51
|
Samaržija I, Dekanić A, Humphries JD, Paradžik M, Stojanović N, Humphries MJ, Ambriović-Ristov A. Integrin Crosstalk Contributes to the Complexity of Signalling and Unpredictable Cancer Cell Fates. Cancers (Basel) 2020; 12:E1910. [PMID: 32679769 PMCID: PMC7409212 DOI: 10.3390/cancers12071910] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/10/2020] [Accepted: 07/12/2020] [Indexed: 12/12/2022] Open
Abstract
Integrins are heterodimeric cell surface receptors composed of α and β subunits that control adhesion, proliferation and gene expression. The integrin heterodimer binding to ligand reorganises the cytoskeletal networks and triggers multiple signalling pathways that can cause changes in cell cycle, proliferation, differentiation, survival and motility. In addition, integrins have been identified as targets for many different diseases, including cancer. Integrin crosstalk is a mechanism by which a change in the expression of a certain integrin subunit or the activation of an integrin heterodimer may interfere with the expression and/or activation of other integrin subunit(s) in the very same cell. Here, we review the evidence for integrin crosstalk in a range of cellular systems, with a particular emphasis on cancer. We describe the molecular mechanisms of integrin crosstalk, the effects of cell fate determination, and the contribution of crosstalk to therapeutic outcomes. Our intention is to raise awareness of integrin crosstalk events such that the contribution of the phenomenon can be taken into account when researching the biological or pathophysiological roles of integrins.
Collapse
Affiliation(s)
- Ivana Samaržija
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (I.S.); (M.P.); (N.S.)
| | - Ana Dekanić
- Laboratory for Protein Dynamics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | - Jonathan D. Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, UK; (J.D.H.); (M.J.H.)
| | - Mladen Paradžik
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (I.S.); (M.P.); (N.S.)
| | - Nikolina Stojanović
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (I.S.); (M.P.); (N.S.)
| | - Martin J. Humphries
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine & Health, University of Manchester, Manchester M13 9PT, UK; (J.D.H.); (M.J.H.)
| | - Andreja Ambriović-Ristov
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (I.S.); (M.P.); (N.S.)
| |
Collapse
|
52
|
Jaudon F, Thalhammer A, Cingolani LA. Integrin adhesion in brain assembly: From molecular structure to neuropsychiatric disorders. Eur J Neurosci 2020; 53:3831-3850. [PMID: 32531845 DOI: 10.1111/ejn.14859] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/21/2020] [Accepted: 06/02/2020] [Indexed: 02/07/2023]
Abstract
Integrins are extracellular matrix receptors that mediate biochemical and mechanical bi-directional signals between the extracellular and intracellular environment of a cell thanks to allosteric conformational changes. In the brain, they are found in both neurons and glial cells, where they play essential roles in several aspects of brain development and function, such as cell migration, axon guidance, synaptogenesis, synaptic plasticity and neuro-inflammation. Although there are many successful examples of how regulating integrin adhesion and signaling can be used for therapeutic purposes, for example for halting tumor progression, this is not the case for the brain, where the growing evidence of the importance of integrins for brain pathophysiology has not translated yet into medical applications. Here, we review recent literature showing how alterations in integrin structure, expression and signaling may be involved in the etiology of autism spectrum disorder, epilepsy, schizophrenia, addiction, depression and Alzheimer's disease. We focus on common mechanisms and recurrent signaling pathways, trying to bridge studies on the genetics and molecular structure of integrins with those on synaptic physiology and brain pathology. Further, we discuss integrin-targeting strategies and their potential benefits for therapeutic purposes in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Fanny Jaudon
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Agnes Thalhammer
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Lorenzo A Cingolani
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia (IIT), Genoa, Italy.,Department of Life Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
53
|
Kumar M, Gouw M, Michael S, Sámano-Sánchez H, Pancsa R, Glavina J, Diakogianni A, Valverde JA, Bukirova D, Čalyševa J, Palopoli N, Davey NE, Chemes LB, Gibson TJ. ELM-the eukaryotic linear motif resource in 2020. Nucleic Acids Res 2020; 48:D296-D306. [PMID: 31680160 PMCID: PMC7145657 DOI: 10.1093/nar/gkz1030] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/18/2019] [Accepted: 10/23/2019] [Indexed: 12/20/2022] Open
Abstract
The eukaryotic linear motif (ELM) resource is a repository of manually curated experimentally validated short linear motifs (SLiMs). Since the initial release almost 20 years ago, ELM has become an indispensable resource for the molecular biology community for investigating functional regions in many proteins. In this update, we have added 21 novel motif classes, made major revisions to 12 motif classes and added >400 new instances mostly focused on DNA damage, the cytoskeleton, SH2-binding phosphotyrosine motifs and motif mimicry by pathogenic bacterial effector proteins. The current release of the ELM database contains 289 motif classes and 3523 individual protein motif instances manually curated from 3467 scientific publications. ELM is available at: http://elm.eu.org.
Collapse
Affiliation(s)
- Manjeet Kumar
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Marc Gouw
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Sushama Michael
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Hugo Sámano-Sánchez
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.,Collaboration for Joint PhD Degree between EMBL and Heidelberg University, Faculty of Biosciences
| | - Rita Pancsa
- Institute of Enzymology, Research Centre for Natural Sciences, Budapest 1117, Hungary
| | - Juliana Glavina
- Instituto de Investigaciones Biotecnológicas (IIBio) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de San Martín. Av. 25 de Mayo y Francia, CP1650, Buenos Aires, Argentina
| | - Athina Diakogianni
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Jesús Alvarado Valverde
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Dayana Bukirova
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.,Nazarbayev University, Nur-Sultan 010000, Kazakhstan
| | - Jelena Čalyševa
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.,Collaboration for Joint PhD Degree between EMBL and Heidelberg University, Faculty of Biosciences
| | - Nicolas Palopoli
- Department of Science and Technology, Universidad Nacional de Quilmes - CONICET, Bernal B1876BXD, Buenos Aires, Argentina
| | - Norman E Davey
- The Institute of Cancer Research, Chester Beatty Laboratories, 237 Fulham Rd, Chelsea, London SW3 6JB, UK
| | - Lucía B Chemes
- Instituto de Investigaciones Biotecnológicas (IIBio) and Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de San Martín. Av. 25 de Mayo y Francia, CP1650, Buenos Aires, Argentina
| | - Toby J Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| |
Collapse
|
54
|
Cell matrix adhesion in cell migration. Essays Biochem 2020; 63:535-551. [PMID: 31444228 DOI: 10.1042/ebc20190012] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 07/22/2019] [Accepted: 08/06/2019] [Indexed: 02/06/2023]
Abstract
The ability of cells to migrate is a fundamental physiological process involved in embryonic development, tissue homeostasis, immune surveillance and wound healing. In order for cells to migrate, they must interact with their environment using adhesion receptors, such as integrins, and form specialized adhesion complexes that mediate responses to different extracellular cues. In this review, we discuss the role of integrin adhesion complexes (IACs) in cell migration, highlighting the layers of regulation that are involved, including intracellular signalling cascades, mechanosensing and reciprocal feedback to the extracellular environment. We also discuss the role of IACs in extracellular matrix remodeling and how they impact upon cell migration.
Collapse
|
55
|
Lukácsi S, Gerecsei T, Balázs K, Francz B, Szabó B, Erdei A, Bajtay Z. The differential role of CR3 (CD11b/CD18) and CR4 (CD11c/CD18) in the adherence, migration and podosome formation of human macrophages and dendritic cells under inflammatory conditions. PLoS One 2020; 15:e0232432. [PMID: 32365067 PMCID: PMC7197861 DOI: 10.1371/journal.pone.0232432] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 04/14/2020] [Indexed: 11/19/2022] Open
Abstract
CR3 and CR4, the leukocyte specific β2-integrins, involved in cellular adherence, migration and phagocytosis, are often assumed to have similar functions. Previously however, we proved that under physiological conditions CR4 is dominant in the adhesion to fibrinogen of human monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs). Here, using inflammatory conditions, we provide further evidence that the expression and function of CR3 and CR4 are not identical in these cell types. We found that LPS treatment changes their expression differently on MDMs and MDDCs, suggesting a cell type specific regulation. Using mAb24, specific for the high affinity conformation of CD18, we proved that the activation and recycling of β2-integrins is significantly enhanced upon LPS treatment. Adherence to fibrinogen was assessed by two fundamentally different approaches: a classical adhesion assay and a computer-controlled micropipette, capable of measuring adhesion strength. While both receptors participated in adhesion, we demonstrated that CR4 exerts a dominant role in the strong attachment of MDDCs. Studying the formation of podosomes we found that MDMs retain podosome formation after LPS activation, whereas MDDCs lose this ability, resulting in a significantly reduced adhesion force and an altered cellular distribution of CR3 and CR4. Our results suggest that inflammatory conditions reshape differentially the expression and role of CR3 and CR4 in macrophages and dendritic cells.
Collapse
Affiliation(s)
- Szilvia Lukácsi
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary
| | - Tamás Gerecsei
- Department of Biological Physics, Eötvös Loránd University, Budapest, Hungary
- Nanobiosensorics “Lendület” Group, Institute of Technical Physics and Material Sciences, Centre for Energy Research, Hungarian Academy of Sciences, Budapest, Hungary
| | - Katalin Balázs
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | | | - Bálint Szabó
- Department of Biological Physics, Eötvös Loránd University, Budapest, Hungary
- CellSorter Company for Innovations, Budapest, Hungary
| | - Anna Erdei
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
| | - Zsuzsa Bajtay
- MTA-ELTE Immunology Research Group, Eötvös Loránd University, Budapest, Hungary
- Department of Immunology, Eötvös Loránd University, Budapest, Hungary
- * E-mail:
| |
Collapse
|
56
|
Conformationally active integrin endocytosis and traffic: why, where, when and how? Biochem Soc Trans 2020; 48:83-93. [PMID: 32065228 PMCID: PMC7054750 DOI: 10.1042/bst20190309] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/16/2020] [Accepted: 01/28/2020] [Indexed: 12/30/2022]
Abstract
Spatiotemporal control of integrin-mediated cell adhesion to the extracellular matrix (ECM) is critical for physiological and pathological events in multicellular organisms, such as embryonic development, angiogenesis, platelet aggregation, leukocytes extravasation, and cancer cell metastatic dissemination. Regulation of integrin adhesive function and signaling relies on the modulation of both conformation and traffic. Indeed, integrins exist in a dynamic equilibrium between a bent/closed (inactive) and an extended/open (active) conformation, respectively endowed with low and high affinity for ECM ligands. Increasing evidence proves that, differently to what hypothesized in the past, detachment from the ECM and conformational inactivation are not mandatory for integrin to get endocytosed and trafficked. Specific transmembrane and cytosolic proteins involved in the control of ECM proteolytic fragment-bound active integrin internalization and recycling exist. In the complex masterplan that governs cell behavior, active integrin traffic is key to the turnover of ECM polymers and adhesion sites, the polarized secretion of endogenous ECM proteins and modifying enzymes, the propagation of motility and survival endosomal signals, and the control of cell metabolism.
Collapse
|
57
|
Abstract
Integrins, and integrin-mediated adhesions, have long been recognized to provide the main molecular link attaching cells to the extracellular matrix (ECM) and to serve as bidirectional hubs transmitting signals between cells and their environment. Recent evidence has shown that their combined biochemical and mechanical properties also allow integrins to sense, respond to and interact with ECM of differing properties with exquisite specificity. Here, we review this work first by providing an overview of how integrin function is regulated from both a biochemical and a mechanical perspective, affecting integrin cell-surface availability, binding properties, activation or clustering. Then, we address how this biomechanical regulation allows integrins to respond to different ECM physicochemical properties and signals, such as rigidity, composition and spatial distribution. Finally, we discuss the importance of this sensing for major cell functions by taking cell migration and cancer as examples.
