Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 295(39): 13474-13487, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32690605

ABSTRACT

Yes-associated protein (YAP) signaling has emerged as a crucial pathway in several normal and pathological processes. Although the main upstream effectors that regulate its activity have been extensively studied, the role of the endosomal system has been far less characterized. Here, we identified the late endosomal/lysosomal adaptor MAPK and mTOR activator (LAMTOR) complex as an important regulator of YAP signaling in a preosteoblast cell line. We found that p18/LAMTOR1-mediated peripheral positioning of late endosomes allows delivery of SRC proto-oncogene, nonreceptor tyrosine kinase (SRC) to the plasma membrane and promotes activation of an SRC-dependent signaling cascade that controls YAP nuclear shuttling. Moreover, ß1 integrin engagement and mechano-sensitive cues, such as external stiffness and related cell contractility, controlled LAMTOR targeting to the cell periphery and thereby late endosome recycling and had a major impact on YAP signaling. Our findings identify the late endosome recycling pathway as a key mechanism that controls YAP activity and explains YAP mechano-sensitivity.


Subject(s)
Cell Cycle Proteins/metabolism , Endosomes/metabolism , Integrin beta1/metabolism , Transcription Factors/metabolism , src-Family Kinases/metabolism , Animals , Cell Cycle Proteins/deficiency , Cell Line , HEK293 Cells , Humans , Mice , Mice, Knockout , Proto-Oncogene Mas , Signal Transduction , Transcription Factors/deficiency , src-Family Kinases/deficiency
2.
PLoS One ; 13(4): e0196021, 2018.
Article in English | MEDLINE | ID: mdl-29677202

ABSTRACT

Osteoblast differentiation is a highly regulated process that requires coordinated information from both soluble factors and the extracellular matrix. Among these extracellular stimuli, chemical and physical properties of the matrix are sensed through cell surface receptors such as integrins and transmitted into the nucleus to drive specific gene expression. Here, we showed that the conditional deletion of ß1 integrins in the osteo-precursor population severely impacts bone formation and homeostasis both in vivo and in vitro. Mutant mice displayed a severe bone deficit characterized by bone fragility and reduced bone mass. We showed that ß1 integrins are required for proper BMP2 dependent signaling at the pre-osteoblastic stage, by positively modulating Smad1/5-dependent transcriptional activity at the nuclear level. The lack of ß1 integrins results in a transcription modulation that relies on a cooperative defect with other transcription factors rather than a plain blunted BMP2 response. Our results point to a nuclear modulation of Smad1/5 transcriptional activity by ß1 integrins, allowing a tight control of osteoblast differentiation.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Integrin beta1/genetics , Osteoblasts/cytology , Osteogenesis , Smad1 Protein/genetics , Smad5 Protein/genetics , Animals , Cell Differentiation , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cells, Cultured , Gene Expression Regulation , Gene Knockout Techniques , Homeostasis , Mice , Osteoblasts/metabolism , Signal Transduction , Transcription, Genetic
3.
J Biol Chem ; 292(47): 19179-19197, 2017 11 24.
Article in English | MEDLINE | ID: mdl-28972170

