Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 1840: 73-80, 2018.
Article in English | MEDLINE | ID: mdl-30141039

ABSTRACT

The mechanical properties of the cellular microenvironment can impact many aspects of cell behavior, including molecular processes in the nucleus. Recent studies indicate that the LINC complex and its associated nuclear envelope transmit and transduce mechanical stress into biochemical pathways that ultimately regulate nuclear structure or gene expression. Here we describe a method to apply tensional forces to the LINC complex of isolated nuclei. Using magnetic beads and magnets, this technique can be used to explore the biochemical pathways that are activated in response to tension applied to the surface of isolated nuclei.


Subject(s)
Cytoskeleton/metabolism , Mechanotransduction, Cellular , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Cell Fractionation , Cell Nucleus/metabolism , HeLa Cells , Humans
2.
J Vis Exp ; (121)2017 03 08.
Article in English | MEDLINE | ID: mdl-28362397

ABSTRACT

Mechanosensitive cell surface adhesion complexes allow cells to sense the mechanical properties of their surroundings. Recent studies have identified both force-sensing molecules at adhesion sites, and force-dependent transcription factors that regulate lineage-specific gene expression and drive phenotypic outputs. However, the signaling networks converting mechanical tension into biochemical pathways have remained elusive. To explore the signaling pathways engaged upon mechanical tension applied to cell surface receptor, superparamagnetic microbeads can be used. Here we present a protocol for using magnetic beads to apply forces to cell surface adhesion proteins. Using this approach, it is possible to investigate not only force-dependent cytoplasmic signaling pathways by various biochemical approaches, but also adhesion remodeling by magnetic isolation of adhesion complexes attached to the ligand-coated beads. This protocol includes the preparation of ligand-coated superparamagnetic beads, and the application of define tensile forces followed by biochemical analyses. Additionally, we provide a representative sample of data demonstrating that tension applied to integrin-based adhesion triggers adhesion remodeling and alters protein tyrosine phosphorylation.


Subject(s)
Cell Adhesion/physiology , Cell Membrane/metabolism , Magnets , Receptors, Cell Surface/metabolism , Stress, Mechanical , Phosphorylation , Signal Transduction/physiology
3.
Proteomics Clin Appl ; 10(1): 51-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26147903

ABSTRACT

PURPOSE: Multipotent mesenchymal stem cells (MSCs) have the capability to differentiate down adipocyte, osteocyte and chondrocyte lineages and as such offer a range of potential therapeutic applications. The composition and stiffness of the extracellular matrix (ECM) environment that surrounds cells dictates their transcriptional programme, thereby affecting stem cell lineage decision-making. Cells sense force via linkages between themselves and their microenvironment, and this is transmitted by integrin receptors and associated adhesion signalling complexes. To identify regulators of MSC force sensing, we sought to catalogue MSC integrin-associated adhesion complex composition. EXPERIMENTAL DESIGN: Adhesion complexes formed by MSCs plated on the ECM ligand fibronectin were isolated and characterised by MS. Identified proteins were interrogated by comparison to a literature-based reference set of cell adhesion-related components and using ontological and protein-protein interaction network analyses. RESULTS: Adhesion complex-specific proteins in MSCs were identified that comprised predominantly cell adhesion-related adaptors and actin cytoskeleton regulators. Furthermore, LIM domain-containing proteins in MSC adhesion complexes were highlighted, which may act as force-sensing components. CONCLUSION AND CLINICAL RELEVANCE: These data provide a valuable resource of information regarding the molecular connections that link integrins and adhesion signalling in MSCs, and as such may present novel opportunities for therapeutic intervention.


Subject(s)
Databases, Protein , Integrins/metabolism , Mesenchymal Stem Cells/metabolism , Humans , Integrins/genetics , Proteomics
4.
Nat Cell Biol ; 17(12): 1577-1587, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26479319

ABSTRACT

Integrin receptor activation initiates the formation of integrin adhesion complexes (IACs) at the cell membrane that transduce adhesion-dependent signals to control a multitude of cellular functions. Proteomic analyses of isolated IACs have revealed an unanticipated molecular complexity; however, a global view of the consensus composition and dynamics of IACs is lacking. Here, we have integrated several IAC proteomes and generated a 2,412-protein integrin adhesome. Analysis of this data set reveals the functional diversity of proteins in IACs and establishes a consensus adhesome of 60 proteins. The consensus adhesome is likely to represent a core cell adhesion machinery, centred around four axes comprising ILK-PINCH-kindlin, FAK-paxillin, talin-vinculin and α-actinin-zyxin-VASP, and includes underappreciated IAC components such as Rsu-1 and caldesmon. Proteomic quantification of IAC assembly and disassembly detailed the compositional dynamics of the core cell adhesion machinery. The definition of this consensus view of integrin adhesome components provides a resource for the research community.


