features of pADSCs, which are beneficial to cell therapy. Furthermore, the differentiation of pPiPSCs is more favorable for producing high-quality reconstructed embryos.
Collapse
Affiliation(s)
- Chao Wei
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and BreedingCollege of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Xia Li
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and BreedingCollege of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Pengfei Zhang
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and BreedingCollege of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yu Zhang
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and BreedingCollege of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Tong Liu
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and BreedingCollege of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Shaoshuai Jiang
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and BreedingCollege of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Fei Han
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and BreedingCollege of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| | - Yunhai Zhang
- Anhui Provincial Laboratory for Local Livestock and Poultry Genetic Resource Conservation and BreedingCollege of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui 230036, China
| |
Collapse
|
28
|
Baxter SS, Dibble CF, Byrd WC, Carlson J, Mack CR, Saldarriaga I, Bencharit S. Role of cytoskeletal proteins in cerebral cavernous malformation signaling pathways: a proteomic analysis. MOLECULAR BIOSYSTEMS 2015; 10:1881-9. [PMID: 24770493 PMCID: PMC4043921 DOI: 10.1039/c3mb70199a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Three genetic mutations were found to cause cerebral cavernous malformation (CCM), a vascular anomaly predisposing affected individuals to hemorrhagic stroke. These CCM proteins function together as a protein complex in the cell. Loss of expression of each CCM gene results in loss of in vitro endothelial tube formation. Label-free differential protein expression analysis using multidimensional liquid chromatography/tandem mass spectrometry (2D-LC-MS/MS) was applied to explore the proteomic profile for loss of each CCM gene expression in mouse endothelial stem cells (MEES) compared to mock shRNA and no shRNA control cell-lines. Differentially expressed proteins were identified (p < 0.05). 120 proteins were differentially expressed among the cell-lines. Principal component analysis and cluster analysis show the effects of individual knockdown. In all knockdown cell-lines, altered expression of cytoskeletal proteins is the most common. While all CCM mutations result in similar pathology, different CCM mutations have their own distinct pathogenesis in cell signaling.
Collapse
Affiliation(s)
- Sarah Schwartz Baxter
- David H. Murdock Research Institute, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Janus-faces of NME-oncoprotein interactions. Naunyn Schmiedebergs Arch Pharmacol 2014; 388:175-87. [PMID: 25366701 DOI: 10.1007/s00210-014-1062-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 10/15/2014] [Indexed: 12/26/2022]
Abstract
Since the identification of Nm23 (NME1, NME/NM23 nucleoside diphosphate kinase 1) as the first non-metastatic protein, a great deal of research on members of the NME family of proteins has focused on roles in processes implicated in carcinogenesis and particularly their regulation of cellular motility and the process of metastatic spread. To date, there are ten identified members of this family of genes, and these can be dichotomized into groups both taxonomically and by the presence or absence of their nucleoside diphosphate kinase activity with NMEs 1-4 encoding nucleoside diphosphate kinases (NDPKs) and NMEs 5-9 plus RP2 displaying little if any NDPK activity. NMEs are relatively small proteins that can form hetero-oligomers (typically hexamers), and given the apparent genetic redundancy of some NMEs and the number of different isoforms, it is perhaps not surprising that there remains a great deal of uncertainty regarding their function and even more regarding cellular mechanisms of action. Since residues that contribute to NDPK activity span much of the protein, it seems likely that the consequences of NME expression must be mediated through their NDPK activity, through interactions with other structures in cells including protein-protein interactions or through combinations of these. Our goal in this review is to focus on some of the protein-protein interactions that have been identified and to highlight some of the challenges that face this area of research.
