Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 296: 100109, 2021.
Article in English | MEDLINE | ID: mdl-33853759

ABSTRACT

Cell-extracellular matrix (ECM) detachment is known to decrease extracellular signal-regulated kinase (ERK) signaling, an intracellular pathway that is central for control of cell behavior. How cell-ECM detachment is linked to downregulation of ERK signaling, however, is incompletely understood. We show here that focal adhesion protein Ras Suppressor 1 (RSU1) plays a critical role in cell-ECM detachment induced suppression of ERK signaling. We have identified prohibitin 2 (PHB2), a component of membrane lipid rafts, as a novel binding protein of RSU1, and mapped a major RSU1-binding site to PHB2 amino acids 150 to 206 in the C-terminal region of the PHB/SPFH (stomatin/prohibitin/flotillin/HflKC) domain. The PHB2 binding is mediated by multiple sites located in the N-terminal leucine-rich repeat region of RSU1. Depletion of PHB2 suppressed cell-ECM adhesion-induced ERK activation. Furthermore, cell-ECM detachment increased RSU1 association with membrane lipid rafts and interaction with PHB2. Finally, knockout of RSU1 or inhibition of RSU1 interaction with PHB2 by overexpression of the major RSU1-binding PHB2 fragment (amino acids 150-206) effectively suppressed the cell-ECM detachment induced downregulation of ERK signaling. Additionally, expression of venus-tagged wild-type RSU1 restored ERK signaling, while expression of venus-tagged PHB2-binding defective RSU1 mutant in which the N-terminal leucine-rich repeat region is deleted did not. Taken together, Our findings identify a novel RSU1-PHB2 signaling axis that senses cell-ECM detachment and links it to decreased ERK signaling.


Subject(s)
Down-Regulation , Extracellular Matrix/metabolism , MAP Kinase Signaling System , Repressor Proteins/metabolism , Transcription Factors/metabolism , Cell Adhesion/genetics , Cell Line, Tumor , Extracellular Matrix/genetics , Humans , Prohibitins , Repressor Proteins/genetics
2.
J Biol Chem ; 289(45): 31001-13, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25237194

ABSTRACT

Integrin-mediated cell-extracellular matrix (ECM) adhesion is critical for control of intracellular signaling; however, the mechanisms underlying this "outside-in" signaling are incompletely understood. Here we show that depletion of kindlin-2 impairs integrin outside-in signaling. Kindlin-2 is tyrosine-phosphorylated upon cell-ECM adhesion. Furthermore, kindlin-2 binds Src in a cell-ECM adhesion-regulatable fashion. At the molecular level, the kindlin-2·Src interaction is mediated by the kindlin-2 F0 and the Src SH2 and SH3 domains. Src activation increases kindlin-2 tyrosine phosphorylation and the kindlin-2·Src interaction. Conversely, inhibition of Src reduces kindlin-2 tyrosine phosphorylation and diminishes the kindlin-2·Src interaction. Finally, disruption of the kindlin-2·Src interaction, unlike depletion of kindlin-2, impairs neither cell-ECM adhesion nor cell-ECM adhesion-induced focal adhesion kinase Tyr-397 phosphorylation. However, it markedly inhibits cell-ECM adhesion-induced paxillin tyrosine phosphorylation, cell migration, and proliferation. These results suggest that kindlin-2 tyrosine phosphorylation and interaction with Src serve as a regulatable switch downstream of focal adhesion kinase in the integrin outside-in signaling circuit, relaying signals from cell-ECM adhesion to paxillin that control cell migration and proliferation.


Subject(s)
Cytoskeletal Proteins/metabolism , Integrins/metabolism , Membrane Proteins/metabolism , Muscle Proteins/metabolism , Neoplasm Proteins/metabolism , src-Family Kinases/metabolism , Animals , CSK Tyrosine-Protein Kinase , Cell Adhesion , Cell Movement , Cell Proliferation , Extracellular Matrix/metabolism , Fibroblasts/cytology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression Regulation , Humans , Kidney Glomerulus/metabolism , Mice , Paxillin/metabolism , Phosphorylation , Podocytes/cytology , Protein Binding , Recombinant Proteins/metabolism , Signal Transduction , Tyrosine , src Homology Domains
3.
J Biol Chem ; 287(25): 21450-60, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22556421