ABSTRACT

Cell adhesion to the extracellular matrix or to surrounding cells plays a key role in cell proliferation and differentiation and is critical for proper tissue homeostasis. An important pathway in adhesion-dependent cell proliferation is the Hippo signaling cascade, which is coregulated by the transcription factors Yes-associated protein 1 (YAP1) and transcriptional coactivator with PDZ-binding motif (TAZ). However, how cells integrate extracellular information at the molecular level to regulate YAP1's nuclear localization is still puzzling. Herein, we investigated the role of ß1 integrins in regulating this process. We found that ß1 integrin-dependent cell adhesion is critical for supporting cell proliferation in mesenchymal cells both in vivo and in vitro ß1 integrin-dependent cell adhesion relied on the relocation of YAP1 to the nucleus after the down-regulation of its phosphorylated state mediated by large tumor suppressor gene 1 and 2 (LATS1/2). We also found that this phenotype relies on ß1 integrin-dependent local activation of the small GTPase RAC1 at the plasma membrane to control the activity of P21 (RAC1)-activated kinase (PAK) of group 1. We further report that the regulatory protein merlin (neurofibromin 2, NF2) interacts with both YAP1 and LATS1/2 via its C-terminal moiety and FERM domain, respectively. PAK1-mediated merlin phosphorylation on Ser-518 reduced merlin's interactions with both LATS1/2 and YAP1, resulting in YAP1 dephosphorylation and nuclear shuttling. Our results highlight RAC/PAK1 as major players in YAP1 regulation triggered by cell adhesion.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Genes, Neurofibromatosis 2/physiology , Integrin beta1/physiology , Neurofibromin 2/metabolism , Phosphoproteins/metabolism , p21-Activated Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Adhesion , Cell Cycle Proteins , Cell Proliferation , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Mice , Mice, Knockout , Neurofibromin 2/genetics , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins , p21-Activated Kinases/genetics , rac1 GTP-Binding Protein/genetics
4.
PLoS One ; 10(10): e0141205, 2015.
Article in English | MEDLINE | ID: mdl-26509443

ABSTRACT

We previously reported the anti-migratory function of 3-aryl-2-quinolone derivatives, chemically close to flavonoids (Joseph et al., 2002). Herein we show that 3-arylquinoline or 3-aryl-2-quinolone derivatives disrupt cell adhesion in a dose dependent and reversible manner yet antagonized by artificial integrin activation such as manganese. Relying on this anti-adhesive activity, a Structure-Activity Relationship (SAR) study was established on 20 different compounds to throw the bases of future optimization strategies. Active drugs efficiently inhibit platelet spreading, aggregation, and clot retraction, processes that rely on αllbß3 integrin activation and clustering. In vitro these derivatives interfere with ß3 cytoplasmic tail interaction with kindlin-2 in pulldown assays albeit little effect was observed with pure proteins suggesting that the drugs may block an alternative integrin activation process that may not be directly related to kindlin recruitment. Ex vivo, these drugs blunt integrin signaling assayed using focal adhesion kinase auto-phosphorylation as a read-out. Hence, 3-arylquinoline and 3-aryl-2-quinolone series are a novel class of integrin activation and signaling antagonists.


Subject(s)
Integrins/metabolism , Quinolones/metabolism , Animals , Cattle , Cell Adhesion/drug effects , Cell Line , Humans , Manganese/pharmacology , Quinolones/pharmacology , Signal Transduction/drug effects , Structure-Activity Relationship
5.
Sci Rep ; 5: 14532, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26459014

ABSTRACT

Video microscopy offers outstanding capabilities to investigate the dynamics of biological and pathological mechanisms in optimal culture conditions. Contact imaging is one of the simplest imaging architectures to digitally record images of cells due to the absence of any objective between the sample and the image sensor. However, in the framework of in-line holography, other optical components, e.g., an optical filter or a pinhole, are placed underneath the light source in order to illuminate the cells with a coherent or quasi-coherent incident light. In this study, we demonstrate that contact imaging with an incident light of both limited temporal and spatial coherences can be achieved with sufficiently high quality for most applications in cell biology, including monitoring of cell sedimentation, rolling, adhesion, spreading, proliferation, motility, death and detachment. Patterns of cells were recorded at various distances between 0 and 1000 µm from the pixel array of the image sensors. Cells in suspension, just deposited or at mitosis focalise light into photonic nanojets which can be visualised by contact imaging. Light refraction by cells significantly varies during the adhesion process, the cell cycle and among the cell population in connection with every modification in the tridimensional morphology of a cell.