Collapse
|
30
|
Shekaran A, Shoemaker JT, Kavanaugh TE, Lin AS, LaPlaca MC, Fan Y, Guldberg RE, García AJ. The effect of conditional inactivation of beta 1 integrins using twist 2 Cre, Osterix Cre and osteocalcin Cre lines on skeletal phenotype. Bone 2014; 68:131-41. [PMID: 25183373 PMCID: PMC4189988 DOI: 10.1016/j.bone.2014.08.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Revised: 08/13/2014] [Accepted: 08/16/2014] [Indexed: 11/27/2022]
Abstract
Skeletal development and growth are complex processes regulated by multiple microenvironmental cues, including integrin-ECM interactions. The β1 sub-family of integrins is the largest integrin sub-family and constitutes the main integrin binding partners of collagen I, the major ECM component of bone. As complete β1 integrin knockout results in embryonic lethality, studies of β1 integrin function in vivo rely on tissue-specific gene deletions. While multiple in vitro studies indicate that β1 integrins are crucial regulators of osteogenesis and mineralization, in vivo osteoblast-specific perturbations of β1 integrins have resulted in mild and sometimes contradictory skeletal phenotypes. To further investigate the role of β1 integrins on skeletal phenotype, we used the Twist2-Cre, Osterix-Cre and osteocalcin-Cre lines to generate conditional β1 integrin deletions, where Cre is expressed primarily in mesenchymal condensation, pre-osteoblast, and mature osteoblast lineage cells respectively within these lines. Mice with Twist2-specific β1 integrin disruption were smaller, had impaired skeletal development, especially in the craniofacial and vertebral tissues at E19.5, and did not survive beyond birth. Osterix-specific β1 integrin deficiency resulted in viable mice which were normal at birth but displayed early defects in calvarial ossification, incisor eruption and growth as well as femoral bone mineral density, structure, and mechanical properties. Although these defects persisted into adulthood, they became milder with age. Finally, a lack of β1 integrins in mature osteoblasts and osteocytes resulted in minor alterations to femur structure but had no effect on mineral density, biomechanics or fracture healing. Taken together, our data indicate that β1 integrin expression in early mesenchymal condensations play an important role in skeletal ossification, while β1 integrin-ECM interactions in pre-osteoblast, odontoblast- and hypertrophic chondryocyte-lineage cells regulate incisor eruption and perinatal bone formation in both intramembranously and endochondrally formed bones in young, rapidly growing mice. In contrast, the osteocalcin-specific β1 integrin deletion had only minor effects on skeletal phenotype.
Collapse
Affiliation(s)
- Asha Shekaran
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332, USA; Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332, USA
| | - James T Shoemaker
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Taylor E Kavanaugh
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Angela S Lin
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332, USA; School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332, USA
| | - Michelle C LaPlaca
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Yuhong Fan
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332, USA; School of Biology, Georgia Institute of Technology, 310 Ferst Drive, Atlanta, GA 30332, USA
| | - Robert E Guldberg
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332, USA; School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, Atlanta, GA 30332, USA; School of Mechanical Engineering, Georgia Institute of Technology, 801 Ferst Drive, Atlanta, GA 30332, USA.
| |
Collapse
|
31
|
Hytönen VP, Wehrle-Haller B. Protein conformation as a regulator of cell–matrix adhesion. Phys Chem Chem Phys 2014; 16:6342-57. [DOI: 10.1039/c3cp54884h] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Conformational changes within proteins play key roles in the regulation of cell–matrix adhesion. We discuss the mechanisms involved in conformational regulation, including mechanical signals, posttranslational modifications and intrinsically disordered proteins.
Collapse
Affiliation(s)
- Vesa P. Hytönen
- University of Tampere
- Institute of Biomedical Technology and BioMediTech
- 33520 Tampere, Finland
- Fimlab Laboratories
- 33014 Tampere, Finland
| | - Bernhard Wehrle-Haller
- University of Geneva
- Department of Cell Physiology and Metabolism
- Centre Médical Universitaire
- 1211 Geneva 4, Switzerland
| |
Collapse
|
32
|
Fisher OS, Boggon TJ. Signaling pathways and the cerebral cavernous malformations proteins: lessons from structural biology. Cell Mol Life Sci 2013; 71:1881-92. [PMID: 24287896 DOI: 10.1007/s00018-013-1532-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/19/2013] [Accepted: 11/21/2013] [Indexed: 10/26/2022]
Abstract
Cerebral cavernous malformations (CCM) are neurovascular dysplasias that result in mulberry-shaped lesions predominantly located in brain and spinal tissues. Mutations in three genes are associated with CCM. These genes encode for the proteins KRIT1/CCM1 (krev interaction trapped 1/cerebral cavernous malformations 1), cerebral cavernous malformations 2, osmosensing scaffold for MEKK3 (CCM2/malcavernin/OSM), and cerebral cavernous malformations 3/programmed cell death 10 (CCM3/PDCD10). There have been many significant recent advances in our understanding of the structure and function of these proteins, as well as in their roles in cellular signaling. Here, we provide an update on the current knowledge of the structure of the CCM proteins and their functions within cellular signaling, particularly in cellular adhesion complexes and signaling cascades. We go on to discuss subcellular localization of the CCM proteins, the formation and regulation of the CCM complex signaling platform, and current progress towards targeted therapy for CCM disease. Recent structural studies have begun to shed new light on CCM protein function, and we focus here on how these studies have helped inform the current understanding of these roles and how they may aid future studies into both CCM-related biology and disease mechanisms.