ABSTRACT

Bone remodeling is a complex process that must be precisely controlled to maintain a healthy life. We show here that filamin-binding LIM protein 1 (FBLP-1, also known as migfilin), a kindlin- and filamin-binding focal adhesion protein, is essential for proper control of bone remodeling. Genetic inactivation of FBLIM1 (the gene encoding FBLP-1) in mice resulted in a severe osteopenic phenotype. Primary FBLP-1 null bone marrow stromal cells (BMSCs) exhibited significantly reduced extracellular matrix adhesion and migration compared with wild type BMSCs. Loss of FBLP-1 significantly impaired the growth and survival of BMSCs in vitro and decreased the number of osteoblast (OB) progenitors in bone marrow and OB differentiation in vivo. Furthermore, the loss of FBLP-1 caused a dramatic increase of osteoclast (OCL) differentiation in vivo. The level of receptor activator of nuclear factor κB ligand (RANKL), a key regulator of OCL differentiation, was markedly increased in FBLP-1 null BMSCs. The capacity of FBLP-1 null bone marrow monocytes (BMMs) to differentiate into multinucleated OCLs in response to exogenously supplied RANKL, however, was not different from that of WT BMMs. Finally, we show that a loss of FBLP-1 promotes activating phosphorylation of ERK1/2. Inhibition of ERK1/2 activation substantially suppressed the increase of RANKL induced by the loss of FBLP-1. Our results identify FBLP-1 as a key regulator of bone homeostasis and suggest that FBLP-1 functions in this process through modulating both the intrinsic properties of OB/BMSCs (i.e., BMSC-extracellular matrix adhesion and migration, cell growth, survival, and differentiation) and the communication between OB/BMSCs and BMMs (i.e., RANKL expression) that controls osteoclastogenesis.


Subject(s)
Bone Remodeling/physiology , Cell Adhesion Molecules/metabolism , Cell Differentiation/physiology , Cytoskeletal Proteins/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Adhesion/physiology , Cell Adhesion Molecules/genetics , Cell Movement/physiology , Cytoskeletal Proteins/genetics , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Monocytes/cytology , Monocytes/metabolism , Osteoblasts/cytology , Osteoclasts/cytology , Phosphorylation/physiology , Stromal Cells/cytology , Stromal Cells/metabolism
4.
PLoS One ; 6(11): e27789, 2011.
Article in English | MEDLINE | ID: mdl-22110760

ABSTRACT

The Non-structural 1 (NS1) protein of avian influenza (AI) viruses is important for pathogenicity. Here, we identify a previously unrecognized tandem PDZ-ligand (TPL) domain in the extreme carboxy terminus of NS1 proteins from a subset of globally circulating AI viruses. By using protein arrays we have identified several human PDZ-cellular ligands of this novel domain, one of which is the RIL protein, a known regulator of the cellular tyrosine kinase Src. We found that the AI NS1 proteins bind and stimulate human Src tyrosine kinase, through their carboxy terminal Src homology type 3-binding (SHB) domain. The physical interaction between NS1 and Src and the ability of AI viruses to modulate the phosphorylation status of Src during the infection, were found to be influenced by the TPL arrangement. These results indicate the potential for novel host-pathogen interactions mediated by the TPL and SHB domains of AI NS1 protein.


Subject(s)
Epidemics , Influenza in Birds/virology , Protein Interaction Domains and Motifs , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , src-Family Kinases/metabolism , Amino Acid Sequence , Animals , Birds , Cell Line , DNA-Binding Proteins/metabolism , Humans , Influenza A Virus, H7N1 Subtype , LIM Domain Proteins/metabolism , Ligands , Molecular Sequence Data , PDZ Domains , Protein Array Analysis , src Homology Domains
5.
J Biol Chem ; 286(35): 30795-30805, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21680739