Subject(s)
Focal Adhesions/metabolism , Integrins/metabolism , Proteome/metabolism , Proteomics/methods , Actinin/metabolism , Animals , Cell Adhesion/drug effects , Cell Line, Tumor , Cells, Cultured , Cluster Analysis , Focal Adhesions/drug effects , Humans , Immunoblotting , K562 Cells , Kinetics , Mass Spectrometry , Mice , Microscopy, Fluorescence , Nocodazole/pharmacology , Paxillin/metabolism , Protein Interaction Maps , Proteome/classification , Talin/metabolism , Tubulin Modulators/pharmacology , Vinculin/metabolism , Zyxin/metabolism
5.
Curr Protoc Cell Biol ; 66: 9.8.1-9.8.15, 2015 Mar 02.
Article in English | MEDLINE | ID: mdl-25727331

ABSTRACT

The integration of cells with their extracellular environment is facilitated by cell surface adhesion receptors, such as integrins, which play important roles in both normal development and the onset of pathologies. Engagement of integrins with their ligands in the extracellular matrix, or counter-receptors on other cells, initiates the intracellular assembly of a wide variety of proteins into adhesion complexes such as focal contacts, focal adhesions, and fibrillar adhesions. The proteins recruited to these complexes mediate bidirectional signaling across the plasma membrane, and, as such, help to coordinate and/or modulate the multitude of physical and chemical signals to which the cell is subjected. The protocols in this unit describe two approaches for the isolation or enrichment of proteins contained within integrin-associated adhesion complexes, together with their local plasma membrane/cytosolic environments, from cells in culture. In the first protocol, integrin-associated adhesion structures are affinity isolated using microbeads coated with extracellular ligands or antibodies. The second protocol describes the isolation of ventral membrane preparations that are enriched for adhesion complex structures. The protocols permit the determination of adhesion complex components via subsequent downstream analysis by western blotting or mass spectrometry.


Subject(s)
Cytological Techniques/methods , Integrins/isolation & purification , Integrins/metabolism , Animals , Cattle , Cell Adhesion , Fibroblasts/cytology , Fibroblasts/drug effects , Fibronectins/pharmacology , Humans , K562 Cells , Male , Microspheres , Proteomics
6.
PLoS One ; 9(12): e115213, 2014.
Article in English | MEDLINE | ID: mdl-25526367

ABSTRACT

The microtubule network regulates the turnover of integrin-containing adhesion complexes to stimulate cell migration. Disruption of the microtubule network results in an enlargement of adhesion complex size due to increased RhoA-stimulated actomyosin contractility, and inhibition of adhesion complex turnover; however, the microtubule-dependent changes in adhesion complex composition have not been studied in a global, unbiased manner. Here we used label-free quantitative mass spectrometry-based proteomics to determine adhesion complex changes that occur upon microtubule disruption with nocodazole. Nocodazole-treated cells displayed an increased abundance of the majority of known adhesion complex components, but no change in the levels of the fibronectin-binding α5ß1 integrin. Immunofluorescence analyses confirmed these findings, but revealed a change in localisation of adhesion complex components. Specifically, in untreated cells, α5-integrin co-localised with vinculin at peripherally located focal adhesions and with tensin at centrally located fibrillar adhesions. In nocodazole-treated cells, however, α5-integrin was found in both peripherally located and centrally located adhesion complexes that contained both vinculin and tensin, suggesting a switch in the maturation state of adhesion complexes to favour focal adhesions. Moreover, the switch to focal adhesions was confirmed to be force-dependent as inhibition of cell contractility with the Rho-associated protein kinase inhibitor, Y-27632, prevented the nocodazole-induced conversion. These results highlight a complex interplay between the microtubule cytoskeleton, adhesion complex maturation state and intracellular contractile force, and provide a resource for future adhesion signaling studies. The proteomics data have been deposited in the ProteomeXchange with identifier PXD001183.