Collapse
|
58
|
The Extracellular Matrix: An Accomplice in Gastric Cancer Development and Progression. Cells 2020; 9:cells9020394. [PMID: 32046329 PMCID: PMC7072625 DOI: 10.3390/cells9020394] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) is a dynamic and highly organized tissue structure, providing support and maintaining normal epithelial architecture. In the last decade, increasing evidence has emerged demonstrating that alterations in ECM composition and assembly strongly affect cellular function and behavior. Even though the detailed mechanisms underlying cell-ECM crosstalk are yet to unravel, it is well established that ECM deregulation accompanies the development of many pathological conditions, such as gastric cancer. Notably, gastric cancer remains a worldwide concern, representing the third most frequent cause of cancer-associated deaths. Despite increased surveillance protocols, patients are usually diagnosed at advanced disease stages, urging the identification of novel diagnostic biomarkers and efficient therapeutic strategies. In this review, we provide a comprehensive overview regarding expression patterns of ECM components and cognate receptors described in normal gastric epithelium, pre-malignant lesions, and gastric carcinomas. Important insights are also discussed for the use of ECM-associated molecules as predictive biomarkers of the disease or as potential targets in gastric cancer.
Collapse
|
59
|
Abstract
Chronic liver diseases, such as fibrosis and cancer, lead to a rigid or stiff liver because of perpetual activation of hepatic stellate cells or portal fibroblasts into matrix-producing myofibroblasts. Mechanical forces, as determined by the mechanical properties of extracellular matrix or pressure of circulating blood flow/shear stress, are sensed by mechanoreceptors at the plasma membrane and transmitted into a cell to impact cell function. This process is termed as mechanotransduction. This review includes basic knowledge regarding how external forces are sensed, amplified, and transmitted into the interior of a cell as far as the nucleus to regulate gene transcription and generate biological responses. It also reviews literatures to highlight the mechanisms by which mechanical forces in a normal or diseased liver influence the phenotype of hepatocytes, hepatic stellate cells, portal fibroblasts, and sinusoidal endothelial cells, and these cells in turn participate in the initiation and progression of liver diseases.
Collapse
Affiliation(s)
- Ningling Kang
- Section of Tumor Microenvironment and Metastasis, Hormel Institute, University of Minnesota, Austin, Minnesota
| |
Collapse
|
60
|
Activation and suppression of hematopoietic integrins in hemostasis and immunity. Blood 2020; 135:7-16. [DOI: 10.1182/blood.2019003336] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022] Open
Abstract
Nolte and Margadant review the current understanding of the activation and inactivation of integrin receptors expressed by hematopoietic cells and the role of these conformational changes in modulating platelet and leukocyte function.
Collapse
|
61
|
Deville SS, Cordes N. The Extracellular, Cellular, and Nuclear Stiffness, a Trinity in the Cancer Resistome-A Review. Front Oncol 2019; 9:1376. [PMID: 31867279 PMCID: PMC6908495 DOI: 10.3389/fonc.2019.01376] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
Alterations in mechano-physiological properties of a tissue instigate cancer burdens in parallel to common genetic and epigenetic alterations. The chronological and mechanistic interrelation between the various extra- and intracellular aspects remains largely elusive. Mechano-physiologically, integrins and other cell adhesion molecules present the main mediators for transferring and distributing forces between cells and the extracellular matrix (ECM). These cues are channeled via focal adhesion proteins, termed the focal adhesomes, to cytoskeleton and nucleus and vice versa thereby affecting the pathophysiology of multicellular cancer tissues. In combination with simultaneous activation of diverse downstream signaling pathways, the phenotypes of cancer cells are created and driven characterized by deregulated transcriptional and biochemical cues that elicit the hallmarks of cancer. It, however, remains unclear how elastostatic modifications, i.e., stiffness, in the extracellular, intracellular, and nuclear compartment contribute and control the resistance of cancer cells to therapy. In this review, we discuss how stiffness of unique tumor components dictates therapy response and what is known about the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Sara Sofia Deville
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Nils Cordes
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- Germany German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
62
|
Evans JJ, Alkaisi MM, Sykes PH. Tumour Initiation: a Discussion on Evidence for a "Load-Trigger" Mechanism. Cell Biochem Biophys 2019; 77:293-308. [PMID: 31598831 PMCID: PMC6841748 DOI: 10.1007/s12013-019-00888-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/23/2019] [Indexed: 12/18/2022]
Abstract
Appropriate mechanical forces on cells are vital for normal cell behaviour and this review discusses the possibility that tumour initiation depends partly on the disruption of the normal physical architecture of the extracellular matrix (ECM) around a cell. The alterations that occur thence promote oncogene expression. Some questions, that are not answered with certainty by current consensus mechanisms of tumourigenesis, are elegantly explained by the triggering of tumours being a property of the physical characteristics of the ECM, which is operative following loading of the tumour initiation process with a relevant gene variant. Clinical observations are consistent with this alternative hypothesis which is derived from studies that have, together, accumulated an extensive variety of data incorporating biochemical, genetic and clinical findings. Thus, this review provides support for the view that the ECM may have an executive function in induction of a tumour. Overall, reported observations suggest that either restoring an ECM associated with homeostasis or targeting the related signal transduction mechanisms may possibly be utilised to modify or control the early progression of cancers. The review provides a coherent template for discussing the notion, in the context of contemporary knowledge, that tumourigenesis is an alliance of biochemistry, genetics and biophysics, in which the physical architecture of the ECM may be a fundamental component. For more definitive clarification of the concept there needs to be a phalanx of experiments conceived around direct questions that are raised by this paper.
Collapse
Affiliation(s)
- John J Evans
- Department of Obstetrics and Gynaecology, University of Otago Christchurch, Christchurch, New Zealand.
- MacDiarmid Institute of Advanced Materials and Nanotechnology, Christchurch, New Zealand.
| | - Maan M Alkaisi
- MacDiarmid Institute of Advanced Materials and Nanotechnology, Christchurch, New Zealand
- Department of Electrical and Computer Engineering, University of Canterbury, Christchurch, New Zealand
| | - Peter H Sykes
- Department of Obstetrics and Gynaecology, University of Otago Christchurch, Christchurch, New Zealand
| |
Collapse
|
63
|
Alam M, Bertolini M, Gherardini J, Keren A, Ponce L, Chéret J, Alenfall J, Dunér P, Nilsson AH, Gilhar A, Paus R. An osteopontin-derived peptide inhibits human hair growth at least in part by decreasing fibroblast growth factor-7 production in outer root sheath keratinocytes. Br J Dermatol 2019; 182:1404-1414. [PMID: 31487385 DOI: 10.1111/bjd.18479] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Given that unwanted hair growth (hirsutism, hypertrichosis) can cause major psychological distress, new pharmacological treatment strategies with safe and effective hair growth inhibitors that do not destroy the hair follicle (HF) and its stem cells need to be developed. OBJECTIVES To establish if osteopontin-derived fragments may modulate human hair growth given that human HFs express the multifunctional, immunomodulatory glycoprotein, osteopontin. METHODS Our hypothesis was tested ex vivo and in vivo by using a newly generated, toxicologically well-characterized, modified osteopontin-derived peptide (FOL-005), which binds to the HF. RESULTS In organ-cultured human HFs and scalp skin, and in human scalp skin xenotransplants onto SCID mice, FOL-005 treatment (60 nmol L-1 to 3 μmol L-1 ) significantly promoted premature catagen development without reducing the number of keratin 15-positive HF stem cells or showing signs of drug toxicity. Genome-wide DNA microarray, quantitative reverse-transcriptase polymerase chain reaction and immunohistochemistry revealed decreased expression of the hair growth promoter, fibroblast growth factor-7 (FGF7) by FOL-005, while cotreatment of HFs with recombinant FGF7 partially abrogated FOL-005-induced catagen promotion. CONCLUSIONS With caveats in mind, our study identifies this osteopontin-derived peptide as an effective, novel inhibitory principle for human hair growth ex vivo and in vivo, which deserves systematic clinical testing in hirsutism and hypertrichosis. What's already known about this topic? The treatment of unwanted hair growth (hypertrichosis, hirsutism) lacks pharmacological intervention, with only few and often unsatisfactory treatments available. Osteopontin is prominently expressed in human HFs and has been reported to be elevated during catagen in the murine hair cycle. What does this study add? We tested the effects on hair growth of a novel, osteopontin-derived fragment (FOL-005) ex vivo and in vivo. In human hair follicles, high-dose FOL-005 significantly reduces hair growth both ex vivo and in vivo. What is the translational message? High-dose FOL-005 may provide a new therapeutic opportunity as a treatment for unwanted hair growth.
Collapse
Affiliation(s)
- M Alam
- Monasterium Laboratory - Skin and Hair Research Solutions GmbH, Münster, Germany.,Mediteknia Skin & Hair Lab, Las Palmas de Gran Canaria, Spain.,Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - M Bertolini
- Monasterium Laboratory - Skin and Hair Research Solutions GmbH, Münster, Germany
| | - J Gherardini
- Monasterium Laboratory - Skin and Hair Research Solutions GmbH, Münster, Germany
| | - A Keren
- Laboratory for Skin Research, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - L Ponce
- Monasterium Laboratory - Skin and Hair Research Solutions GmbH, Münster, Germany
| | - J Chéret
- Monasterium Laboratory - Skin and Hair Research Solutions GmbH, Münster, Germany
| | | | - P Dunér
- Follicum AB, Lund, Sweden.,Department of Clinical Sciences, Lund University, Lund, Sweden
| | - A H Nilsson
- Follicum AB, Lund, Sweden.,Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - A Gilhar
- Laboratory for Skin Research, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - R Paus
- Centre for Dermatology Research, University of Manchester and NIHR Manchester Biomedical Research Centre, Manchester, U.K.,Dr. Philip Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, U.S.A
| |
Collapse
|
64
|
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
|
65
|
Wang Q, Onuma K, Liu C, Wong H, Bloom MS, Elliott EE, Cao RR, Hu N, Lingampalli N, Sharpe O, Zhao X, Sohn DH, Lepus CM, Sokolove J, Mao R, Cisar CT, Raghu H, Chu CR, Giori NJ, Willingham SB, Prohaska SS, Cheng Z, Weissman IL, Robinson WH. Dysregulated integrin αVβ3 and CD47 signaling promotes joint inflammation, cartilage breakdown, and progression of osteoarthritis. JCI Insight 2019; 4:128616. [PMID: 31534047 PMCID: PMC6795293 DOI: 10.1172/jci.insight.128616] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 08/13/2019] [Indexed: 12/13/2022] Open
Abstract
Osteoarthritis (OA) is the leading cause of joint failure, yet the underlying mechanisms remain elusive, and no approved therapies that slow progression exist. Dysregulated integrin function was previously implicated in OA pathogenesis. However, the roles of integrin αVβ3 and the integrin-associated receptor CD47 in OA remain largely unknown. Here, transcriptomic and proteomic analyses of human and murine osteoarthritic tissues revealed dysregulated expression of αVβ3, CD47, and their ligands. Using genetically deficient mice and pharmacologic inhibitors, we showed that αVβ3, CD47, and the downstream signaling molecules Fyn and FAK are crucial to OA pathogenesis. MicroPET/CT imaging of a mouse model showed elevated ligand-binding capacities of integrin αVβ3 and CD47 in osteoarthritic joints. Further, our in vitro studies demonstrated that chondrocyte breakdown products, derived from articular cartilage of individuals with OA, induced αVβ3/CD47-dependent expression of inflammatory and degradative mediators, and revealed the downstream signaling network. Our findings identify a central role for dysregulated αVβ3 and CD47 signaling in OA pathogenesis and suggest that activation of αVβ3 and CD47 signaling in many articular cell types contributes to inflammation and joint destruction in OA. Thus, the data presented here provide a rationale for targeting αVβ3, CD47, and their signaling pathways as a disease-modifying therapy.