ABSTRACT

Genetic alterations of α-actinin-4 can cause podocyte injury through multiple mechanisms. Although a mechanism involving gain-of-α-actinin-4 function was well described and is responsible for a dominantly inherited form of human focal segmental glomerulosclerosis (FSGS), evidence supporting mechanisms involving loss-of-α-actinin-4 function in human glomerular diseases remains elusive. Here we show that α-actinin-4 deficiency occurs in multiple human primary glomerulopathies including sporadic FSGS, minimal change disease, and IgA nephropathy. Furthermore, we identify a close correlation between the levels of α-actinin-4 and CLP36, which form a complex in normal podocytes, in human glomerular diseases. siRNA-mediated depletion of α-actinin-4 in human podocytes resulted in a marked reduction of the CLP36 level. Additionally, two FSGS-associated α-actinin-4 mutations (R310Q and Q348R) inhibited the complex formation between α-actinin-4 and CLP36. Inhibition of the α-actinin-4-CLP36 complex, like loss of α-actinin-4, markedly reduced the level of CLP36 in podocytes. Finally, reduction of the CLP36 level or disruption of the α-actinin-4-CLP36 complex significantly inhibited RhoA activity and generation of traction force in podocytes. Our studies reveal a critical role of the α-actinin-4-CLP36 complex in podocytes and provide an explanation as to how α-actinin-4 deficiency or mutations found in human patients could contribute to podocyte defects and glomerular failure through a loss-of-function mechanism.


Subject(s)
Actinin/genetics , Kidney Glomerulus/metabolism , Microfilament Proteins/genetics , Podocytes/metabolism , Actinin/deficiency , Animals , Biopsy , Detergents/pharmacology , Glomerulonephritis, IGA/metabolism , Glomerulosclerosis, Focal Segmental/metabolism , Humans , Immunohistochemistry/methods , Kidney/metabolism , LIM Domain Proteins , Mice , Microfilament Proteins/deficiency , Mutation , Protein Interaction Mapping/methods , Proteinuria/metabolism , Transcription Factors
6.
J Cell Sci ; 124(Pt 6): 879-91, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21325030

ABSTRACT

Kindlin-2 is a FERM and PH domain-containing integrin-binding protein that is emerging as an important regulator of integrin activation. How kindlin-2 functions in integrin activation, however, is not known. We report here that kindlin-2 interacts with multiple phosphoinositides, preferentially with phosphatidylinositol 3,4,5-trisphosphate. Although integrin-binding is essential for focal adhesion localization of kindlin-2, phosphoinositide-binding is not required for this process. Using biologically and clinically relevant glomerular podocytes as a model system, we show that integrin activation and dependent processes are tightly regulated by kindlin-2: depletion of kindlin-2 reduced integrin activation, matrix adhesion and fibronectin matrix deposition, whereas overexpression of kindlin-2 promoted these processes. Furthermore, we provide evidence showing that kindlin-2 is involved in phosphoinositide-3-kinase-mediated regulation of podocyte-matrix adhesion and fibronectin matrix deposition. Mechanistically, kindlin-2 promotes integrin activation and integrin-dependent processes through interacting with both integrins and phosphoinositides. TGF-ß1, a mediator of progressive glomerular failure, markedly increased the level of kindlin-2 and fibronectin matrix deposition, and the latter process was reversed by depletion of kindlin-2. Our results reveal important functions of kindlin-2 in the regulation of podocyte-matrix adhesion and matrix deposition and shed new light on the mechanism whereby kindlin-2 functions in these processes.


Subject(s)
Fibronectins/metabolism , Integrin beta1/metabolism , Integrin beta3/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Phosphatidylinositols/metabolism , Podocytes/cytology , Podocytes/metabolism , Cell Adhesion , Cell Line , Extracellular Matrix/chemistry , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Fibronectins/genetics , Humans , Integrin beta1/genetics , Integrin beta3/genetics , Kidney Glomerulus/cytology , Kidney Glomerulus/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Podocytes/chemistry , Protein Binding , Protein Structure, Tertiary , Transforming Growth Factor beta1/metabolism
7.
J Biol Chem ; 284(49): 34308-20, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19833732

ABSTRACT

Integrin-mediated cell-extracellular matrix (ECM) adhesion is essential for protection of epithelial cells against apoptosis, but the underlying mechanism is incompletely understood. Here we show that migfilin, an integrin-proximal adaptor protein, interacts with Src and contributes to cell-ECM-mediated survival signaling. Loss of cell-ECM adhesion markedly reduces the migfilin level in untransformed epithelial cells and concomitantly induces apoptosis. Overexpression of migfilin substantially desensitizes cell detachment-induced apoptosis. Conversely, depletion of migfilin promotes apoptosis despite the presence of cell-ECM adhesion. At the molecular level migfilin directly interacts with Src, and the migfilin binding surface overlaps with the inhibitory intramolecular interaction sites in Src. Consequently, the binding of migfilin activates Src, resulting in suppression of apoptosis. Our results reveal a novel mechanism by which cell-ECM adhesion regulates Src activation and survival signaling. This migfilin-mediated signaling pathway is dysfunctional in multiple types of carcinoma cells, which likely contributes to aberrant Src activation and anoikis resistance in the cancerous cells.