Subject(s)
Focal Adhesions/metabolism , Integrin alpha5beta1/metabolism , Microtubules/drug effects , Nocodazole/pharmacology , Tubulin Modulators/pharmacology , Cell Adhesion , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Foreskin/cytology , Humans , Male , Mass Spectrometry/methods , Microtubules/metabolism , Proteomics/methods , Vinculin/metabolism
7.
J Biol Chem ; 288(28): 20248-60, 2013 Jul 12.
Article in English | MEDLINE | ID: mdl-23720740

ABSTRACT

Focal adhesion turnover during cell migration is an integrated cyclic process requiring tight regulation of integrin function. Interaction of integrin with its ligand depends on its activation state, which is regulated by the direct recruitment of proteins onto the ß integrin chain cytoplasmic domain. We previously reported that ICAP-1α, a specific cytoplasmic partner of ß1A integrins, limits both talin and kindlin interaction with ß1 integrin, thereby restraining focal adhesion assembly. Here we provide evidence that the calcium and calmodulin-dependent serine/threonine protein kinase type II (CaMKII) is an important regulator of ICAP-1α for controlling focal adhesion dynamics. CaMKII directly phosphorylates ICAP-1α and disrupts an intramolecular interaction between the N- and the C-terminal domains of ICAP-1α, unmasking the PTB domain, thereby permitting ICAP-1α binding onto the ß1 integrin tail. ICAP-1α direct interaction with the ß1 integrin tail and the modulation of ß1 integrin affinity state are required for down-regulating focal adhesion assembly. Our results point to a molecular mechanism for the phosphorylation-dependent control of ICAP-1α function by CaMKII, allowing the dynamic control of ß1 integrin activation and cell adhesion.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Focal Adhesions/metabolism , Integrin beta1/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Benzylamines/pharmacology , CHO Cells , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Cricetinae , Cricetulus , Focal Adhesions/drug effects , Focal Adhesions/genetics , Immunoblotting , Integrin beta1/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Microscopy, Confocal , Models, Biological , Mutation , NIH 3T3 Cells , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Rats , Sulfonamides/pharmacology , Threonine/genetics , Threonine/metabolism , Time-Lapse Imaging
8.
J Cell Biol ; 194(2): 307-22, 2011 Jul 25.
Article in English | MEDLINE | ID: mdl-21768292

ABSTRACT

The morphogenetic and differentiation events required for bone formation are orchestrated by diffusible and insoluble factors that are localized within the extracellular matrix. In mice, the deletion of ICAP-1, a modulator of ß1 integrin activation, leads to severe defects in osteoblast proliferation, differentiation, and mineralization and to a delay in bone formation. Deposition of fibronectin and maturation of fibrillar adhesions, adhesive structures that accompany fibronectin deposition, are impaired upon ICAP-1 loss, as are type I collagen deposition and mineralization. Expression of ß1 integrin with a mutated binding site for ICAP-1 recapitulates the ICAP-1-null phenotype. Follow-up experiments demonstrated that ICAP-1 negatively regulates kindlin-2 recruitment onto the ß1 integrin cytoplasmic domain, whereas an excess of kindlin-2 binding has a deleterious effect on fibrillar adhesion formation. These results suggest that ICAP-1 works in concert with kindlin-2 to control the dynamics of ß1 integrin-containing fibrillar adhesions and, thereby, regulates fibronectin deposition and osteoblast mineralization.


Subject(s)
Calcification, Physiologic , Fibronectins/metabolism , Integrin beta1/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Osteoblasts/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cytoskeletal Proteins/metabolism , Extracellular Matrix/metabolism , Intracellular Signaling Peptides and Proteins/deficiency , Mice , Muscle Proteins/metabolism , Osteoblasts/cytology , Protein Binding
9.
Eur J Cell Biol ; 90(2-3): 261-9, 2011.
Article in English | MEDLINE | ID: mdl-20971526

ABSTRACT

Cells exert actomyosin contractility and cytoskeleton-dependent force in response to matrix stiffness cues. Cells dynamically adapt to force by modifying their behavior and remodeling their microenvironment. This adaptation is favored by integrin activation switch and their ability to modulate their clustering and the assembly of an intracellular hub in response to force. Indeed integrins are mechanoreceptors and mediate mechanotransduction by transferring forces to specific adhesion proteins into focal adhesions which are sensitive to tension and activate intracellular signals. α(5)ß(1) integrin is considered of major importance for the formation of an elaborate meshwork of fibronectin fibrils and for the extracellular matrix deposition and remodeling. Here we summarize recent progress in the study of mechanisms regulating the activation cycle of ß(1) integrin and the specificity of α(5)ß(1) integrin in mechanotransduction.