Collapse
Affiliation(s)
- Oriana S Fisher
- Department of Pharmacology, Yale University School of Medicine, SHM B-316A, 333 Cedar Street, New Haven, CT, 06520, USA
| | | |
Collapse
|
33
|
Faurobert E, Rome C, Lisowska J, Manet-Dupé S, Boulday G, Malbouyres M, Balland M, Bouin AP, Kéramidas M, Bouvard D, Coll JL, Ruggiero F, Tournier-Lasserve E, Albiges-Rizo C. CCM1-ICAP-1 complex controls β1 integrin-dependent endothelial contractility and fibronectin remodeling. ACTA ACUST UNITED AC 2013; 202:545-61. [PMID: 23918940 PMCID: PMC3734079 DOI: 10.1083/jcb.201303044] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Loss of CCM1/2 leads to destabilization of ICAP-1 and up-regulation of β1 integrin, resulting in the destabilization of intercellular junctions due to increased cell contractility and aberrant extracellular matrix remodeling. The endothelial CCM complex regulates blood vessel stability and permeability. Loss-of-function mutations in CCM genes are responsible for human cerebral cavernous malformations (CCMs), which are characterized by clusters of hemorrhagic dilated capillaries composed of endothelium lacking mural cells and altered sub-endothelial extracellular matrix (ECM). Association of the CCM1/2 complex with ICAP-1, an inhibitor of β1 integrin, prompted us to investigate whether the CCM complex interferes with integrin signaling. We demonstrate that CCM1/2 loss resulted in ICAP-1 destabilization, which increased β1 integrin activation and led to increased RhoA-dependent contractility. The resulting abnormal distribution of forces led to aberrant ECM remodeling around lesions of CCM1- and CCM2-deficient mice. ICAP-1–deficient vessels displayed similar defects. We demonstrate that a positive feedback loop between the aberrant ECM and internal cellular tension led to decreased endothelial barrier function. Our data support that up-regulation of β1 integrin activation participates in the progression of CCM lesions by destabilizing intercellular junctions through increased cell contractility and aberrant ECM remodeling.
Collapse
Affiliation(s)
- Eva Faurobert
- INSERM U823, Institut Albert Bonniot, Grenoble F-38042, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Bouvard D, Pouwels J, De Franceschi N, Ivaska J. Integrin inactivators: balancing cellular functions in vitro and in vivo. Nat Rev Mol Cell Biol 2013; 14:430-42. [DOI: 10.1038/nrm3599] [Citation(s) in RCA: 173] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
Millon-Frémillon A, Brunner M, Abed N, Collomb E, Ribba AS, Block MR, Albigès-Rizo C, Bouvard D. Calcium and calmodulin-dependent serine/threonine protein kinase type II (CaMKII)-mediated intramolecular opening of integrin cytoplasmic domain-associated protein-1 (ICAP-1α) negatively regulates β1 integrins. J Biol Chem 2013; 288:20248-60. [PMID: 23720740 DOI: 10.1074/jbc.m113.455956] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Focal adhesion turnover during cell migration is an integrated cyclic process requiring tight regulation of integrin function. Interaction of integrin with its ligand depends on its activation state, which is regulated by the direct recruitment of proteins onto the β integrin chain cytoplasmic domain. We previously reported that ICAP-1α, a specific cytoplasmic partner of β1A integrins, limits both talin and kindlin interaction with β1 integrin, thereby restraining focal adhesion assembly. Here we provide evidence that the calcium and calmodulin-dependent serine/threonine protein kinase type II (CaMKII) is an important regulator of ICAP-1α for controlling focal adhesion dynamics. CaMKII directly phosphorylates ICAP-1α and disrupts an intramolecular interaction between the N- and the C-terminal domains of ICAP-1α, unmasking the PTB domain, thereby permitting ICAP-1α binding onto the β1 integrin tail. ICAP-1α direct interaction with the β1 integrin tail and the modulation of β1 integrin affinity state are required for down-regulating focal adhesion assembly. Our results point to a molecular mechanism for the phosphorylation-dependent control of ICAP-1α function by CaMKII, allowing the dynamic control of β1 integrin activation and cell adhesion.
Collapse
|
36
|
Liu W, Boggon TJ. Cocrystal structure of the ICAP1 PTB domain in complex with a KRIT1 peptide. Acta Crystallogr Sect F Struct Biol Cryst Commun 2013; 69:494-8. [PMID: 23695561 DOI: 10.1107/s1744309113010762] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 04/19/2013] [Indexed: 11/11/2022]
Abstract
Integrin cytoplasmic domain-associated protein-1 (ICAP1) is a suppressor of integrin activation and directly binds to the cytoplasmic tail of β1 integrins; its binding suppresses integrin activation by competition with talin. Krev/Rap1 interaction trapped-1 (KRIT1) releases ICAP1 suppression of integrin activation by sequestering ICAP1 away from integrin cytoplasmic tails. Here, the cocrystal structure of the PTB domain of ICAP1 in complex with a 29-amino-acid fragment (residues 170-198) of KRIT1 is presented to 1.7 Å resolution [the resolution at which 〈I/σ(I)〉 = 2.9 was 1.83 Å]. In previous studies, the structure of ICAP1 with integrin β1 was determined to 3.0 Å resolution and that of ICAP1 with the N-terminal portion of KRIT1 (residues 1-198) was determined to 2.54 Å resolution; therefore, this study provides the highest resolution structure yet of ICAP1 and allows further detailed analysis of the interaction of ICAP1 with its minimal binding region in KRIT1.