Collapse
Affiliation(s)
- Qian Wang
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Kazuhiro Onuma
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Changhao Liu
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, California, USA
| | - Heidi Wong
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Michelle S. Bloom
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Eileen E. Elliott
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Richard R.L. Cao
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Nick Hu
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Nithya Lingampalli
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Orr Sharpe
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Xiaoyan Zhao
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Dong Hyun Sohn
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Microbiology and Immunology, Pusan National University School of Medicine, Yangsan, Gyeongsangnam-do, South Korea
| | - Christin M. Lepus
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Jeremy Sokolove
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Rong Mao
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Cecilia T. Cisar
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Harini Raghu
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Constance R. Chu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Orthopedic Surgery
| | - Nicholas J. Giori
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
- Department of Orthopedic Surgery
| | - Stephen B. Willingham
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, and
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Susan S. Prohaska
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, and
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - Zhen Cheng
- Molecular Imaging Program at Stanford (MIPS), Canary Center at Stanford for Cancer Early Detection, Department of Radiology and Bio-X Program, Stanford University School of Medicine, Stanford, California, USA
| | - Irving L. Weissman
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer Center, and
- Departments of Pathology and Developmental Biology, Stanford University School of Medicine, Stanford, California, USA
| | - William H. Robinson
- Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
66
|
Jayadev R, Chi Q, Keeley DP, Hastie EL, Kelley LC, Sherwood DR. α-Integrins dictate distinct modes of type IV collagen recruitment to basement membranes. J Cell Biol 2019; 218:3098-3116. [PMID: 31387941 PMCID: PMC6719451 DOI: 10.1083/jcb.201903124] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/16/2019] [Accepted: 07/10/2019] [Indexed: 01/04/2023] Open
Abstract
Basement membranes (BMs) are cell-associated extracellular matrices that support tissue integrity, signaling, and barrier properties. Type IV collagen is critical for BM function, yet how it is directed into BMs in vivo is unclear. Through live-cell imaging of endogenous localization, conditional knockdown, and misexpression experiments, we uncovered distinct mechanisms of integrin-mediated collagen recruitment to Caenorhabditis elegans postembryonic gonadal and pharyngeal BMs. The putative laminin-binding αINA-1/βPAT-3 integrin was selectively activated in the gonad and recruited laminin, which directed moderate collagen incorporation. In contrast, the putative Arg-Gly-Asp (RGD)-binding αPAT-2/βPAT-3 integrin was activated in the pharynx and recruited high levels of collagen in an apparently laminin-independent manner. Through an RNAi screen, we further identified the small GTPase RAP-3 (Rap1) as a pharyngeal-specific PAT-2/PAT-3 activator that modulates collagen levels. Together, these studies demonstrate that tissues can use distinct mechanisms to direct collagen incorporation into BMs to precisely control collagen levels and construct diverse BMs.
Collapse
Affiliation(s)
- Ranjay Jayadev
- Department of Biology, Regeneration Next, Duke University, Durham, NC
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| | - Qiuyi Chi
- Department of Biology, Regeneration Next, Duke University, Durham, NC
| | - Daniel P Keeley
- Department of Biology, Regeneration Next, Duke University, Durham, NC
| | - Eric L Hastie
- Department of Biology, Regeneration Next, Duke University, Durham, NC
| | - Laura C Kelley
- Department of Biology, Regeneration Next, Duke University, Durham, NC
| | - David R Sherwood
- Department of Biology, Regeneration Next, Duke University, Durham, NC
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC
| |
Collapse
|
67
|
Garcia-Keller C, Neuhofer D, Bobadilla AC, Spencer S, Chioma VC, Monforton C, Kalivas PW. Extracellular Matrix Signaling Through β3 Integrin Mediates Cocaine Cue-Induced Transient Synaptic Plasticity and Relapse. Biol Psychiatry 2019; 86:377-387. [PMID: 31126696 PMCID: PMC6697624 DOI: 10.1016/j.biopsych.2019.03.982] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/05/2019] [Accepted: 03/25/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND Cue-induced relapse to drug use is a primary symptom of cocaine addiction. Cue-induced transient excitatory synaptic potentiation (t-SP) induced in the nucleus accumbens mediates cued cocaine seeking in rat models of relapse. Cue-induced t-SP depends on extracellular signaling by matrix metalloproteases (MMPs), but it is unknown how this catalytic activity communicates with nucleus accumbens neurons to induce t-SP and cocaine seeking. METHODS Male Sprague Dawley rats (N = 125) were trained to self-administer cocaine, after which self-administration was extinguished and then reinstated by cocaine-conditioned cues. We used a morpholino antisense strategy to knock down the β1 or β3 integrin subunits or inhibitors to prevent phosphorylation of the integrin signaling kinases focal adhesion kinase (FAK) or integrin-linked kinase. We quantified protein changes with immunoblotting and t-SP by measuring dendritic spine morphology and alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid/N-methyl-D-aspartate glutamate currents. Integrin signaling was stimulated by microinjecting an MMP activator or integrin peptide ligand into the accumbens. RESULTS Knockdown of β3 integrin or FAK inhibitor, but not β1 integrin or integrin-linked kinase inhibitor, prevented cue-induced cocaine seeking but not sucrose seeking. β3 integrin knockdown prevented t-SP as measured by preventing the cue-induced increases in both alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid/N-methyl-D-aspartate glutamate ratio and spine head diameter. Activating MMP gelatinases with tissue plasminogen activator potentiated cue-induced reinstatement, which was prevented by β3 integrin knockdown and FAK inhibition. Stimulating integrin receptors with the RGD ligand liberated by MMP gelatinase activity also potentiated cued cocaine seeking. CONCLUSIONS Activation of MMP gelatinase in the extracellular space is necessary for and potentiates cued cocaine seeking. This extracellular catalysis stimulates β3 integrins and activates FAK to induce t-SP and promote cue-induced cocaine seeking.
Collapse
Affiliation(s)
- Constanza Garcia-Keller
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina.
| | - Daniela Neuhofer
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Ana-Clara Bobadilla
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Sade Spencer
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota
| | - Vivian C Chioma
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Cara Monforton
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
68
|
Gao J, Bao Y, Ge S, Sun P, Sun J, Liu J, Chen F, Han L, Cao Z, Qin J, White GC, Xu Z, Ma YQ. Sharpin suppresses β1-integrin activation by complexing with the β1 tail and kindlin-1. Cell Commun Signal 2019; 17:101. [PMID: 31429758 PMCID: PMC6700787 DOI: 10.1186/s12964-019-0407-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 07/28/2019] [Indexed: 12/30/2022] Open
Abstract
Background Previously sharpin has been identified as an endogenous inhibitor of β1-integrin activation by directly binding to a conserved region in the cytoplasmic tails (CTs) of the integrin β1-associated α subunits. Methods Here we employed biochemical approaches and cellular analyses to evaluate the function and molecular mechanism of the sharpin-kindlin-1 complex in regulating β1-integrin activation. Results In this study, we found that although the inhibition of sharpin on β1-integrin activation could be confirmed, sharpin had no apparent effect on integrin αIIbβ3 activation in CHO cell system. Notably, a direct interaction between sharpin and the integrin β1 CT was detected, while the interaction of sharpin with the integrin αIIb and the β3 CTs were substantially weaker. Importantly, sharpin was able to inhibit the talin head domain binding to the integrin β1 CT, which can mechanistically contribute to inhibiting β1-integrin activation. Interestingly, we also found that sharpin interacted with kindlin-1, and the interaction between sharpin and the integrin β1 CT was significantly enhanced when kindlin-1 was present. Consistently, we observed that instead of acting as an activator, kindlin-1 actually suppressed the talin head domain mediated β1-integrin activation, indicating that kindlin-1 may facilitate recruitment of sharpin to the integrin β1 CT. Conclusion Taken together, our findings suggest that sharpin may complex with both kindlin-1 and the integrin β1 CT to restrict the talin head domain binding, thus inhibiting β1-integrin activation.
Collapse
Affiliation(s)
- Juan Gao
- Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China
| | - Yun Bao
- Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China
| | - Shushu Ge
- Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China
| | - Peisen Sun
- Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China
| | - Jiaojiao Sun
- Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China
| | - Jianmin Liu
- Department of Molecular Cardiology, Lerner Research Institute Cleveland Clinic, Cleveland, OH, USA
| | - Feng Chen
- Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China
| | - Li Han
- Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China
| | - Zhongyuan Cao
- Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China
| | - Jun Qin
- Department of Molecular Cardiology, Lerner Research Institute Cleveland Clinic, Cleveland, OH, USA
| | - Gilbert C White
- Blood Research Institute, Versiti, 8727 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Biochemistry, Medical College of Milwaukee, Milwaukee, WI, USA
| | - Zhen Xu
- Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China. .,Blood Research Institute, Versiti, 8727 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Yan-Qing Ma
- Collaborative Research Program for Cell Adhesion Molecules, Shanghai University School of Life Sciences, Shanghai, China. .,Blood Research Institute, Versiti, 8727 Watertown Plank Road, Milwaukee, WI, 53226, USA. .,Department of Biochemistry, Medical College of Milwaukee, Milwaukee, WI, USA.
| |
Collapse
|
69
|
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: 4.0] [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
|
70
|
CKAP4 Regulates Cell Migration via the Interaction with and Recycling of Integrin. Mol Cell Biol 2019; 39:MCB.00073-19. [PMID: 31160493 DOI: 10.1128/mcb.00073-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 05/28/2019] [Indexed: 02/06/2023] Open
Abstract
Cytoskeleton-associated protein 4 (CKAP4) is an endoplasmic reticulum protein that is also present in the cell surface membrane, where it acts as a receptor for Dickkopf1 (DKK1). In this study, we found that CKAP4 interacts with β1 integrin and controls the recycling of α5β1 integrin independently of DKK1. In S2-CP8 cells, knockdown of CKAP4 but not DKK1 enlarged the size of cell adhesion sites and enhanced cell adhesion to fibronectin, resulting in decreased cell migration. When CKAP4 was depleted, the levels of α5 but not β1 integrin were increased in the cell surface membrane. A similar phenotype was observed in other cells expressing low levels of DKK1. In S2-CP8 cells, α5 integrin was trafficked with β1 integrin and CKAP4 to the lysosome or recycled with β1 integrin. In CKAP4-depleted cells, the internalization of α5β1 integrin was unchanged, but its recycling was upregulated. Knockdown of sorting nexin 17 (SNX17), a mediator of integrin recycling, abrogated the increased α5 integrin levels caused by CKAP4 knockdown. CKAP4 bound to SNX17, and its knockdown enhanced the recruitment of α5β1 integrin to SNX17. These results suggest that CKAP4 suppresses the recycling of α5β1 integrin and coordinates cell adhesion sites and migration independently of DKK1.