Subject(s)
Microscopy, Video/methods , Time-Lapse Imaging/methods , Cell Adhesion , Cell Culture Techniques , Cell Movement , Cell Proliferation , Cells, Cultured , Humans
6.
Biomacromolecules ; 16(3): 715-22, 2015 Mar 09.
Article in English | MEDLINE | ID: mdl-25629300

ABSTRACT

In this work, cell-responsive polysaccharide hydrogels were prepared by a simple procedure based on the sequential bioconjugation and cross-linking of the polysaccharide backbone with bioactive peptides and poly(ethylene glycol)-bis(thiol) (PEG-(SH)2), respectively. Using thiol-ene reactions, we successfully functionalized hyaluronic acid (HA) and carboxymethylcellulose (CMC) with short and long peptides (5-mer and 15-mer derivatives, respectively) derived from adhesive proteins of bone extracellular matrix. The resulting HA-peptide and CMC-peptide conjugates with varying degrees of substitution were then carefully characterized by (1)H NMR spectroscopy to precisely control the peptide density into the hydrogels cross-linked with PEG-(SH)2. Preosteoblast seeded on the hydrogels with controlled identical stiffness spread in a manner that was strongly dependent on ligand density. Surprisingly, increasing the density of the adhesive peptide anchors did not result in a plateau of initial cell spreading but rather in a bell-shaped cell response that varies with the nature of both polysaccharide backbone and functional peptide. Placing the cells under optimal conditions for cell/hydrogel interaction, we showed that in HA hydrogels, the polysaccharide moiety is not solely a passive scaffold that presents the active peptides but is an active player in cell microenvironment to control and sustain cell activity.


Subject(s)
Carboxymethylcellulose Sodium/chemistry , Cell Differentiation , Fibronectins/chemistry , Hyaluronic Acid/chemistry , Integrin-Binding Sialoprotein/chemistry , Osteoblasts/physiology , Animals , Cell Adhesion , Cell Proliferation , Cells, Cultured , Culture Media , Hydrogels , Mice , Osteogenesis , Peptide Fragments/chemistry
7.
Int Rev Cell Mol Biol ; 305: 1-68, 2013.
Article in English | MEDLINE | ID: mdl-23890379

ABSTRACT

Mineralized tissues that are protective scaffolds in the most primitive species have evolved and acquired more specific functions in modern animals. These are as diverse as support in locomotion, ion homeostasis, and precise hormonal regulation. Bone formation is tightly controlled by a balance between anabolism, in which osteoblasts are the main players, and catabolism mediated by the osteoclasts. The bone matrix is deposited in a cyclic fashion during homeostasis and integrates several environmental cues. These include diffusible elements that would include estrogen or growth factors and physicochemical parameters such as bone matrix composition, stiffness, and mechanical stress. Therefore, the microenvironment is of paramount importance for controlling this delicate equilibrium. Here, we provide an overview of the most recent data highlighting the role of cell-adhesion molecules during bone formation. Due to the very large scope of the topic, we focus mainly on the role of the integrin receptor family during osteogenesis. Bone phenotypes of some deficient mice as well as diseases of human bones involving cell adhesion during this process are discussed in the context of bone physiology.


Subject(s)
Bone and Bones/metabolism , Osteogenesis , Signal Transduction , Animals , Bone and Bones/cytology , Cell Adhesion , Humans , Integrins/metabolism
8.
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
9.
Biomacromolecules ; 13(6): 1818-27, 2012 Jun 11.
Article in English | MEDLINE | ID: mdl-22559074