Collapse
Affiliation(s)
- Weizhi Liu
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | |
Collapse
|
37
|
Liu W, Draheim KM, Zhang R, Calderwood DA, Boggon TJ. Mechanism for KRIT1 release of ICAP1-mediated suppression of integrin activation. Mol Cell 2013; 49:719-29. [PMID: 23317506 DOI: 10.1016/j.molcel.2012.12.005] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Revised: 09/04/2012] [Accepted: 11/07/2012] [Indexed: 10/27/2022]
Abstract
KRIT1 (Krev/Rap1 Interaction Trapped-1) mutations are observed in ∼40% of autosomal-dominant cerebral cavernous malformations (CCMs), a disease occurring in up to 0.5% of the population. We show that KRIT1 functions as a switch for β1 integrin activation by antagonizing ICAP1 (Integrin Cytoplasmic Associated Protein-1)-mediated modulation of "inside-out" activation. We present cocrystal structures of KRIT1 with ICAP1 and ICAP1 with integrin β1 cytoplasmic tail to 2.54 and 3.0 Å resolution (the resolutions at which I/σI = 2 are 2.75 and 3.0 Å, respectively). We find that KRIT1 binds ICAP1 by a bidentate surface, that KRIT1 directly competes with integrin β1 to bind ICAP1, and that KRIT1 antagonizes ICAP1-modulated integrin activation using this site. We also find that KRIT1 contains an N-terminal Nudix domain, in a region previously designated as unstructured. We therefore provide insights to integrin regulation and CCM-associated KRIT1 function.
Collapse
Affiliation(s)
- Weizhi Liu
- Department of Pharmacology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | | | | | | | | |
Collapse
|
38
|
New insights into adhesion signaling in bone formation. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2013; 305:1-68. [PMID: 23890379 DOI: 10.1016/b978-0-12-407695-2.00001-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mineralized tissues that are protective scaffolds in the most primitive species have evolved and acquired more specific functions in modern animals. These are as diverse as support in locomotion, ion homeostasis, and precise hormonal regulation. Bone formation is tightly controlled by a balance between anabolism, in which osteoblasts are the main players, and catabolism mediated by the osteoclasts. The bone matrix is deposited in a cyclic fashion during homeostasis and integrates several environmental cues. These include diffusible elements that would include estrogen or growth factors and physicochemical parameters such as bone matrix composition, stiffness, and mechanical stress. Therefore, the microenvironment is of paramount importance for controlling this delicate equilibrium. Here, we provide an overview of the most recent data highlighting the role of cell-adhesion molecules during bone formation. Due to the very large scope of the topic, we focus mainly on the role of the integrin receptor family during osteogenesis. Bone phenotypes of some deficient mice as well as diseases of human bones involving cell adhesion during this process are discussed in the context of bone physiology.
Collapse
|
39
|
Pouwels J, Nevo J, Pellinen T, Ylänne J, Ivaska J. Negative regulators of integrin activity. J Cell Sci 2012; 125:3271-80. [PMID: 22822081 DOI: 10.1242/jcs.093641] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Integrins are heterodimeric transmembrane adhesion receptors composed of α- and β-subunits. They are ubiquitously expressed and have key roles in a number of important biological processes, such as development, maintenance of tissue homeostasis and immunological responses. The activity of integrins, which indicates their affinity towards their ligands, is tightly regulated such that signals inside the cell cruicially regulate the switching between active and inactive states. An impaired ability to activate integrins is associated with many human diseases, including bleeding disorders and immune deficiencies, whereas inappropriate integrin activation has been linked to inflammatory disorders and cancer. In recent years, the molecular details of integrin 'inside-out' activation have been actively investigated. Binding of cytoplasmic proteins, such as talins and kindlins, to the cytoplasmic tail of β-integrins is widely accepted as being the crucial step in integrin activation. By contrast, much less is known with regard to the counteracting mechanism involved in switching integrins into an inactive conformation. In this Commentary, we aim to discuss the known mechanisms of integrin inactivation and the molecules involved.