Collapse
|
71
|
Siitonen R, Peuhu E, Autio A, Liljenbäck H, Mattila E, Metsälä O, Käkelä M, Saanijoki T, Dijkgraaf I, Jalkanen S, Ivaska J, Roivainen A. 68Ga-DOTA-E[c(RGDfK)] 2 PET Imaging of SHARPIN-Regulated Integrin Activity in Mice. J Nucl Med 2019; 60:1380-1387. [PMID: 30850498 DOI: 10.2967/jnumed.118.222026] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 03/04/2019] [Indexed: 12/26/2022] Open
Abstract
Shank-associated RH domain-interacting protein (SHARPIN) is a cytosolic protein that plays a key role in activation of nuclear factor κ-light-chain enhancer of activated B cells and regulation of inflammation. Furthermore, SHARPIN controls integrin-dependent cell adhesion and migration in several normal and malignant cell types, and loss of SHARPIN correlates with increased integrin activity in mice. Arginyl-glycyl-aspartic acid (RGD), a cell adhesion tripeptide motif, is an integrin recognition sequence that facilitates PET imaging of integrin upregulation during tumor angiogenesis. We hypothesized that increased integrin activity due to loss of SHARPIN protein would affect the uptake of αvβ3-selective cyclic, dimeric peptide 68Ga-DOTA-E[c(RGDfK)]2, where E[c(RGDfk)]2 = glutamic acid-[cyclo(arginyl-glycyl-aspartic acid-D-phenylalanine-lysine)], both in several tissue types and in the tumor microenvironment. To test this hypothesis, we used RGD-based in vivo PET imaging to evaluate wild-type (wt) and SHARPIN-deficient mice (Sharpin cpdm , where cpdm = chronic proliferative dermatitis in mice) with and without melanoma tumor allografts. Methods: Sharpin cpdm mice with spontaneous null mutation in the Sharpin gene and their wt littermates with or without B16-F10-luc melanoma tumors were studied by in vivo imaging and ex vivo measurements with cyclic-RGD peptide 68Ga-DOTA-E[c(RGDfK)]2 After the last 68Ga-DOTA-E[c(RGDfK)]2 peptide PET/CT, tumors were cut into cryosections for autoradiography, histology, and immunohistochemistry. Results: The ex vivo uptake of 68Ga-DOTA-E[c(RGDfK)]2 in the mouse skin and tumor was significantly higher in Sharpin cpdm mice than in wt mice. B16-F10-luc tumors were detected 4 d after inoculation, without differences in volume or blood flow between the mouse strains. PET imaging with 68Ga-DOTA-E[c(RGDfK)]2 peptide at day 10 after inoculation revealed significantly higher uptake in the tumors transplanted into Sharpin cpdm mice than in wt mice. Furthermore, tumor vascularization was increased in the Sharpin cpdm mice. Conclusion: Sharpin cpdm mice demonstrated increased integrin activity and vascularization in B16-F10-luc melanoma tumors, as demonstrated by RGD-based in vivo PET imaging. These data indicate that SHARPIN, a protein previously associated with increased cancer growth and metastasis, may also have important regulatory roles in controlling the tumor microenvironment.
Collapse
Affiliation(s)
| | - Emilia Peuhu
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.,FICAN West Cancer Research Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Anu Autio
- Turku PET Centre, University of Turku, Turku, Finland
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, Turku, Finland.,Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Elina Mattila
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Olli Metsälä
- Turku PET Centre, University of Turku, Turku, Finland
| | - Meeri Käkelä
- Turku PET Centre, University of Turku, Turku, Finland
| | | | - Ingrid Dijkgraaf
- Department of Biochemistry, University of Maastricht, Maastricht, the Netherlands
| | - Sirpa Jalkanen
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.,Department of Biochemistry, University of Turku, Turku, Finland; and
| | - Anne Roivainen
- Turku PET Centre, University of Turku, Turku, Finland .,Turku Center for Disease Modeling, University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital, Turku, Finland
| |
Collapse
|
72
|
Moreno-Layseca P, Icha J, Hamidi H, Ivaska J. Integrin trafficking in cells and tissues. Nat Cell Biol 2019; 21:122-132. [PMID: 30602723 PMCID: PMC6597357 DOI: 10.1038/s41556-018-0223-z] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/25/2018] [Indexed: 12/28/2022]
Abstract
Cell adhesion to the extracellular matrix is fundamental to metazoan multicellularity and is accomplished primarily through the integrin family of cell-surface receptors. Integrins are internalized and enter the endocytic-exocytic pathway before being recycled back to the plasma membrane. The trafficking of this extensive protein family is regulated in multiple context-dependent ways to modulate integrin function in the cell. Here, we discuss recent advances in understanding the mechanisms and cellular roles of integrin endocytic trafficking.
Collapse
Affiliation(s)
- Paulina Moreno-Layseca
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Jaroslav Icha
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Hellyeh Hamidi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.
- Department of Biochemistry, University of Turku, Turku, Finland.
| |
Collapse
|
73
|
Kim DH, Lu Q, Chen YH. Claudin-7 modulates cell-matrix adhesion that controls cell migration, invasion and attachment of human HCC827 lung cancer cells. Oncol Lett 2019; 17:2890-2896. [PMID: 30854065 PMCID: PMC6365970 DOI: 10.3892/ol.2019.9909] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 12/31/2018] [Indexed: 11/12/2022] Open
Abstract
Claudins are a family of tight junction proteins, and serve important roles in epithelial barrier, selective ion transports and cancer metastasis. Although the exact role of claudin-7 in human lung cancer has not been completely elucidated, recent clinical studies have demonstrated that claudin-7 is associated with the survival of patients with lung cancer. Our previous studies have demonstrated that claudin-7 forms a protein complex with integrin β1 in human lung cancer cells. The knockdown (KD) of claudin-7 by short hairpin RNA (shRNA) reduced integrin β1 expression and increased the cell proliferative rate, whereas claudin-7 re-expression in the KD cells decreased the cell proliferation. It is unknown as to whether claudin-7 and integrin β1 regulate cell proliferation and invasion synergistically or independently. In the present study, it was observed that ectopic expression of integrin β1 in claudin-7 KD lung cancer cells did not reduce the cell proliferation. However, integrin β1-transfected cells migrated more effectively in wound healing and cell invasion assays and were more adhesive in a cell attachment assay when compared with those of claudin-7 KD cells. This indicates that claudin-7 controls cell proliferation, while cell attachment and motility were regulated partially through integrin β1. Additionally, claudin-7 overexpression in claudin-7 KD cells resulted in an improved ability to attach to the surface of cell culture plates and a higher expression of focal adhesion proteins when compared with claudin-7 non-KD control cells, which supports the role of claudin-7 in cell adhesion and motility. Taken together, these data suggest that claudin-7 regulates cell motility through integrin β1, providing additional insight into the roles of claudins in carcinogenesis and cancer cell metastasis.
Collapse
Affiliation(s)
- Do Hyung Kim
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Qun Lu
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.,Leo Jenkins Cancer Center, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Yan-Hua Chen
- Department of Anatomy and Cell Biology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.,Leo Jenkins Cancer Center, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
74
|
Integrin activation by talin, kindlin and mechanical forces. Nat Cell Biol 2019; 21:25-31. [PMID: 30602766 DOI: 10.1038/s41556-018-0234-9] [Citation(s) in RCA: 319] [Impact Index Per Article: 63.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 10/16/2018] [Indexed: 01/15/2023]
Abstract
Integrins are the major family of adhesion molecules that mediate cell adhesion to the extracellular matrix. They are essential for embryonic development and influence numerous diseases, including inflammation, cancer cell invasion and metastasis. In this Perspective, we discuss the current understanding of how talin, kindlin and mechanical forces regulate integrin affinity and avidity, and how integrin inactivators function in this framework.
Collapse
|
75
|
The extracellular matrix-myosin pathway in mechanotransduction: from molecule to tissue. Emerg Top Life Sci 2018; 2:727-737. [PMID: 33530663 PMCID: PMC7289002 DOI: 10.1042/etls20180043] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/26/2018] [Accepted: 09/28/2018] [Indexed: 12/23/2022]
Abstract
Mechanotransduction via the extracellular matrix (ECM)–myosin pathway is involved in determining cell morphology during development and in coupling external transient mechanical stimuli to the reorganization of the cytoskeleton. Here, we present a review on the molecular mechanisms involved in this pathway and how they influence cellular development and organization. We investigate key proteins involved in the ECM–myosin pathway and discuss how specific binding events and conformational changes under force are related to mechanical signaling. We connect these molecular mechanisms with observed morphological changes at the cellular and organism level. Finally, we propose a model encompassing the biomechanical signals along the ECM–myosin pathway and how it could be involved in cell adhesion, cell migration, and tissue architecture.
Collapse
|
76
|
Andriu A, Crockett J, Dall'Angelo S, Piras M, Zanda M, Fleming IN. Binding of α vβ 3 Integrin-Specific Radiotracers Is Modulated by Both Integrin Expression Level and Activation Status. Mol Imaging Biol 2018; 20:27-36. [PMID: 28695371 PMCID: PMC5775384 DOI: 10.1007/s11307-017-1100-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE Molecular imaging of αvβ3 integrin has exhibited real potential to guide the appropriate use of anti-angiogenic therapies. However, an incomplete understanding of the factors that influence binding of αvβ3 integrin-specific radiotracers currently limits their use for assessing response to therapy in cancer patients. This study identifies two fundamental factors that modulate uptake of these radiotracers. Procedures Experiments were performed in prostate cancer (PC3) and glioblastoma (U87MG) cells, which differentially express αvβ3 integrin. αvβ3 integrin-specific radiotracers were used to investigate the effect of manipulating αvβ3 integrin expression or activation in cellular binding assays. β3 integrin and αvβ3 integrin expression were measured by western blotting and flow cytometry, respectively. The effect of select pharmacological inhibitors on αvβ3 integrin activation and expression was also determined. RESULTS Radiotracer binding was proportional to αvβ3 integrin expression when it was decreased (β3 knock-down cells) or increased, either using pharmacological inhibitors of cell signalling or by culturing cells for different times. Studies with both small molecule and arginine-glycine-aspartic acid (RGD)-based radiotracers revealed increased radiotracer binding after activation of αvβ3 integrin with Mn2+ or talin head domain. Moreover, inhibition of fundamental signalling pathways (mitogen-activated protein kinase kinase (MEK), Src and VEGFR2) decreased radiotracer binding, reflecting reduced αvβ3 integrin activity. CONCLUSION Binding of small molecule ligands and radiolabelled RGD peptides is modulated by expression and activation status of αvβ3 integrin. αvβ3 integrin-specific radiotracers can provide otherwise inaccessible information of the effect of signalling pathways on αvβ3 integrin. This has significant implications for assessing response to anti-angiogenic therapies in clinical studies.
Collapse
Affiliation(s)
- Alexandra Andriu
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Julie Crockett
- Arthritis and Musculoskeletal Medicine Research Programme, Division of Applied Medicine, Institute of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Sergio Dall'Angelo
- Kosterlitz Centre for Therapeutics, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, AB25 2ZD, UK
| | - Monica Piras
- Kosterlitz Centre for Therapeutics, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, AB25 2ZD, UK
| | - Matteo Zanda
- Kosterlitz Centre for Therapeutics, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, AB25 2ZD, UK
| | - Ian N Fleming
- Aberdeen Biomedical Imaging Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
77
|
Kawamura M, Yamamoto T, Yamashiro K, Kochi S, Yoshihara-Hirata C, Ideguchi H, Aoyagi H, Omori K, Takashiba S. Induction of migration of periodontal ligament cells by selective regulation of integrin subunits. J Cell Mol Med 2018; 23:1211-1223. [PMID: 30511442 PMCID: PMC6349235 DOI: 10.1111/jcmm.14023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 08/06/2018] [Accepted: 10/20/2018] [Indexed: 01/09/2023] Open
Abstract
The recruitment of tissue-resident stem cells is important for wound regeneration. Periodontal ligament cells (PDL cells) are heterogeneous cell populations with stemness features that migrate into wound sites to regenerate periodontal fibres and neighbouring hard tissues. Cell migration is regulated by the local microenvironment, coordinated by growth factors and the extracellular matrix (ECM). Integrin-mediated cell adhesion to the ECM provides essential signals for migration. We hypothesized that PDL cell migration could be enhanced by selective expression of integrins. The migration of primary cultured PDL cells was induced by platelet-derived growth factor-BB (PDGF-BB). The effects of blocking specific integrins on migration and ECM adhesion were investigated based on the integrin expression profiles observed during migration. Up-regulation of integrins α3, α5, and fibronectin was identified at distinct localizations in migrating PDL cells. Treatment with anti-integrin α5 antibodies inhibited PDL cell migration. Treatment with anti-integrin α3, α3-blocking peptide, and α3 siRNA significantly enhanced cell migration, comparable to treatment with PDGF-BB. Furthermore, integrin α3 inhibition preferentially enhanced adhesion to fibronectin via integrin α5. These findings indicate that PDL cell migration is reciprocally regulated by integrin α3-mediated inhibition and α5-mediated promotion. Thus, targeting integrin expression is a possible therapeutic strategy for periodontal regeneration.