ABSTRACT

Hyaluronic acid (HA) is a natural polysaccharide abundant in biological tissues with excellent potential for constructing synthetic extracellular matrix analogues. In this work, we established a simple and dependable approach to prepare hyaluronic acid-based hydrogels with controlled stiffness and cell recognition properties for use as cell-interactive substrates. This approach relied on a new procedure for the synthesis of methacrylate-modified HA macromers (HA-MA) and, on photorheometry allowing real time monitoring of gelation during photopolymerization. We showed in this way the ability to obtain gels that encompass the range of physiologically relevant elastic moduli while still maintaining the recognition properties of HA by specific cell surface receptors. These hydrogels were prepared from HA macromers having a degree of methacrylation <0.5, which allows to minimize compromising effects on the binding affinity of HA to its cell receptors due to high substitution on the one hand, and to achieve nearly 100% conversion of the methacrylate groups on the other. When the HA hydrogels were immobilized on glass substrates, it was observed that the attachment and the spreading of a variety of mammalian cells rely on CD44 and its coreceptor RHAMM. The attachment and spreading were also shown to be modulated by the elastic properties of the HA matrix. All together, these results highlight the biological potential of these HA hydrogel systems and the needs of controlling their chemical and physical properties for applications in cell culture and tissue engineering.


Subject(s)
Biomimetic Materials/chemical synthesis , Hyaluronic Acid/chemistry , Hydrogels/chemical synthesis , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/metabolism , Cells, Cultured , Hyaluronic Acid/metabolism , Hyaluronoglucosaminidase/chemistry , Hyaluronoglucosaminidase/metabolism , Hydrogels/chemistry , Hydrogels/metabolism , Macromolecular Substances/chemical synthesis , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Mice , NIH 3T3 Cells , Surface Properties
10.
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
11.
Curr Opin Cell Biol ; 23(5): 597-606, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21550788

ABSTRACT

Invadosomes are adhesive mechanosensory modules composed of a dense F-actin core surrounded by a ring of adhesion molecules and able to infiltrate compact tissue environment in physiological and pathological conditions. These structures comprise podosomes that are found in a variety of cells under physiological conditions and invadopodia in transformed or cancer cells. Invadosomes are regulated by extracellular matrix signals and are endowed with degradative machinery for extracellular matrix. The ability of extracellular matrix signals to orchestrate the building, dynamics, and function of invadosomes is based on mechano-chemical integrin outside-in signaling and requires integrin cross-talk. This review highlights recent findings that place Src as an inducer and PKC as an amplifier in the assembly of integrin stimulated invadosome through mechanotransduction and polarized endo/exocytic trafficking pathways for key proteolytic and enzymatic activities in a temporally and spatially confined manner.


Subject(s)
Cell Adhesion Molecules/metabolism , Mechanotransduction, Cellular , Actins/metabolism , Animals , Cell Movement , Cytoskeleton/metabolism , Extracellular Matrix/metabolism , Humans , Integrins/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism
12.
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
13.
Mol Biol Cell ; 21(23): 4108-19, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20926684

ABSTRACT

Invadosomes are adhesion structures involved in tissue invasion that are characterized by an intense actin polymerization-depolymerization associated with ß1 and ß3 integrins and coupled to extracellular matrix (ECM) degradation activity. We induced the formation of invadosomes by expressing the constitutive active form of Src, SrcYF, in different cell types. Use of ECM surfaces micropatterned at the subcellular scale clearly showed that in mesenchymal cells, integrin signaling controls invadosome activity. Using ß1⁻/⁻ or ß3⁻/⁻ cells, it seemed that ß1A but not ß3 integrins are essential for initiation of invadosome formation. Protein kinase C activity was shown to regulate autoassembly of invadosomes into a ring-like metastructure (rosette), probably by phosphorylation of Ser785 on the ß1A tail. Moreover, our study clearly showed that ß1A links actin dynamics and ECM degradation in invadosomes. Finally, a new strategy based on fusion of the photosensitizer KillerRed to the ß1A cytoplasmic domain allowed specific and immediate loss of function of ß1A, resulting in disorganization and disassembly of invadosomes and formation of focal adhesions.