Subject(s)
Cell Adhesion/physiology , Integrin beta1/metabolism , Animals , Humans , Integrin alpha5beta1/metabolism , Mechanotransduction, Cellular , Mice , Signal Transduction
10.
Eur J Cell Biol ; 87(8-9): 491-506, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18417250

ABSTRACT

Cell-matrix adhesions are essential for cell migration, tissue organization and differentiation, therefore playing central roles in embryonic development, remodeling and homeostasis of tissues and organs. Matrix adhesion-dependent signals cooperate with other pathways to regulate biological functions such as cell survival, cell proliferation, wound healing, and tumorigenesis. Cell migration and invasion are integrated processes requiring the continuous, coordinated assembly and disassembly of integrin-mediated adhesions. An understanding of how integrins regulate cell migration and invasiveness through the dynamic regulation of adhesions is fundamental to both physiological and pathological situations. A variety of cell-matrix adhesions has been identified, namely, focal complexes, focal adhesions, fibrillar adhesions, podosomes, and invadopodia (podosome-type adhesions). These adhesion sites contain integrin clusters able to develop specialized structures, which are different in their architecture and dynamics although they share almost the same proteins. Here we compare recent advances and developments in the elucidation of the organization and dynamics of focal adhesions and podosome-type adhesions, in order to understand how such subcellular sites - though closely related in their composition - can be structurally and functionally different. The underlying question is how their respective physiological or pathological roles are related to their distinct organization.


Subject(s)
Cell-Matrix Junctions/metabolism , Focal Adhesions/metabolism , Actins/metabolism , Animals , Cell Adhesion , Cell Movement/physiology , Extracellular Matrix/metabolism , Humans , Integrins/metabolism , Models, Biological
11.
J Cell Biol ; 180(2): 427-41, 2008 Jan 28.
Article in English | MEDLINE | ID: mdl-18227284

ABSTRACT

Cell migration is an integrated process requiring the continuous coordinated assembly and disassembly of adhesion structures. How cells orchestrate adhesion turnover is only partially understood. We provide evidence for a novel mechanistic insight into focal adhesion (FA) dynamics by demonstrating that integrin cytoplasmic domain-associated protein 1 (ICAP-1) slows down FA assembly. Live cell imaging, which was performed in both Icap-1-deficient mouse embryonic fibroblasts and cells expressing active beta(1) integrin, shows that the integrin high affinity state favored by talin is antagonistically controlled by ICAP-1. This affinity switch results in modulation in the speed of FA assembly and, consequently, of cell spreading and migration. Unexpectedly, the ICAP-1-dependent decrease in integrin affinity allows cell sensing of matrix surface density, suggesting that integrin conformational changes are important in mechanotransduction. Our results clarify the function of ICAP-1 in cell adhesion and highlight the central role it plays in the cell's integrated response to the extracellular microenvironment.


Subject(s)
Extracellular Matrix/metabolism , Fibroblasts/cytology , Focal Adhesions , Integrin beta1/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Cell Movement , Cells, Cultured , Integrin beta1/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Mice , Protein Conformation , Talin/metabolism
12.
Eur J Cell Biol ; 85(3-4): 275-82, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16546571

ABSTRACT

Cell adhesion to either the extracellular matrix (ECM) or to neighboring cells is of critical importance during both physiological and pathological situations. Integrins are a large family of cell adhesion receptors composed of two non-covalently linked alpha and beta subunits. They have a well-identified dual function of mediating both firm adhesion and signaling. The short cytoplasmic domain of integrin can interact with cytoplasmic proteins that are either shared by several different integrins or specific for one type of integrin. Integrin cytoplasmic domain-associated protein-1 (ICAP-1) is a small cytoplasmic protein that specifically interacts with the beta1 integrin subunit. In this review we will discuss recent findings on ICAP-1, not only at the structural and functional level, but also its possible interconnection in other signaling pathways such as those that control cell proliferation.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Cell Proliferation , Gene Expression Regulation , Humans , Integrins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Models, Biological , Protein Binding , Protein Structure, Tertiary , Signal Transduction/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...