Collapse
Affiliation(s)
- Mari Kawamura
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tadashi Yamamoto
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Keisuke Yamashiro
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shinsuke Kochi
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Chiaki Yoshihara-Hirata
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hidetaka Ideguchi
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroaki Aoyagi
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kazuhiro Omori
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shogo Takashiba
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
78
|
Tseng HY, Samarelli AV, Kammerer P, Scholze S, Ziegler T, Immler R, Zent R, Sperandio M, Sanders CR, Fässler R, Böttcher RT. LCP1 preferentially binds clasped αMβ2 integrin and attenuates leukocyte adhesion under flow. J Cell Sci 2018; 131:jcs.218214. [PMID: 30333137 DOI: 10.1242/jcs.218214] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Integrins are α/β heterodimers that interconvert between inactive and active states. In the active state the α/β cytoplasmic domains recruit integrin-activating proteins and separate the transmembrane and cytoplasmic (TMcyto) domains (unclasped TMcyto). Conversely, in the inactive state the α/β TMcyto domains bind integrin-inactivating proteins, resulting in the association of the TMcyto domains (clasped TMcyto). Here, we report the isolation of integrin cytoplasmic tail interactors using either lipid bicelle-incorporated integrin TMcyto domains (α5, αM, αIIb, β1, β2 and β3 integrin TMcyto) or a clasped, lipid bicelle-incorporated αMβ2 TMcyto. Among the proteins found to preferentially bind clasped rather than the isolated αM and β2 subunits was L-plastin (LCP1, also known as plastin-2), which binds to and maintains the inactive state of αMβ2 integrin in vivo and thereby regulates leukocyte adhesion to integrin ligands under flow. Our findings offer a global view on cytoplasmic proteins interacting with different integrins and provide evidence for the existence of conformation-specific integrin interactors.
Collapse
Affiliation(s)
- Hui-Yuan Tseng
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Anna V Samarelli
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Patricia Kammerer
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Sarah Scholze
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Tilman Ziegler
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany
| | - Roland Immler
- Walter Brendel Center for Experimental Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Roy Zent
- Division of Nephrology, Department of Medicine, Vanderbilt University, Nashville, 37232 Tennessee, USA.,Department of Medicine, Veterans Affairs Medical Center, Nashville, 37232 Tennessee, USA
| | - Markus Sperandio
- Walter Brendel Center for Experimental Medicine, Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Charles R Sanders
- Department of Biochemistry, Center for Structural Biology, and Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, 37232 Tennessee, USA
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| | - Ralph T Böttcher
- Department of Molecular Medicine, Max Planck Institute for Biochemistry, 82152 Martinsried, Germany .,DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, 80802 Munich, Germany
| |
Collapse
|
79
|
Cancer-associated fibroblast (CAF)-derived IL32 promotes breast cancer cell invasion and metastasis via integrin β3-p38 MAPK signalling. Cancer Lett 2018; 442:320-332. [PMID: 30391782 DOI: 10.1016/j.canlet.2018.10.015] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/30/2018] [Accepted: 10/22/2018] [Indexed: 12/28/2022]
Abstract
Metastasis is the leading cause of breast cancer-related deaths. Cancer-associated fibroblasts (CAFs), the predominant stromal cell type in the breast tumour microenvironment, may contribute to cancer progression through interaction with tumour cells. Nonetheless, little is known about the details of the underlying mechanism. Here we found that interaction of interleukin 32 (IL32) with integrin β3 (encoded by ITGB3; a member of the integrin family) mediating the cross-talk between CAFs and breast cancer cells plays a crucial role in CAF-induced breast tumour invasiveness. IL32, an 'RGD' motif-containing cytokine, was found to be abundantly expressed in CAFs. Integrin β3 turned out to be up-regulated in breast cancer cells during epithelial-mesenchymal transition (EMT). CAF-derived IL32 specifically bound to integrin β3 through the RGD motif, thus activating intracellular downstream p38 MAPK signalling in breast cancer cells. This signalling increased the expression of EMT markers (fibronectin, N-cadherin, and vimentin) and promoted tumour cell invasion. Counteracting IL32 activity, a knockdown of IL32 or integrin β3 led to specific inactivation of p38 MAPK signalling in tumour cells. Blockage of the p38 MAPK pathway also diminished IL32-induced expression of EMT markers and breast cancer cell invasion and metastasis. Thus, our data indicate that CAF-secreted IL32 promotes breast cancer cell invasion and metastasis via integrin β3-p38 MAPK signalling.
Collapse
|
80
|
Hozumi K, Nomizu M. Mixed Peptide-Conjugated Chitosan Matrices as Multi-Receptor Targeted Cell-Adhesive Scaffolds. Int J Mol Sci 2018; 19:E2713. [PMID: 30208645 PMCID: PMC6165449 DOI: 10.3390/ijms19092713] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 08/28/2018] [Accepted: 09/07/2018] [Indexed: 01/05/2023] Open
Abstract
Biomaterials are important for cell and tissue engineering. Chitosan is widely used as a scaffold because it is easily modified using its amino groups, can easily form a matrix, is stable under physiological conditions, and is inactive for cell adhesion. Chitosan is an excellent platform for peptide ligands, especially cell adhesive peptides derived from extracellular matrix (ECM) proteins. ECM proteins, such as collagen, fibronectin, and laminin, are multifunctional and have diverse cell attachment sites. Various cell adhesive peptides have been identified from the ECM proteins, and these are useful to design functional biomaterials. The cell attachment activity of peptides is influenced by the solubility, conformation, and coating efficiency to solid materials, whereas immobilization of peptides to a polysaccharide such as chitosan avoids these problems. Peptide⁻chitosan matrices promote various biological activities depending on the peptide. When the peptides are immobilized to chitosan, the activity of the peptides is significantly enhanced. Further, mixed peptide⁻chitosan matrices, conjugated with more than one peptide on a chitosan matrix, interact with multiple cellular receptors and promote specific biological responses via receptor cross-talk. Receptor cross-talk is important for mimicking the biological activity of ECM and the proteins. The mixed peptide⁻chitosan matrix approach is useful to develop biomaterials as a synthetic ECM for cell and tissue engineering.
Collapse
Affiliation(s)
- Kentaro Hozumi
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan.
- Department of Applied Clinical Dietetics, Kitasato Junior College of Health and Hygienic Sciences, Minamiuonuma, Niigata 949-7241, Japan.
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan.
| |
Collapse
|
81
|
Abstract
Cell adhesion to the extracellular matrix is fundamental to tissue integrity and human health. Integrins are the main cellular adhesion receptors that through multifaceted roles as signalling molecules, mechanotransducers and key components of the cell migration machinery are implicated in nearly every step of cancer progression from primary tumour development to metastasis. Altered integrin expression is frequently detected in tumours, where integrins have roles in supporting oncogenic growth factor receptor (GFR) signalling and GFR-dependent cancer cell migration and invasion. In addition, integrins determine colonization of metastatic sites and facilitate anchorage-independent survival of circulating tumour cells. Investigations describing integrin engagement with a growing number of versatile cell surface molecules, including channels, receptors and secreted proteins, continue to lead to the identification of novel tumour-promoting pathways. Integrin-mediated sensing, stiffening and remodelling of the tumour stroma are key steps in cancer progression supporting invasion, acquisition of cancer stem cell characteristics and drug resistance. Given the complexity of integrins and their adaptable and sometimes antagonistic roles in cancer cells and the tumour microenvironment, therapeutic targeting of these receptors has been a challenge. However, novel approaches to target integrins and antagonism of specific integrin subunits in stringently stratified patient cohorts are emerging as potential ways forward.
Collapse
Affiliation(s)
- Hellyeh Hamidi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, Turku, Finland.
- Department of Biochemistry, University of Turku, Turku, Finland.
| |
Collapse
|
82
|
De Franceschi N, Miihkinen M, Hamidi H, Alanko J, Mai A, Picas L, Guzmán C, Lévy D, Mattjus P, Goult BT, Goud B, Ivaska J. ProLIF - quantitative integrin protein-protein interactions and synergistic membrane effects on proteoliposomes. J Cell Sci 2018; 132:jcs.214270. [PMID: 30072441 DOI: 10.1242/jcs.214270] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 07/07/2018] [Indexed: 01/23/2023] Open
Abstract
Integrin transmembrane receptors control a wide range of biological interactions by triggering the assembly of large multiprotein complexes at their cytoplasmic interface. Diverse methods have been used to investigate interactions between integrins and intracellular proteins, and predominantly include peptide-based pulldowns and biochemical immuno-isolations from detergent-solubilised cell lysates. However, quantitative methods to probe integrin-protein interactions in a more biologically relevant context where the integrin is embedded within a lipid bilayer have been lacking. Here, we describe 'protein-liposome interactions by flow cytometry' (denoted ProLIF), a technique to reconstitute recombinant integrin transmembrane domains (TMDs) and cytoplasmic tail (CT) fragments in liposomes as individual subunits or as αβ heterodimers and, via flow cytometry, allow rapid and quantitative measurement of protein interactions with these membrane-embedded integrins. Importantly, the assay can analyse binding of fluorescent proteins directly from cell lysates without further purification steps. Moreover, the effect of membrane composition, such as PI(4,5)P2 incorporation, on protein recruitment to the integrin CTs can be analysed. ProLIF requires no specific instrumentation and can be applied to measure a broad range of membrane-dependent protein-protein interactions with the potential for high-throughput/multiplex analyses.This article has associated First Person interviews with the first authors of the paper (see doi: 10.1242/jcs.223644 and doi: 10.1242/jcs.223719).
Collapse
Affiliation(s)
- Nicola De Franceschi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland.,Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR 168, 75005 Paris, France.,Sorbonne Universités, UPMC, 75005 Paris, France
| | - Mitro Miihkinen
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Hellyeh Hamidi
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Jonna Alanko
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Anja Mai
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Laura Picas
- Institut Curie, PSL Research University, UMR 168, Centre de Recherche, 75248 Paris, France
| | - Camilo Guzmán
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland
| | - Daniel Lévy
- Institut Curie, PSL Research University, UMR 168, Centre de Recherche, 75248 Paris, France
| | - Peter Mattjus
- Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, UK
| | - Bruno Goud
- Institut Curie, PSL Research University, UMR 168, Centre de Recherche, 75248 Paris, France
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, 20520 Turku, Finland .,Department of Biochemistry, University of Turku, 20520 Turku, Finland
| |
Collapse
|
83
|
Jahan F, Madhavan S, Rolova T, Viazmina L, Grönholm M, Gahmberg CG. Phosphorylation of the α-chain in the integrin LFA-1 enables β2-chain phosphorylation and α-actinin binding required for cell adhesion. J Biol Chem 2018; 293:12318-12330. [PMID: 29903913 DOI: 10.1074/jbc.ra118.004318] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 06/11/2018] [Indexed: 11/06/2022] Open
Abstract
The integrin leukocyte function-associated antigen-1 (LFA-1) plays a pivotal role in leukocyte adhesion and migration, but the mechanism(s) by which this integrin is regulated has remained incompletely understood. LFA-1 integrin activity requires phosphorylation of its β2-chain and interactions of its cytoplasmic tail with various cellular proteins. The α-chain is constitutively phosphorylated and necessary for cellular adhesion, but how the α-chain regulates adhesion has remained enigmatic. We now show that substitution of the α-chain phosphorylation site (S1140A) in T cells inhibits the phosphorylation of the functionally important Thr-758 in the β2-chain, binding of α-actinin and 14-3-3 protein, and expression of an integrin-activating epitope after treatment with the stromal cell-derived factor-1α. The presence of this substitution resulted in a loss of cell adhesion and directional cell migration. Moreover, LFA-1 activation through the T-cell receptor in cells expressing the S1140A LFA-1 variant resulted in less Thr-758 phosphorylation, α-actinin and talin binding, and cell adhesion. The finding that the LFA-1 α-chain regulates adhesion through the β-chain via specific phosphorylation at Ser-1140 in the α-chain has not been previously reported and emphasizes that both chains are involved in the regulation of LFA-1 integrin activity.