Subject(s)
Actins/metabolism , Cell Adhesion Molecules/metabolism , Cell Adhesion , Cell Membrane Structures/physiology , Integrin beta1/metabolism , Animals , Cell Membrane Structures/ultrastructure , Cell Movement , Cells, Cultured , Extracellular Matrix/metabolism , Focal Adhesions/metabolism , Gene Knockout Techniques , Genes, src , Integrin beta1/chemistry , Integrin beta3/metabolism , Mesoderm/metabolism , Mice , Phosphorylation , Polymerization , Protein Kinase C/metabolism , Signal Transduction
15.
J Cell Sci ; 122(Pt 17): 3037-49, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19692590

ABSTRACT

The invasiveness of cells is correlated with the presence of dynamic actin-rich membrane structures called invadopodia, which are membrane protrusions that are associated with localized polymerization of sub-membrane actin filaments. Similar to focal adhesions and podosomes, invadopodia are cell-matrix adhesion sites. Indeed, invadopodia share several features with podosomes, but whether they are distinct structures is still a matter of debate. Invadopodia are built upon an N-WASP-dependent branched actin network, and the Rho GTPase Cdc42 is involved in inducing invadopodial-membrane protrusion, which is mediated by actin filaments that are organized in bundles to form an actin core. Actin-core formation is thought to be an early step in invadopodium assembly, and the actin core is perpendicular to the extracellular matrix and the plasma membrane; this contrasts with the tangential orientation of actin stress fibers anchored to focal adhesions. In this Commentary, we attempt to summarize recent insights into the actin dynamics of invadopodia and podosomes, and the forces that are transmitted through these invasive structures. Although the mechanisms underlying force-dependent regulation of invadopodia and podosomes are largely unknown compared with those of focal adhesions, these structures do exhibit mechanosensitivity. Actin dynamics and associated forces might be key elements in discriminating between invadopodia, podosomes and focal adhesions. Targeting actin-regulatory molecules that specifically promote invadopodium formation is an attractive strategy against cancer-cell invasion.


Subject(s)
Actin Cytoskeleton/chemistry , Cell Membrane Structures/chemistry , Cells/chemistry , Focal Adhesions/chemistry , Neoplasms/chemistry , Actin Cytoskeleton/metabolism , Animals , Biomechanical Phenomena , Cell Membrane Structures/metabolism , Cells/metabolism , Focal Adhesions/metabolism , Humans , Mechanotransduction, Cellular , Neoplasm Invasiveness , Neoplasms/metabolism , Neoplasms/pathology
16.
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
17.
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
18.
Exp Cell Res ; 314(3): 478-88, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18067889

ABSTRACT

Time-lapse video-microscopy unambiguously shows that fibroblast filopodia are the scaffold of lamellipodia nucleation that allows anisotropic cell spreading. This process was dissected into elementary stages by monitoring cell adhesion on micropatterned extracellular matrix arrays of various pitches. Adhesion structures are stabilized by contact with the adhesive plots and subsequently converted into lamellipodia-like extensions starting at the filopodia tips. This mechanism progressively leads to full cell spreading. Stable expression of the dominant-negative Rac1 N17 impairs this change in membrane extension mode and stops cell spreading on matrix arrays. Similar expression of the dominant-negative Cdc42 N17 impairs cell spreading on homogenous and structured substrate, suggesting that filopodia extension is a prerequisite for cell spreading in this model. The differential polarity of the nucleation of lamellipodial structures by filopodia on homogenous and structured surfaces starting from the cell body and of filopodia tip, respectively, suggested that this process is triggered by areas that are in contact with extracellular matrix proteins for longer times. Consistent with this view, wild-type cells cannot spread on microarrays made of function blocking or neutral anti-beta 1 integrin antibodies. However, stable expression of a constitutively active Rac1 mutant rescues the cell ability to spread on these integrin microarrays. Thereby, lamellipodia nucleation by filopodia requires integrin occupancy by matrix substrate and downstream Rac1 signaling.