Collapse
Affiliation(s)
- Jeroen Pouwels
- Centre for Biotechnology, University of Turku, Turku, Finland
| | | | | | | | | |
Collapse
|
40
|
Cheng TY, Wu MS, Lin JT, Lin MT, Shun CT, Huang HY, Hua KT, Kuo ML. Annexin A1 is associated with gastric cancer survival and promotes gastric cancer cell invasiveness through the formyl peptide receptor/extracellular signal-regulated kinase/integrin beta-1-binding protein 1 pathway. Cancer 2012; 118:5757-67. [PMID: 22736399 DOI: 10.1002/cncr.27565] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2011] [Revised: 11/23/2011] [Accepted: 02/29/2012] [Indexed: 01/08/2023]
Abstract
BACKGROUND Annexin A1 (AnxA1) has been well-known as a glucocorticoid-regulated anti-inflammatory protein, and it is implicated in tumorigenesis in a tumor type-specific pattern. However, the role of AnxA1 in gastric cancer (GC) is indeterminate, and the underlying mechanism is not clear. The purpose of this study was to evaluate the prognostic significance and associated mechanism of AnxA1 in GC. METHODS Immunohistochemical staining was employed to analyze 118 GC patients. Both AnxA1 gain-of-function and loss-of-function approaches were performed in GC cells. Western blotting and reverse-transcription polymerase chain reaction were used for assessment of the AnxA1 regulation mechanism in GC cells. An intraperitoneal inoculation model in severe combined immunodeficient mice was used for an in vivo assay. RESULTS High AnxA1 expression was significantly associated with peritoneal metastasis (P = .009) and serosal invasion (P = .044). Cox multivariate analysis showed that high AnxA1 expression was an independent risk factor for poor overall survival in GC patients (P = .037). AnxA1 expression positively correlated with invasiveness of human GC cells both in vitro and in vivo. AnxA1 could regulate the GC cell invasion through the formyl peptide receptor (FPR)/extracellular signal-regulated kinase/integrin beta-1-binding protein pathway, and all 3 FPRs (FPR1 through FPR3) were involved in the regulation process. CONCLUSIONS High AnxA1 expression was associated with more serosal invasion, more peritoneal metastasis, and poorer overall survival in GC patients. The current study demonstrated a novel mechanism involving FPRs, extracellular signal-regulated kinases 1 and 2, and integrin beta-1-binding protein 1 by which AnxA1 regulated GC cell invasion.
Collapse
Affiliation(s)
- Tsu-Yao Cheng
- Graduate Institute of Toxicology, National Taiwan University College of Medicine, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Zheng Y, Qiu J, Hu J, Wang G. Concepts and hypothesis: integrin cytoplasmic domain-associated protein-1 (ICAP-1) as a potential player in cerebral cavernous malformation. J Neurol 2012; 260:10-9. [DOI: 10.1007/s00415-012-6567-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 05/18/2012] [Accepted: 05/18/2012] [Indexed: 11/28/2022]
|
42
|
Cavalcanti DD, Kalani MYS, Martirosyan NL, Eales J, Spetzler RF, Preul MC. Cerebral cavernous malformations: from genes to proteins to disease. J Neurosurg 2011; 116:122-32. [PMID: 21962164 DOI: 10.3171/2011.8.jns101241] [Citation(s) in RCA: 92] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Over the past half century molecular biology has led to great advances in our understanding of angio- and vasculogenesis and in the treatment of malformations resulting from these processes gone awry. Given their sporadic and familial distribution, their developmental and pathological link to capillary telangiectasias, and their observed chromosomal abnormalities, cerebral cavernous malformations (CCMs) are regarded as akin to cancerous growths. Although the exact pathological mechanisms involved in the formation of CCMs are still not well understood, the identification of 3 genetic loci has begun to shed light on key developmental pathways involved in CCM pathogenesis. Cavernous malformations can occur sporadically or in an autosomal dominant fashion. Familial forms of CCMs have been attributed to mutations at 3 different loci implicated in regulating important processes such as proliferation and differentiation of angiogenic precursors and members of the apoptotic machinery. These processes are important for the generation, maintenance, and pruning of every vessel in the body. In this review the authors highlight the latest discoveries pertaining to the molecular genetics of CCMs, highlighting potential new therapeutic targets for the treatment of these lesions.