Collapse
Affiliation(s)
- Farhana Jahan
- From the Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki 00014 UH, Finland
| | - Sudarrshan Madhavan
- From the Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki 00014 UH, Finland
| | - Taisia Rolova
- From the Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki 00014 UH, Finland
| | - Larisa Viazmina
- From the Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki 00014 UH, Finland
| | - Mikaela Grönholm
- From the Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki 00014 UH, Finland
| | - Carl G Gahmberg
- From the Faculty of Biological and Environmental Sciences, Molecular and Integrative Biosciences Research Program, University of Helsinki, Helsinki 00014 UH, Finland
| |
Collapse
|
84
|
Nieuwenhuis B, Haenzi B, Andrews MR, Verhaagen J, Fawcett JW. Integrins promote axonal regeneration after injury of the nervous system. Biol Rev Camb Philos Soc 2018; 93:1339-1362. [PMID: 29446228 PMCID: PMC6055631 DOI: 10.1111/brv.12398] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 12/23/2017] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
Integrins are cell surface receptors that form the link between extracellular matrix molecules of the cell environment and internal cell signalling and the cytoskeleton. They are involved in several processes, e.g. adhesion and migration during development and repair. This review focuses on the role of integrins in axonal regeneration. Integrins participate in spontaneous axonal regeneration in the peripheral nervous system through binding to various ligands that either inhibit or enhance their activation and signalling. Integrin biology is more complex in the central nervous system. Integrins receptors are transported into growing axons during development, but selective polarised transport of integrins limits the regenerative response in adult neurons. Manipulation of integrins and related molecules to control their activation state and localisation within axons is a promising route towards stimulating effective regeneration in the central nervous system.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
| | - Barbara Haenzi
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
| | | | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor SystemsNetherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW)1105 BAAmsterdamThe Netherlands
- Centre for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit Amsterdam1081 HVAmsterdamThe Netherlands
| | - James W. Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical NeurosciencesUniversity of CambridgeCambridgeCB2 0PYU.K.
- Centre of Reconstructive NeuroscienceInstitute of Experimental Medicine142 20Prague 4Czech Republic
| |
Collapse
|
85
|
Osterix regulates corticalization for longitudinal bone growth via integrin β3 expression. Exp Mol Med 2018; 50:1-11. [PMID: 30022046 PMCID: PMC6052162 DOI: 10.1038/s12276-018-0119-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 04/04/2018] [Accepted: 04/11/2018] [Indexed: 01/08/2023] Open
Abstract
Corticalization, coalescence of trabecular bone into the metaphyseal cortex, is important for the longitudinal growth of long bones. However, little is known about the molecular mechanisms controlling corticalization. To understand the molecular mechanisms underlying corticalization, we analyzed osteoblast-specific Osterix-knockout mice (Col-OMT). In control mice, corticalization was initiated after 7 postnatal days, and the number of osteoblasts in the peripheral spongiosa was increased compared to the number in the central spongiosa. In contrast, in Col-OMT mice, corticalization was delayed, and the number of osteoblasts in peripheral zones was unchanged compared to the central zone. Furthermore, femoral length was decreased in Col-OMT mice at 1 month. Because Col-OMT mice exhibited impaired matrix coalescence and osteoblast migration, we evaluated integrin signaling in Col-OMT mice. Osterix bound to the Itgb3 promoter and increased transcription of the Itgb3 gene in osteoblast cells. Interestingly, the inner and outer cortical bones were separated in Itgb3-null mice at postnatal day 7. In Itgb3-null mice, the number of osteoblasts in peripheral zones was not changed, and the femoral length was decreased. Taken together, these results indicate that Osterix regulates corticalization for longitudinal bone growth via the control of integrin β3 expression in osteoblasts. Our findings imply that the ability to control osteoblast function during corticalization may help in the treatment of short stature.
Collapse
|
86
|
Kawahara R, Niwa Y, Simizu S. Integrin β1 is an essential factor in vasculogenic mimicry of human cancer cells. Cancer Sci 2018; 109:2490-2496. [PMID: 29900640 PMCID: PMC6113431 DOI: 10.1111/cas.13693] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/01/2018] [Accepted: 06/08/2018] [Indexed: 12/16/2022] Open
Abstract
Vasculogenic mimicry (VM) formation by cancer cells is known to play a crucial role in tumor progression, but its detailed mechanism is unclear. In the present study, we focused on integrin β1 (ITGB1) and assessed the role of ITGB1 in VM formation. We used in vitro methods to seed cancer cells on Matrigel to evaluate the capability of VM formation. We carried out ITGB1 gene deletion using the CRISPR/Cas9 system, and these ITGB1‐knockout cells did not show a VM‐like network formation. Further, reintroduction of ITGB1 rescued VM‐like network formation in ITGB1‐knockout cells. In conclusion, ITGB1 is a critical factor in VM of human cancer cells, and inhibition of ITGB1 may be a novel therapeutic approach for malignant cancer.
Collapse
Affiliation(s)
- Ryota Kawahara
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Yuki Niwa
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| | - Siro Simizu
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Yokohama, Japan
| |
Collapse
|
87
|
Abstract
The formation of correct synaptic structures and neuronal connections is paramount for normal brain development and a functioning adult brain. The integrin family of cell adhesion receptors and their ligands play essential roles in the control of several processes regulating neuronal connectivity - including neurite outgrowth, the formation and maintenance of synapses, and synaptic plasticity - that are affected in neurodevelopmental disorders, such as autism spectrum disorders (ASDs) and schizophrenia. Many ASD- and schizophrenia-associated genes are linked to alterations in the genetic code of integrins and associated signalling pathways. In non-neuronal cells, crosstalk between integrin-mediated adhesions and the actin cytoskeleton, and the regulation of integrin activity (affinity for extracellular ligands) are widely studied in healthy and pathological settings. In contrast, the roles of integrin-linked pathways in the central nervous system remains less well defined. In this Review, we will provide an overview of the known pathways that are regulated by integrin-ECM interaction in developing neurons and in adult brain. We will also describe recent advances in the identification of mechanisms that regulate integrin activity in neurons, and highlight the interesting emerging links between integrins and neurodevelopment.
Collapse
Affiliation(s)
- Johanna Lilja
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland
| | - Johanna Ivaska
- Turku Centre for Biotechnology, University of Turku, FIN-20520 Turku, Finland .,Department of Biochemistry, University of Turku, FIN-20500 Turku, Finland
| |
Collapse
|
88
|
Spiess M, Hernandez-Varas P, Oddone A, Olofsson H, Blom H, Waithe D, Lock JG, Lakadamyali M, Strömblad S. Active and inactive β1 integrins segregate into distinct nanoclusters in focal adhesions. J Cell Biol 2018; 217:1929-1940. [PMID: 29632027 PMCID: PMC5987715 DOI: 10.1083/jcb.201707075] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 02/16/2018] [Accepted: 03/26/2018] [Indexed: 01/24/2023] Open
Abstract
Through two superresolution microscopy techniques, STED and STORM, Spiess et al. visualize the organization of integrins in focal adhesions and show that active and inactive β1 integrins assemble into distinct nanoclusters within adhesions, suggesting the existence of a novel mechanism that locally coordinates integrin activity. Integrins are the core constituents of cell–matrix adhesion complexes such as focal adhesions (FAs) and play key roles in physiology and disease. Integrins fluctuate between active and inactive conformations, yet whether the activity state influences the spatial organization of integrins within FAs has remained unclear. In this study, we address this question and also ask whether integrin activity may be regulated either independently for each integrin molecule or through locally coordinated mechanisms. We used two distinct superresolution microscopy techniques, stochastic optical reconstruction microscopy (STORM) and stimulated emission depletion microscopy (STED), to visualize active versus inactive β1 integrins. We first reveal a spatial hierarchy of integrin organization with integrin molecules arranged in nanoclusters, which align to form linear substructures that in turn build FAs. Remarkably, within FAs, active and inactive β1 integrins segregate into distinct nanoclusters, with active integrin nanoclusters being more organized. This unexpected segregation indicates synchronization of integrin activities within nanoclusters, implying the existence of a coordinate mechanism of integrin activity regulation.
Collapse
Affiliation(s)
- Matthias Spiess
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Pablo Hernandez-Varas
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden.,Wolfson Imaging Centre, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, UK
| | - Anna Oddone
- Institut de Ciències Fotòniques, Barcelona, Spain
| | - Helene Olofsson
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Hans Blom
- Science for Life Laboratory, Royal Institute of Technology, Solna, Sweden
| | - Dominic Waithe
- Wolfson Imaging Centre, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, UK
| | - John G Lock
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | | | - Staffan Strömblad
- Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
89
|
Thinn AMM, Wang Z, Zhu J. The membrane-distal regions of integrin α cytoplasmic domains contribute differently to integrin inside-out activation. Sci Rep 2018; 8:5067. [PMID: 29568062 PMCID: PMC5864728 DOI: 10.1038/s41598-018-23444-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 03/13/2018] [Indexed: 12/20/2022] Open
Abstract
Functioning as signal receivers and transmitters, the integrin α/β cytoplasmic tails (CT) are pivotal in integrin activation and signaling. 18 α integrin subunits share a conserved membrane-proximal region but have a highly diverse membrane-distal (MD) region at their CTs. Recent studies demonstrated that the presence of α CTMD region is essential for talin-induced integrin inside-out activation. However, it remains unknown whether the non-conserved α CTMD regions differently regulate the inside-out activation of integrin. Using αIIbβ3, αLβ2, and α5β1 as model integrins and by replacing their α CTMD regions with those of α subunits that pair with β3, β2, and β1 subunits, we analyzed the function of CTMD regions of 17 α subunits in talin-mediated integrin activation. We found that the α CTMD regions play two roles on integrin, which are activation-supportive and activation-regulatory. The regulatory but not the supportive function depends on the sequence identity of α CTMD region. A membrane-proximal tyrosine residue present in the CTMD regions of a subset of α integrins was identified to negatively regulate integrin inside-out activation. Our study provides a useful resource for investigating the function of α integrin CTMD regions.
Collapse
Affiliation(s)
- Aye Myat Myat Thinn
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, 53226, USA
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Zhengli Wang
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, 53226, USA
| | - Jieqing Zhu
- Blood Research Institute, BloodCenter of Wisconsin, Milwaukee, WI, 53226, USA.