Subject(s)
Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Cytoskeletal Proteins/metabolism , Gene Expression Regulation , src-Family Kinases/metabolism , Anoikis , Apoptosis , Cell Adhesion , Cell Line, Tumor , Cell Survival , Cytoplasm/metabolism , Epithelial Cells/cytology , Extracellular Matrix/metabolism , Glutathione Transferase/metabolism , Humans , Magnetic Resonance Spectroscopy , RNA Interference , Signal Transduction
8.
Eur J Oral Sci ; 117(5): 518-27, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19758247

ABSTRACT

Kindlin-1 is an intracellular focal adhesion protein that regulates the actin cytoskeleton. Patients suffering from Kindler syndrome have a homologous mutation of the kindlin-1 gene and develop skin blisters, periodontal disease, and intestinal complications because of deficient adhesion of the basal epithelial cells. We investigated kindlin-1 localization in periodontal tissue and its functions in cultured keratinocytes and showed that kindlin-1 co-localizes with migfilin and paxillin in the basal epithelial cells of oral mucosa and in cultured keratinocytes. The kindlin-1-deficient oral mucosal tissue from a patient with Kindler syndrome showed a complete lack of paxillin and reduced migfilin immunostaining in the basal keratinocytes. Co-immunoprecipitation showed that migfilin directly interacted with kindlin-1. RNA interference-induced kindlin-1 deficiency in keratinocytes led to an altered distribution of migfilin-containing focal adhesions, reduced cell spreading, decreased cell proliferation, and decelerated cell migration. Disruption of microtubules in the kindlin-1-deficient cells further reduced cell spreading, suggesting that microtubules can partially compensate for kindlin-1 deficiency. Kindlin-1 supported mature cell-extracellular matrix adhesions of keratinocytes, as downregulation of kindlin-1 expression significantly reduced the cell-adhesion strength. In summary, kindlin-1 interacts with migfilin and plays a crucial role in actin-dependent keratinocyte cell adhesion essential for epidermal and periodontal health.


Subject(s)
Membrane Proteins/analysis , Neoplasm Proteins/analysis , Periodontium/pathology , Cell Adhesion/physiology , Cell Adhesion Molecules/analysis , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation , Cytoskeletal Proteins/analysis , Epithelial Cells/pathology , Extracellular Matrix/ultrastructure , Focal Adhesions/ultrastructure , Humans , Intestinal Diseases/genetics , Keratinocytes/pathology , Membrane Proteins/genetics , Membrane Proteins/physiology , Microtubules/ultrastructure , Mouth Mucosa/pathology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Paxillin/analysis , Periodontal Diseases/genetics , Protein Serine-Threonine Kinases/analysis , RNA Interference , Skin Diseases, Genetic/pathology , Skin Diseases, Vesiculobullous/genetics , Syndrome
9.
J Cell Biol ; 184(6): 785-92, 2009 Mar 23.
Article in English | MEDLINE | ID: mdl-19307596

ABSTRACT

Aberrant Src activation plays prominent roles in cancer progression. However, how Src is activated in cancer cells is largely unknown. Genetic Src-activating mutations are rare and, therefore, are insufficient to account for Src activation commonly found in human cancers. In this study, we show that reversion-induced LIM (RIL), which is frequently lost in colon and other cancers as a result of epigenetic silencing, suppresses Src activation. Mechanistically, RIL suppresses Src activation through interacting with Src and PTPL1, allowing PTPL1-dependent dephosphorylation of Src at the activation loop. Importantly, the binding of RIL to Src is drastically reduced upon Src inactivation. Our results reveal a novel Src inactivation cycle in which RIL preferentially recognizes active Src and facilitates PTPL1-mediated inactivation of Src. Inactivation of Src, in turn, promotes dissociation of RIL from Src, allowing the initiation of a new Src inactivation cycle. Epigenetic silencing of RIL breaks this Src inactivation cycle and thereby contributes to aberrant Src activation in human cancers.