Collapse
Affiliation(s)
- Daniel D Cavalcanti
- Division of Neurological Surgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona 85013, USA
| | | | | | | | | | | |
Collapse
|
43
|
Holmes RS, Rout UK. Comparative studies of vertebrate Beta integrin genes and proteins: ancient genes in vertebrate evolution. Biomolecules 2011; 1:3-31. [PMID: 24970121 PMCID: PMC4030831 DOI: 10.3390/biom1010003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 08/14/2011] [Accepted: 08/15/2011] [Indexed: 12/31/2022] Open
Abstract
Intregins are heterodimeric α- and β-subunit containing membrane receptor proteins which serve various cell adhesion roles in tissue repair, hemostasis, immune response, embryogenesis and metastasis. At least 18 α- (ITA or ITGA) and 8 β-integrin subunits (ITB or ITGB) are encoded on mammalian genomes. Comparative ITB amino acid sequences and protein structures and ITB gene locations were examined using data from several vertebrate genome projects. Vertebrate ITB genes usually contained 13-16 coding exons and encoded protein subunits with ~800 amino acids, whereas vertebrate ITB4 genes contained 36-39 coding exons and encoded larger proteins with ~1800 amino acids. The ITB sequences exhibited several conserved domains including signal peptide, extracellular β-integrin, β-tail domain and integrin β-cytoplasmic domains. Sequence alignments of the integrin β-cytoplasmic domains revealed highly conserved regions possibly for performing essential functions and its maintenance during vertebrate evolution. With the exception of the human ITB8 sequence, the other ITB sequences shared a predicted 19 residue α-helix for this region. Potential sites for regulating human ITB gene expression were identified which included CpG islands, transcription factor binding sites and microRNA binding sites within the 3'-UTR of human ITB genes. Phylogenetic analyses examined the relationships of vertebrate beta-integrin genes which were consistent with four major groups: 1: ITB1, ITB2, ITB7; 2: ITB3, ITB5, ITB6; 3: ITB4; and 4: ITB8 and a common evolutionary origin from an ancestral gene, prior to the appearance of fish during vertebrate evolution. The phylogenetic analyses revealed that ITB4 is the most likely primordial form of the vertebrate β integrin subunit encoding genes, that is the only β subunit expressed as a constituent of the sole integrin receptor 'α6β4' in the hemidesmosomes of unicellular organisms.
Collapse
Affiliation(s)
- Roger S Holmes
- School of Biomolecular and Physical Sciences, Griffith University, Nathan, 4111QLD, Australia.
| | - Ujjwal K Rout
- Department of Surgery, University of Mississippi Medical Center, Jackson, MS 38677, USA.
| |
Collapse
|
44
|
Brunner M, Millon-Frémillon A, Chevalier G, Nakchbandi IA, Mosher D, Block MR, Albigès-Rizo C, Bouvard D. Osteoblast mineralization requires beta1 integrin/ICAP-1-dependent fibronectin deposition. ACTA ACUST UNITED AC 2011; 194:307-22. [PMID: 21768292 PMCID: PMC3144405 DOI: 10.1083/jcb.201007108] [Citation(s) in RCA: 94] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
ICAP-1 prevents recruitment of kindlin-2 to β1 integrin to control
dynamics of fibrillar adhesion sites, fibronectin deposition, and osteoblast
mineralization during bone formation. The morphogenetic and differentiation events required for bone formation are
orchestrated by diffusible and insoluble factors that are localized within the
extracellular matrix. In mice, the deletion of ICAP-1, a modulator of β1
integrin activation, leads to severe defects in osteoblast proliferation,
differentiation, and mineralization and to a delay in bone formation. Deposition
of fibronectin and maturation of fibrillar adhesions, adhesive structures that
accompany fibronectin deposition, are impaired upon ICAP-1 loss, as are type I
collagen deposition and mineralization. Expression of β1 integrin with a
mutated binding site for ICAP-1 recapitulates the ICAP-1–null phenotype.
Follow-up experiments demonstrated that ICAP-1 negatively regulates kindlin-2
recruitment onto the β1 integrin cytoplasmic domain, whereas an excess of
kindlin-2 binding has a deleterious effect on fibrillar adhesion formation.
These results suggest that ICAP-1 works in concert with kindlin-2 to control the
dynamics of β1 integrin–containing fibrillar adhesions and,
thereby, regulates fibronectin deposition and osteoblast mineralization.
Collapse
Affiliation(s)
- Molly Brunner
- Equipe 1 Dynamique des Systèmes d'Adhérence et Différenciation Cellulaire, Institut National de la Santé et de la Recherche Médicale U823, Institut Albert Bonniot, 38042 Grenoble, Cedex 09, France
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Regulation of cell-cell and cell-matrix interaction is essential for the normal physiology of metazoans and is important in many diseases. Integrin adhesion receptors can rapidly increase their affinity (integrin activation) in response to intracellular signaling events in a process termed inside-out signaling. The transmembrane domains of integrins and their interactions with the membrane are important in inside-out signaling. Moreover, integrin activation is tightly regulated by a complex network of signaling pathways. Here, we review recent progress in understanding how the membrane environment can, in cooperation with integrin-binding proteins, regulate integrin activation.