Subject(s)
Cell Movement/physiology , Fibroblasts/metabolism , Integrins/metabolism , Pseudopodia/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Cell Adhesion/physiology , Cell Polarity/physiology , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fibroblasts/ultrastructure , Mice , Microscopy, Video/methods , Mutation/genetics , NIH 3T3 Cells , Protein Array Analysis/methods , Pseudopodia/ultrastructure , Signal Transduction/physiology , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics
19.
Mol Biol Cell ; 19(2): 633-45, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18045996

ABSTRACT

In Rous sarcoma virus (RSV)-transformed baby hamster kidney (BHK) cells, invadopodia can self-organize into rings and belts, similarly to podosome distribution during osteoclast differentiation. The composition of individual invadopodia is spatiotemporally regulated and depends on invadopodia localization along the ring section: the actin core assembly precedes the recruitment of surrounding integrins and integrin-linked proteins, whereas the loss of the actin core was a prerequisite to invadopodia disassembly. We have shown that invadopodia ring expansion is controlled by paxillin phosphorylations on tyrosine 31 and 118, which allows invadopodia disassembly. In BHK-RSV cells, ectopic expression of the paxillin mutant Y31F-Y118F induces a delay in invadopodia disassembly and impairs their self-organization. A similar mechanism is unraveled in osteoclasts by using paxillin knockdown. Lack of paxillin phosphorylation, calpain or extracellular signal-regulated kinase inhibition, resulted in similar phenotype, suggesting that these proteins belong to the same regulatory pathways. Indeed, we have shown that paxillin phosphorylation promotes Erk activation that in turn activates calpain. Finally, we observed that invadopodia/podosomes ring expansion is required for efficient extracellular matrix degradation both in BHK-RSV cells and primary osteoclasts, and for transmigration through a cell monolayer.


Subject(s)
Paxillin/metabolism , Pseudopodia/metabolism , Animals , Calpain/antagonists & inhibitors , Cell Communication/drug effects , Cell Movement/drug effects , Cell Transformation, Viral/drug effects , Cricetinae , Enzyme Activation/drug effects , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , HeLa Cells , Humans , Mice , Mutant Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Protein Kinase Inhibitors/pharmacology , Pseudopodia/drug effects , Pseudopodia/enzymology , Rous sarcoma virus/metabolism , Vanadates/pharmacology
20.
Cancer Res ; 67(20): 9781-90, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17942908

ABSTRACT

Depending on its cellular localization, p120 catenin (p120ctn) can participate in various processes, such as cadherin-dependent cell-cell adhesion, actin cytoskeleton remodeling, and intracellular trafficking. Recent studies also indicate that p120ctn could regulate cell proliferation and contact inhibition. This report describes a new function of p120ctn in the regulation of cell cycle progression. Overexpression of the p120ctn isoform 3A in human colon adenocarcinoma cells (HT-29) results in cytoplasmic accumulation of the protein, as observed in many tumors. This cytoplasmic increase is correlated with a reduction in proliferation and inhibition of DNA synthesis. Under these conditions, experiments on synchronized cells revealed a prolonged S phase associated with cyclin E stabilization. Both confocal microscopy and biochemical analysis showed that cyclin E and cyclin-dependent kinase 2 colocalized with p120ctn in centrosomes during mitosis. These proteins are associated in a functional complex evidenced by coimmunoprecipitation experiments and the emergence of Thr199-phosphorylated nucleophosmin/B23. Such post-translational modification of this centrosomal target has been shown to trigger the initiation of centrosome duplication. Therefore, p120ctn-mediated accumulation of cyclin E in centrosomes may participate in abnormal amplification of centrosomes and the inhibition of DNA replication, thus leading to aberrant mitosis and polyploidy. Because these modifications are often observed in cancer, p120ctn may represent a new therapeutic target for future therapy.


Subject(s)
Cell Adhesion Molecules/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Phosphoproteins/metabolism , Catenins , Cell Adhesion Molecules/biosynthesis , Cell Adhesion Molecules/genetics , Cell Cycle/physiology , Cell Growth Processes/physiology , Centrosome/metabolism , Colonic Neoplasms/genetics , Cytoplasm/metabolism , Disease Progression , Gene Amplification , Genomic Instability , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , HT29 Cells , Humans , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Phosphorylation , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Up-Regulation , Delta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...