Subject(s)
Colonic Neoplasms/metabolism , DNA-Binding Proteins/metabolism , src-Family Kinases/metabolism , Binding Sites , Cell Proliferation , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA-Binding Proteins/genetics , Enzyme Activation , Gene Expression Regulation, Neoplastic , Gene Silencing , HCT116 Cells , Humans , LIM Domain Proteins , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 13/metabolism , Recombinant Fusion Proteins/metabolism , Transfection , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
10.
J Biol Chem ; 284(9): 5836-44, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19117955

ABSTRACT

The LIM-only adaptor PINCH (the particularly interesting cysteine- and histidine-rich protein) plays a pivotal role in the assembly of focal adhesions (FAs), supramolecular complexes that transmit mechanical and biochemical information between extracellular matrix and actin cytoskeleton, regulating diverse cell adhesive processes such as cell migration, cell spreading, and survival. A key step for the PINCH function is its localization to FAs, which depends critically on the tight binding of PINCH to integrin-linked kinase (ILK). Here we report the solution NMR structure of the core ILK.PINCH complex (28 kDa, K(D) approximately 68 nm) involving the N-terminal ankyrin repeat domain (ARD) of ILK and the first LIM domain (LIM1) of PINCH. We show that the ILK ARD exhibits five sequentially stacked ankyrin repeat units, which provide a large concave surface to grip the two contiguous zinc fingers of the PINCH LIM1. The highly electrostatic interface is evolutionally conserved but differs drastically from those of known ARD and LIM bound to other types of protein domains. Consistently mutation of a hot spot in LIM1, which is not conserved in other LIM domains, disrupted the PINCH binding to ILK and abolished the PINCH targeting to FAs. These data provide atomic insight into a novel modular recognition and demonstrate how PINCH is specifically recruited by ILK to mediate the FA assembly and cell-extracellular matrix communication.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Focal Adhesions/physiology , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Ankyrin Repeat , Binding Sites , Calorimetry , Cell Movement , Fluorescent Antibody Technique , Humans , Immunoprecipitation , LIM Domain Proteins , Magnetic Resonance Spectroscopy , Membrane Proteins , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Transfection , Zinc Fingers
11.
J Biol Chem ; 283(5): 2508-17, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18063582

ABSTRACT

Resistance to apoptosis is a hallmark of cancer cells. We report here that PINCH-1, a cytoplasmic component of cell-extracellular matrix adhesions, is required for protection of multiple types of cancer cells from apoptosis. Furthermore, using HT-1080 fibrosarcoma cells as a model system, we have investigated the signaling pathway through which PINCH-1 contributes to apoptosis resistance. Loss of PINCH-1 markedly increases the level of Bim and promotes Bim translocation to mitochondria, resulting in activation of the intrinsic apoptosis pathway. Depletion of Bim completely blocked apoptosis induced by the loss of PINCH-1. Thus, PINCH-1 contributes to apoptosis resistance through suppression of Bim. Mechanistically, PINCH-1 suppresses Bim not only transcriptionally but also post-transcriptionally. PINCH-1 promotes activating phosphorylation of Src family kinase and ERK1/2. Consistent with this, ERK1/2-mediated Ser(69) phosphorylation of Bim, a key signal for turnover of Bim, is suppressed by the removal of PINCH-1. Our results demonstrate a strong dependence of multiple types of apoptosis-resistant cancer cells on PINCH-1 and provide new insights into the molecular mechanism by which cancer cells are protected from apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/physiology , DNA-Binding Proteins/metabolism , MAP Kinase Signaling System/physiology , Membrane Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing , Apoptosis Regulatory Proteins/genetics , Base Sequence , Bcl-2-Like Protein 11 , Binding Sites , Cell Line, Tumor , DNA Primers/genetics , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Focal Adhesions/metabolism , Gene Expression Regulation, Neoplastic , Humans , LIM Domain Proteins , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mitochondria/metabolism , Models, Biological , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/genetics , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/genetics , Transfection , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism , src-Family Kinases/metabolism
12.
J Biol Chem ; 282(28): 20455-66, 2007 Jul 13.
Article in English | MEDLINE | ID: mdl-17513299