Collapse
Affiliation(s)
- Chungho Kim
- Department of Medicine, University of California, San Diego, La Jolla, California 92093, USA
| | | | | |
Collapse
|
46
|
Estrach S, Cailleteau L, Franco CA, Gerhardt H, Stefani C, Lemichez E, Gagnoux-Palacios L, Meneguzzi G, Mettouchi A. Laminin-binding integrins induce Dll4 expression and Notch signaling in endothelial cells. Circ Res 2011; 109:172-82. [PMID: 21474814 DOI: 10.1161/circresaha.111.240622] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
RATIONALE Integrins play a crucial role in controlling endothelial cell proliferation and migration during angiogenesis. The Delta-like 4 (Dll4)/Notch pathway establishes an adequate ratio between stalk and tip cell populations by restricting tip cell formation through "lateral inhibition" in response to a vascular endothelial growth factor gradient. Because angiogenesis requires a tight coordination of these cellular processes, we hypothesized that adhesion, vascular endothelial growth factor, and Notch signaling pathways are interconnected. OBJECTIVE This study was aimed at characterizing the cross-talk between integrin and Notch signaling in endothelial cells. METHODS AND RESULTS Adhesion of primary human endothelial cells to laminin-111 triggers Dll4 expression, leading to subsequent Notch pathway activation. SiRNA-mediated knockdown of α2β1 and α6β1 integrins abolishes Dll4 induction, which discloses a selective integrin signaling acting upstream of Notch pathway. The increase in Foxc2 transcription, triggered by α2β1 binding to laminin, is required but not sufficient per se for Dll4 expression. Furthermore, vascular endothelial growth factor stimulates laminin γ1 deposition, which leads to integrin signaling and Dll4 induction. Interestingly, loss of integrins α2 or α6 mimics the effects of Dll4 silencing and induces excessive network branching in an in vitro sprouting angiogenesis assay on three-dimensional matrigel. CONCLUSIONS We show that, in endothelial cells, ligation of α2β1 and α6β1 integrins induces the Notch pathway, and we disclose a novel role of basement membrane proteins in the processes controlling tip vs stalk cell selection.
Collapse
|
47
|
Régent M, Planus E, Bouin AP, Bouvard D, Brunner M, Faurobert E, Millon-Frémillon A, Block MR, Albiges-Rizo C. Specificities of β1 integrin signaling in the control of cell adhesion and adhesive strength. Eur J Cell Biol 2010; 90:261-9. [PMID: 20971526 DOI: 10.1016/j.ejcb.2010.09.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2010] [Revised: 09/01/2010] [Accepted: 09/02/2010] [Indexed: 11/26/2022] Open
Abstract
Cells exert actomyosin contractility and cytoskeleton-dependent force in response to matrix stiffness cues. Cells dynamically adapt to force by modifying their behavior and remodeling their microenvironment. This adaptation is favored by integrin activation switch and their ability to modulate their clustering and the assembly of an intracellular hub in response to force. Indeed integrins are mechanoreceptors and mediate mechanotransduction by transferring forces to specific adhesion proteins into focal adhesions which are sensitive to tension and activate intracellular signals. α(5)β(1) integrin is considered of major importance for the formation of an elaborate meshwork of fibronectin fibrils and for the extracellular matrix deposition and remodeling. Here we summarize recent progress in the study of mechanisms regulating the activation cycle of β(1) integrin and the specificity of α(5)β(1) integrin in mechanotransduction.
Collapse
Affiliation(s)
- Myriam Régent
- INSERM U823 Institut Albert Bonniot, Université Joseph Fourier, France
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Brütsch R, Liebler SS, Wüstehube J, Bartol A, Herberich SE, Adam MG, Telzerow A, Augustin HG, Fischer A. Integrin Cytoplasmic Domain–Associated Protein-1 Attenuates Sprouting Angiogenesis. Circ Res 2010; 107:592-601. [DOI: 10.1161/circresaha.110.217257] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- René Brütsch
- From the Vascular Biology and Tumor Angiogenesis (R.B., S.S.L., J.W., A.B., S.E.H., M.G.A., A.T., H.G.A., A.F.), Medical Faculty Mannheim (CBTM), Heidelberg University, Mannheim; and Vascular Oncology and Metastasis (S.S.L., J.W., A.B., S.E.H., M.G.A., H.G.A., A.F.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Sven S. Liebler
- From the Vascular Biology and Tumor Angiogenesis (R.B., S.S.L., J.W., A.B., S.E.H., M.G.A., A.T., H.G.A., A.F.), Medical Faculty Mannheim (CBTM), Heidelberg University, Mannheim; and Vascular Oncology and Metastasis (S.S.L., J.W., A.B., S.E.H., M.G.A., H.G.A., A.F.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Joycelyn Wüstehube
- From the Vascular Biology and Tumor Angiogenesis (R.B., S.S.L., J.W., A.B., S.E.H., M.G.A., A.T., H.G.A., A.F.), Medical Faculty Mannheim (CBTM), Heidelberg University, Mannheim; and Vascular Oncology and Metastasis (S.S.L., J.W., A.B., S.E.H., M.G.A., H.G.A., A.F.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Arne Bartol