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
90
|
Lu H, Bowler N, Harshyne LA, Craig Hooper D, Krishn SR, Kurtoglu S, Fedele C, Liu Q, Tang HY, Kossenkov AV, Kelly WK, Wang K, Kean RB, Weinreb PH, Yu L, Dutta A, Fortina P, Ertel A, Stanczak M, Forsberg F, Gabrilovich DI, Speicher DW, Altieri DC, Languino LR. Exosomal αvβ6 integrin is required for monocyte M2 polarization in prostate cancer. Matrix Biol 2018. [PMID: 29530483 DOI: 10.1016/j.matbio.2018.03.009] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Therapeutic approaches aimed at curing prostate cancer are only partially successful given the occurrence of highly metastatic resistant phenotypes that frequently develop in response to therapies. Recently, we have described αvβ6, a surface receptor of the integrin family as a novel therapeutic target for prostate cancer; this epithelial-specific molecule is an ideal target since, unlike other integrins, it is found in different types of cancer but not in normal tissues. We describe a novel αvβ6-mediated signaling pathway that has profound effects on the microenvironment. We show that αvβ6 is transferred from cancer cells to monocytes, including β6-null monocytes, by exosomes and that monocytes from prostate cancer patients, but not from healthy volunteers, express αvβ6. Cancer cell exosomes, purified via density gradients, promote M2 polarization, whereas αvβ6 down-regulation in exosomes inhibits M2 polarization in recipient monocytes. Also, as evaluated by our proteomic analysis, αvβ6 down-regulation causes a significant increase in donor cancer cells, and their exosomes, of two molecules that have a tumor suppressive role, STAT1 and MX1/2. Finally, using the Ptenpc-/- prostate cancer mouse model, which carries a prostate epithelial-specific Pten deletion, we demonstrate that αvβ6 inhibition in vivo causes up-regulation of STAT1 in cancer cells. Our results provide evidence of a novel mechanism that regulates M2 polarization and prostate cancer progression through transfer of αvβ6 from cancer cells to monocytes through exosomes.
Collapse
Affiliation(s)
- Huimin Lu
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Nicholas Bowler
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Larry A Harshyne
- Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - D Craig Hooper
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Shiv Ram Krishn
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Senem Kurtoglu
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Carmine Fedele
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Qin Liu
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA, USA
| | - Hsin-Yao Tang
- Center for Systems and Computational Biology, Wistar Institute, Philadelphia, PA, USA
| | - Andrew V Kossenkov
- Center for Systems and Computational Biology, Wistar Institute, Philadelphia, PA, USA
| | - William K Kelly
- Departments of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Kerith Wang
- Departments of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rhonda B Kean
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Lei Yu
- Flow Cytometry Core Facility, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Anindita Dutta
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Paolo Fortina
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Cancer Genomics and Bioinformatics Laboratory, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Adam Ertel
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Cancer Genomics and Bioinformatics Laboratory, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Maria Stanczak
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Flemming Forsberg
- Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dmitry I Gabrilovich
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Immunology, Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA, USA
| | - David W Speicher
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA, USA; Center for Systems and Computational Biology, Wistar Institute, Philadelphia, PA, USA
| | - Dario C Altieri
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Immunology, Microenvironment and Metastasis Program, Wistar Institute, Philadelphia, PA, USA
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, Pennsylvania, USA; Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
91
|
Demiray YE, Rehberg K, Kliche S, Stork O. Ndr2 Kinase Controls Neurite Outgrowth and Dendritic Branching Through α 1 Integrin Expression. Front Mol Neurosci 2018; 11:66. [PMID: 29559888 PMCID: PMC5845635 DOI: 10.3389/fnmol.2018.00066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 02/16/2018] [Indexed: 12/30/2022] Open
Abstract
The serine/threonine kinase Ndr2 has been shown to control the inside-out activation of the β1subunit of integrins and the formation of neurites in both primary neurons and neurally differentiated pheochromacytoma (PC12) cells. In this study, we demonstrate that Ndr2 kinase furthermore determines the substrate specificity of neurite extension in PC12 cells via expression of α1β1 integrins. We show that stable overexpression of Ndr2 in PC12 cells increases neurite growth and extension on poly-D-lysine substrate, likely involving an increased expression of active β1 integrin in the growth tips of these cells. By contrast, the Ndr2 overexpressing cells do not show the α1β1 integrin-mediated enhancement of neurite growth on collagen IV substrate that can be seen in control cells. Moreover, they entirely fail to increase in response to activation of α1β1 integrins via a soluble KTS ligand and show a diminished accumulation of α1 integrin in neurite tips, although the expression of this subunit is induced during differentiation to comparable levels as in control cells. Finally, we demonstrate that Ndr2 overexpression similarly inhibits the α1β1 integrin-dependent dendritic growth of primary hippocampal neurons on laminin 111 substrate. By contrast, lack of Ndr2 impairs the dendritic growth regardless of the substrate. Together, these results suggest that Ndr2 regulates α1 integrin trafficking in addition to β1 integrin subunit activation and thereby controls the neurite growth on different extracellular matrix (ECM) substrates.
Collapse
Affiliation(s)
- Yunus E Demiray
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Kati Rehberg
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Stefanie Kliche
- Institute of Molecular and Clinical Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Oliver Stork
- Department of Genetics and Molecular Neurobiology, Institute of Biology, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Center for Behavioral Brain Science, Magdeburg, Germany
| |
Collapse
|
92
|
Bouvier C, Macagno N, Nguyen Q, Loundou A, Jiguet-Jiglaire C, Gentet JC, Jouve JL, Rochwerger A, Mattei JC, Bouvard D, Salas S. Prognostic value of the Hippo pathway transcriptional coactivators YAP/TAZ and β1-integrin in conventional osteosarcoma. Oncotarget 2018; 7:64702-64710. [PMID: 27608849 PMCID: PMC5323109 DOI: 10.18632/oncotarget.11876] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 08/01/2016] [Indexed: 11/25/2022] Open
Abstract
Introduction Currently, very few studies are available concerning the mammalian Hippo pathway in bone sarcomas. YAP/TAZ transcription co-activators are key downstream effectors of this pathway and may also have oncogenic properties. Additionally, recent in-vitro experiments showed that expression of β1-integrin promoted metastasis in osteosarcomas. This study investigated the expression of YAP/TAZ and β1-integrin in human osteosarcomas. Materials and methods We performed automated immunohistochemistry on tissue-microarrays (TMA) in which 69 conventional osteosarcomas biopsies performed prior to chemotherapy were embedded. Cellular localization and semi-quantitative analysis of each immunostain was performed using Immunoreactive Score (IRS) and correlated to clinico-pathological data. Results Cytoplasmic expression of β1-integrin was noted in 54/59 osteosarcomas (92%), with 33/59 cases (56%) displaying membranous staining. YAP/TAZ was expressed in 27/45 osteosarcomas (60%), with 14 cases (31%) showing cytoplasmic expression while 13 other cases (28%) displayed nuclear expression. No link was found between YAP/TAZ or β1-integrin expression and response to chemotherapy. In univariate analysis, YAP/TAZ immunoreactive score was pejoratively correlated with overall survival (p = 0.01). Expression of β1-integrin on cell membrane was also pejorative for OS (p = 0.045). In multivariate analysis, YAP/TAZ nuclear expression was an independent prognostic factor for PFS (p = 0.035). Conclusion this study indicates that β1-integrin and YAP/TAZ proteins are linked to prognosis and therefore could be therapeutic targets in conventional osteosarcomas.
Collapse
Affiliation(s)
- Corinne Bouvier
- Aix-Marseille University (AMU), Faculty of Medecine, CRO2, UMR 911 (Equipe IV), Marseille, France.,Department of Pathology, APHM, Timone Hospital, Marseille, France
| | - Nicolas Macagno
- Aix-Marseille University (AMU), Faculty of Medecine, CRO2, UMR 911 (Equipe IV), Marseille, France.,Department of Pathology, APHM, Timone Hospital, Marseille, France
| | - Quy Nguyen
- Aix-Marseille University (AMU), Faculty of Medecine, CRO2, UMR 911 (Equipe IV), Marseille, France
| | - Anderson Loundou
- Department of Public Health, Aix-Marseille University (AMU), Faculty of Medecine, EA 3270 Research Unit, Marseille, France.,Department of Research and Innovation, APHM, Timone Hospital, Support Unit for Clinical Research and Economic Evaluation, Marseille, France
| | - Carine Jiguet-Jiglaire
- Aix-Marseille University (AMU), Faculty of Medecine, CRO2, UMR 911 (Equipe IV), Marseille, France
| | - Jean-Claude Gentet
- Department of Pediatric Oncology, APHM, Timone Hospital, Marseille, France
| | - Jean-Luc Jouve
- Department of Pediatric Orthopaedic Surgery, APHM, Timone Hospital, Marseille, France
| | | | - Jean-Camille Mattei
- Aix-Marseille University (AMU), Faculty of Medecine, CRO2, UMR 911 (Equipe IV), Marseille, France.,Department of Adult Orthopaedic Surgery, APHM, Nord Hospital, Marseille, France
| | | | - Sébastien Salas
- Aix-Marseille University (AMU), Faculty of Medecine, CRO2, UMR 911 (Equipe IV), Marseille, France.,Department of Pathology, APHM, Timone Hospital, Marseille, France
| |
Collapse
|
93
|
Endo Y, Ishiwata-Endo H, Yamada KM. Cell adhesion to anosmin via α5β1, α4β1, and α9β1 integrins. Cell Adh Migr 2018; 12:93-100. [PMID: 27715389 DOI: 10.1080/19336918.2016.1221568] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Anosmin is an extracellular matrix protein, and genetic defects in anosmin result in human Kallmann syndrome. It functions in neural crest formation, cell adhesion, and neuronal migration. Anosmin consists of multiple domains, and it has been reported to bind heparan sulfate, FGF receptor, and UPA. In this study, we establish cell adhesion/spreading assays for anosmin and use them for antibody inhibition analyses to search for an integrin adhesion receptor. We find that α5β1, α4β1, and α9β1 integrins are needed for effective adhesive receptor function in cell adhesion and cell spreading on anosmin; adhesion is inhibited by both RGD and α4β1 CS1-based peptides. This identification of anosmin-integrin adhesion receptors should facilitate studies of anosmin function in cell and developmental biology.
Collapse
Affiliation(s)
- Yukinori Endo
- a Laboratory of Cell and Developmental Biology , National Institute of Dental and Craniofacial Research, National Institutes of Health , Bethesda , MD , USA
| | - Hiroko Ishiwata-Endo
- a Laboratory of Cell and Developmental Biology , National Institute of Dental and Craniofacial Research, National Institutes of Health , Bethesda , MD , USA
| | - Kenneth M Yamada
- a Laboratory of Cell and Developmental Biology , National Institute of Dental and Craniofacial Research, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
94
|
Vasodilator-stimulated phosphoprotein promotes liver metastasis of gastrointestinal cancer by activating a β1-integrin-FAK-YAP1/TAZ signaling pathway. NPJ Precis Oncol 2018; 2:2. [PMID: 29872721 PMCID: PMC5871906 DOI: 10.1038/s41698-017-0045-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 12/06/2017] [Accepted: 12/28/2017] [Indexed: 12/26/2022] Open
Abstract
Extracellular matrix (ECM)-induced β1-integrin-FAK signaling promotes cell attachment, survival, and migration of cancer cells in a distant organ so as to enable cancer metastasis. However, mechanisms governing activation of the β1-integrin-FAK signaling remain incompletely understood. Here, we report that vasodilator-stimulated phosphoprotein (VASP), an actin binding protein, is required for ECM–mediated β1-integrin-FAK-YAP1/TAZ signaling in gastrointestinal (GI) cancer cells and their liver metastasis. In patient-derived samples, VASP is upregulated in 53 of 63 colorectal cancers and 43 of 53 pancreatic ductal adenocarcinomas and high VASP levels correlate with liver metastasis and reduced patient survival. In a Matrigel-based 3-dimensional (3D) culture model, short hairpin RNA (shRNA)–mediated VASP knockdown in colorectal cancer cells (KM12L4, HCT116, and HT29) and pancreatic cancer cells (L3.6 and MIA PaCa-1) suppresses the growth of 3D cancer spheroids. Mechanistic studies reveal that VASP knockdown suppresses FAK phosphorylation and YAP1/TAZ protein levels, but not Akt or Erk-related pathways and that YAP1/TAZ proteins are enhanced by the β1-integrin-FAK signaling. Additionally, VASP regulates the β1-integrin-FAK-YAP1/TAZ signaling by at least two mechanisms: (1) promoting ECM-mediated β1-integrin activation and (2) regulating YAP1/TAZ dephosphorylation at downstream of RhoA to enhance the stability of YAP1/TAZ proteins. In agreement with these, preclinical studies with two experimental liver metastasis mouse models demonstrate that VASP knockdown suppresses GI cancer liver metastasis, β1-integrin activation, and YAP1/TAZ levels of metastatic cancer cells. Together, our data support VASP as a treatment target for liver metastasis of colorectal and pancreatic cancers. A protein involved in cytoskeleton regulation and cell motility control offers a new drug target for cancer spreading to the liver. Ningling Kang Ph.D. from the Hormel Institute in Austin, Minnesota, USA, and colleagues showed that levels of this actin-binding protein, known as vasodilator-stimulated phosphoprotein (VASP), are elevated in most patients with advanced colon and pancreatic cancers and that higher VASP expression levels are linked to liver metastasis and poorer patients’ outcomes. To explore the reasons why, the researchers studied three-dimensional tumor spheroids and mouse metastasis models of these cancers, and identified the signaling pathway by which VASP promotes the survival of cancer cells in distant organs, such as the liver. What’s more, they showed that knocking down VASP of cancer cells in metastasis mouse models suppressed cancer metastatic growth in the liver, suggesting that the same might be true in patients as well.
Collapse
|
95
|
Nordenfelt P, Moore TI, Mehta SB, Kalappurakkal JM, Swaminathan V, Koga N, Lambert TJ, Baker D, Waters JC, Oldenbourg R, Tani T, Mayor S, Waterman CM, Springer TA. Direction of actin flow dictates integrin LFA-1 orientation during leukocyte migration. Nat Commun 2017; 8:2047. [PMID: 29229906 PMCID: PMC5725580 DOI: 10.1038/s41467-017-01848-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 10/20/2017] [Indexed: 12/31/2022] Open
Abstract
Integrin αβ heterodimer cell surface receptors mediate adhesive interactions that provide traction for cell migration. Here, we test whether the integrin, when engaged to an extracellular ligand and the cytoskeleton, adopts a specific orientation dictated by the direction of actin flow on the surface of migrating cells. We insert GFP into the rigid, ligand-binding head of the integrin, model with Rosetta the orientation of GFP and its transition dipole relative to the integrin head, and measure orientation with fluorescence polarization microscopy. Cytoskeleton and ligand-bound integrins orient in the same direction as retrograde actin flow with their cytoskeleton-binding β-subunits tilted by applied force. The measurements demonstrate that intracellular forces can orient cell surface integrins and support a molecular model of integrin activation by cytoskeletal force. Our results place atomic, Å-scale structures of cell surface receptors in the context of functional and cellular, μm-scale measurements.
Collapse
Affiliation(s)
- Pontus Nordenfelt
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Physiology Course, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Program in Cellular and Molecular Medicine, Children's Hospital, and Department of Biological Chemistry and Molecular Pharmacology and Medicine, Harvard Medical School, Boston, MA, 02115, USA
- Division of Infection Medicine, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Lund, 221 84, Sweden
| | - Travis I Moore
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Program in Cellular and Molecular Medicine, Children's Hospital, and Department of Biological Chemistry and Molecular Pharmacology and Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Shalin B Mehta
- Eugene Bell Center, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA
| | - Joseph Mathew Kalappurakkal
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Physiology Course, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- National Center for Biological Sciences, Bangalore, 560065, India
| | - Vinay Swaminathan
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Physiology Course, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Cell Biology and Physiology Center, NHLBI, NIH, Bethesda, MD, 20824, USA
| | - Nobuyasu Koga
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
- Institute for Molecular Science, Myodaiji, Okazaki, 444-8585, Japan
| | - Talley J Lambert
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - David Baker
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, 98195, USA
| | - Jennifer C Waters
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Rudolf Oldenbourg
- Eugene Bell Center, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
| | - Tomomi Tani
- Eugene Bell Center, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
| | - Satyajit Mayor
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Physiology Course, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- National Center for Biological Sciences, Bangalore, 560065, India
| | - Clare M Waterman
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Physiology Course, Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Cell Biology and Physiology Center, NHLBI, NIH, Bethesda, MD, 20824, USA
| | - Timothy A Springer
- Whitman Center, Marine Biological Laboratory, Woods Hole, MA, 02543, USA.
- Physiology Course, Marine Biological Laboratory, Woods Hole, MA, 02543, USA.
- Program in Cellular and Molecular Medicine, Children's Hospital, and Department of Biological Chemistry and Molecular Pharmacology and Medicine, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
96
|
Lockett AD, Wu Y, Gunst SJ. Elastase alters contractility and promotes an inflammatory synthetic phenotype in airway smooth muscle tissues. Am J Physiol Lung Cell Mol Physiol 2017; 314:L626-L634. [PMID: 29212803 DOI: 10.1152/ajplung.00334.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Neutrophil elastase is secreted by inflammatory cells during airway inflammation and can elicit airway hyperreactivity in vivo. Elastase can degrade multiple components of the extracellular matrix. We hypothesized that elastase might disrupt the connections between airway smooth muscle (ASM) cells and the extracellular matrix and that this might have direct effects on ASM tissue responsiveness and inflammation. The effect of elastase treatment on ASM contractility was assessed in vitro in isolated strips of canine tracheal smooth muscle by stimulation of tissues with cumulatively increasing concentrations of acetylcholine (ACh) and measurement of contractile force. Elastase treatment potentiated contractile responses to ACh at low concentrations but suppressed the maximal contractile force generated by the tissues without affecting the phosphorylation of myosin regulatory light chain (RLC). Elastase also promoted the secretion of eotaxin and the activation of Akt in ASM tissues and decreased expression of smooth muscle myosin heavy chain, consistent with promotion of a synthetic inflammatory phenotype. As the degradation of matrix proteins can alter integrin engagement, we evaluated the effect of elastase on the assembly and activation of integrin-associated adhesion junction complexes in ASM tissues. Elastase led to talin cleavage, reduced talin binding to vinculin, and suppressed activation of the adhesome proteins paxillin, focal adhesion kinase, and vinculin, indicating that elastase causes the disassembly of adhesion junction complexes and the inactivation of adhesome signaling proteins. We conclude that elastase promotes an inflammatory phenotype and increased sensitivity to ACh in ASM tissues by disrupting signaling pathways mediated by integrin-associated adhesion complexes.
Collapse
Affiliation(s)
- Angelia D Lockett
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine , Indianapolis, Indiana
| | - Yidi Wu
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine , Indianapolis, Indiana
| | - Susan J Gunst
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine , Indianapolis, Indiana
| |
Collapse
|
97
|
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: 10.7] [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]
|
98
|
Li J, Springer TA. Energy landscape differences among integrins establish the framework for understanding activation. J Cell Biol 2017; 217:397-412. [PMID: 29122968 PMCID: PMC5748972 DOI: 10.1083/jcb.201701169] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 09/15/2017] [Accepted: 10/04/2017] [Indexed: 11/22/2022] Open
Abstract
Li and Springer demonstrate differences between integrins α4β1 and α5β1 in intrinsic affinities and relative free energies of three conformational states. Integrin conformational equilibria are both subunit and cell type specific. The energy landscapes of intact receptors on the cell surface provide a framework for understanding regulation of integrin adhesiveness. Why do integrins differ in basal activity, and how does affinity for soluble ligand correlate with cellular adhesiveness? We show that basal conformational equilibrium set points for integrin α4β1 are cell type specific and differ from integrin α5β1 when the two integrins are coexpressed on the same cell. Although α4β1 is easier to activate, its high-affinity state binds vascular cell adhesion molecule and fibronectin 100- to 1,000-fold more weakly than α5β1 binds fibronectin. Furthermore, the difference in affinity between the high- and low-affinity states is more compressed in α4β1 (600- to 800-fold) than in α5β1 (4,000- to 6,000-fold). α4β1 basal conformational equilibria differ among three cell types, define affinity for soluble ligand and readiness for priming, and may reflect differences in interactions with intracellular adaptors but do not predict cellular adhesiveness for immobilized ligand. The measurements here provide a necessary framework for understanding integrin activation in intact cells, including activation of integrin adhesiveness by application of tensile force by the cytoskeleton, across ligand–integrin–adaptor complexes.
Collapse
Affiliation(s)
- Jing Li
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Timothy A Springer
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA .,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| |
Collapse
|
99
|
Shuai C, Guo W, Gao C, Yang Y, Wu P, Feng P. An nMgO containing scaffold: Antibacterial activity, degradation properties and cell responses. Int J Bioprint 2017; 4:120. [PMID: 33102906 PMCID: PMC7582014 DOI: 10.18063/ijb.v4i1.120] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 10/10/2017] [Indexed: 01/08/2023] Open
Abstract
Bone repair failure caused by implant-related infections is a common and troublesome problem. In this study, an antibacterial scaffold was developed via selective laser sintering with incorporating nano magnesium oxide (nMgO) to poly(3-hydroxybutyrate-co-3-hydroxyvalerate) (PHBV). The results indicated the scaffold exerted high antibacterial activity. The antibacterial mechanism was that nMgO could cause oxidative damage and mechanical damage to bacteria through the production of reactive oxygen species (ROS) and direct contact action, respectively, which resulted in the damage of their structures and functions. Besides, nMgO significantly increased the compressive properties of the scaffold including strength and modulus, due to its excellent mechanical properties and uniform dispersion in the PHBV matrix. Moreover, the degradation tests indicated nMgO neutralized the acid degradation products of PHBV and benefited the degradation of the scaffold. The cell culture demonstrated that nMgO promoted the cellular adhesion and proliferation, as well as osteogenic differentiation. The present work may open the door to exploring nMgO as a promising antibacterial material for tissue engineering.
Collapse
Affiliation(s)
- Cijun Shuai
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
- Jiangxi University of Science and Technology, Ganzhou, China
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Changsha, China
| | - Wang Guo
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Chengde Gao
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Youwen Yang
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| | - Ping Wu
- College of Chemistry, Xiangtan University, Xiangtan, China
| | - Pei Feng
- State Key Laboratory of High Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, China
| |
Collapse
|
100
|
Dogan A, Parmaksız M, Elçin AE, Elçin YM. Extracellular Matrix and Regenerative Therapies from the Cardiac Perspective. Stem Cell Rev Rep 2017; 12:202-13. [PMID: 26668014 DOI: 10.1007/s12015-015-9641-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases are the leading cause of death and a major cause of financial burden. Regenerative therapies for heart diseases bring the promise of alternative treatment modalities for myocardial infarction, ischemic heart disease, and congestive heart failure. Although, clinical trials attest to the safety of stem cell injection therapies, researchers need to overcome the underlying mechanisms that are limiting the success of future regenerative options. This article aims to review the basic scientific concepts in the field of mechanobiology and the effects of extracellular functions on stem cell fate.
Collapse
Affiliation(s)
- Arin Dogan
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Degol Caddesi, Tandogan, 06100, Ankara, Turkey
| | - Mahmut Parmaksız
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Degol Caddesi, Tandogan, 06100, Ankara, Turkey
| | - A Eser Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Degol Caddesi, Tandogan, 06100, Ankara, Turkey
| | - Y Murat Elçin
- Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Degol Caddesi, Tandogan, 06100, Ankara, Turkey.
| |
Collapse
|