- From the Vascular Biology and Tumor Angiogenesis (R.B., S.S.L., J.W., A.B., S.E.H., M.G.A., A.T., H.G.A., A.F.), Medical Faculty Mannheim (CBTM), Heidelberg University, Mannheim; and Vascular Oncology and Metastasis (S.S.L., J.W., A.B., S.E.H., M.G.A., H.G.A., A.F.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Stefanie E. Herberich
- From the Vascular Biology and Tumor Angiogenesis (R.B., S.S.L., J.W., A.B., S.E.H., M.G.A., A.T., H.G.A., A.F.), Medical Faculty Mannheim (CBTM), Heidelberg University, Mannheim; and Vascular Oncology and Metastasis (S.S.L., J.W., A.B., S.E.H., M.G.A., H.G.A., A.F.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - M. Gordian Adam
- From the Vascular Biology and Tumor Angiogenesis (R.B., S.S.L., J.W., A.B., S.E.H., M.G.A., A.T., H.G.A., A.F.), Medical Faculty Mannheim (CBTM), Heidelberg University, Mannheim; and Vascular Oncology and Metastasis (S.S.L., J.W., A.B., S.E.H., M.G.A., H.G.A., A.F.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Anja Telzerow
- From the Vascular Biology and Tumor Angiogenesis (R.B., S.S.L., J.W., A.B., S.E.H., M.G.A., A.T., H.G.A., A.F.), Medical Faculty Mannheim (CBTM), Heidelberg University, Mannheim; and Vascular Oncology and Metastasis (S.S.L., J.W., A.B., S.E.H., M.G.A., H.G.A., A.F.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Hellmut G. Augustin
- From the Vascular Biology and Tumor Angiogenesis (R.B., S.S.L., J.W., A.B., S.E.H., M.G.A., A.T., H.G.A., A.F.), Medical Faculty Mannheim (CBTM), Heidelberg University, Mannheim; and Vascular Oncology and Metastasis (S.S.L., J.W., A.B., S.E.H., M.G.A., H.G.A., A.F.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Andreas Fischer
- From the Vascular Biology and Tumor Angiogenesis (R.B., S.S.L., J.W., A.B., S.E.H., M.G.A., A.T., H.G.A., A.F.), Medical Faculty Mannheim (CBTM), Heidelberg University, Mannheim; and Vascular Oncology and Metastasis (S.S.L., J.W., A.B., S.E.H., M.G.A., H.G.A., A.F.), German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| |
Collapse
|
49
|
Faurobert E, Albiges-Rizo C. Recent insights into cerebral cavernous malformations: a complex jigsaw puzzle under construction. FEBS J 2010; 277:1084-96. [PMID: 20096036 DOI: 10.1111/j.1742-4658.2009.07537.x] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cerebral cavernous malformations (CCM) are common vascular malformations with an unpredictable risk of hemorrhage, the consequences of which range from headache to stroke or death. Three genes, CCM1, CCM2 and CCM3, have been linked to the disease. The encoded CCM proteins interact with each other within a large protein complex. Within the past 2 years, a plethora of new data has emerged on the signaling pathways in which CCM proteins are involved. CCM proteins regulate diverse aspects of endothelial cell morphogenesis and blood vessel stability such as cell-cell junctions, cell shape and polarity, or cell adhesion to the extracellular matrix. Although fascinating, a global picture is hard to depict because little is known about how these pathways coordinate to orchestrate angiogenesis. Here we present what is known about the structural domain organization of CCM proteins, their association as a ternary complex and their subcellular localization. Numerous CCM partners have been identified using two-hybrid screens, genetic analyses or proteomic studies. We focus on the best-characterized partners and review data on the signaling pathways they regulate as a step towards a better understanding of the etiology of CCM disease.
Collapse
Affiliation(s)
- Eva Faurobert
- Centre de recherche, INSERM U823-CNRS ERL 3148, Université J. Fourier, Grenoble, France.
| | | |
Collapse
|
50
|
Diamond B, Huerta PT, Mina-Osorio P, Kowal C, Volpe BT. Losing your nerves? Maybe it's the antibodies. Nat Rev Immunol 2009; 9:449-56. [PMID: 19424277 DOI: 10.1038/nri2529] [Citation(s) in RCA: 183] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
We propose that the normal immunocompetent B cell repertoire is replete with B cells making antibodies that recognize brain antigens. Although B cells that are reactive with self antigen are normally silenced during B cell maturation, the blood-brain barrier (BBB) prevents many brain antigens from participating in this process. This enables the generation of a B cell repertoire that is sufficiently diverse to cope with numerous environmental challenges. It requires, however, that the integrity of the BBBs is uninterrupted throughout life to protect the brain from antibodies that crossreact with microorganisms and brain antigens. Under conditions of BBB compromise, and during fetal development, we think that these antibodies can alter brain function in otherwise healthy individuals.
Collapse
Affiliation(s)
- Betty Diamond
- The Feinstein Institute for Medical Research, Center of Autoimmune and Musculoskeletal Diseases, 350 Community Drive, Manhasset, New York 110301, USA.
| | | | | | | | | |
Collapse
|