ABSTRACT

Integrin-mediated cell-matrix adhesion plays an important role in control of cell behavior. We report here that MIG-2, a widely expressed focal adhesion protein, interacts with beta1 and beta3 integrin cytoplasmic domains. Integrin binding is mediated by a single site within the MIG-2 FERM domain. Functionally, the MIG-2/integrin interaction recruits MIG-2 to focal adhesions. Furthermore, using alphaIIbbeta3 integrin-expressing Chinese hamster ovary cells, a well described model system for integrin activation, we show that MIG-2 promotes integrin activation and enhances cell-extracellular matrix adhesion. Although MIG-2 is expressed in many cell types, it is deficient in certain colon cancer cells. Expression of MIG-2, but not of an integrin binding-defective MIG-2 mutant, in MIG-2-null colon cancer cells strengthened cell-matrix adhesion, promoted focal adhesion formation, and reduced cell motility. These results suggest that the MIG-2/integrin interaction is an important element in the cellular control of integrin-mediated cell-matrix adhesion and that loss of this interaction likely contributes to high motility of colon cancer cells.


Subject(s)
Cell Movement , Colonic Neoplasms/metabolism , Extracellular Matrix/metabolism , Focal Adhesions/metabolism , Gene Expression Regulation, Neoplastic , Integrins/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , CHO Cells , Caco-2 Cells , Cell Adhesion , Cell Movement/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cricetinae , Cricetulus , Focal Adhesions/genetics , Focal Adhesions/pathology , Humans , Membrane Proteins/genetics , Mutation , Neoplasm Proteins/genetics , Protein Structure, Tertiary/genetics
13.
J Biol Chem ; 281(18): 12397-407, 2006 May 05.
Article in English | MEDLINE | ID: mdl-16531412

ABSTRACT

Cell migration is a complex process that is coordinately regulated by cell-matrix adhesion and actin cytoskeleton. We report here that migfilin, a recently identified component of cell-matrix adhesions, is a biphasic regulator of cell migration. Loss of migfilin impairs cell migration. Surprisingly, overexpression of migfilin also reduces cell migration. Molecularly, we have identified vasodilator-stimulated phosphoprotein (VASP) as a new migfilin-binding protein. The interaction is mediated by the VASP EVH1 domain and a single L104PPPPP site located within the migfilin proline-rich domain. Migfilin and VASP form a complex in both suspended and adhered cells, and in the latter, they co-localize in cell-matrix adhesions. Functionally, migfilin facilitates VASP localization to cell-matrix adhesions. Using two different approaches (VASP-binding defective migfilin mutants and small interfering RNA-mediated VASP knockdown), we show that the interaction with VASP is crucially involved in migfilin-mediated regulation of cell migration. Our results identify migfilin as an important regulator of cell migration and provide new information on the mechanism by which migfilin regulates this process.


Subject(s)
Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/physiology , Cytoskeletal Proteins/chemistry , Gene Expression Regulation , Microfilament Proteins/physiology , Phosphoproteins/physiology , Actins/chemistry , Animals , Binding Sites , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Movement , Cytoskeletal Proteins/metabolism , Dogs , HeLa Cells , Humans , Microfilament Proteins/chemistry , Mutation , Phosphoproteins/chemistry , Protein Structure, Tertiary , RNA, Small Interfering/metabolism
14.
Mol Cell ; 17(4): 513-23, 2005 Feb 18.
Article in English | MEDLINE | ID: mdl-15721255

ABSTRACT

Weak protein-protein interactions (PPIs) (K(D) > 10(-6) M) are critical determinants of many biological processes. However, in contrast to a large growing number of well-characterized, strong PPIs, the weak PPIs, especially those with K(D) > 10(-4) M, are poorly explored. Genome wide, there exist few 3D structures of weak PPIs with K(D) > 10(-4) M, and none with K(D) > 10(-3) M. Here, we report the NMR structure of an extremely weak focal adhesion complex (K(D) approximately 3 x 10(-3) M) between Nck-2 SH3 domain and PINCH-1 LIM4 domain. The structure exhibits a remarkably small and polar interface with distinct binding modes for both SH3 and LIM domains. Such an interface suggests a transient Nck-2/PINCH-1 association process that may trigger rapid focal adhesion turnover during integrin signaling. Genetic rescue experiments demonstrate that this interface is indeed involved in mediating cell shape change and migration. Together, the data provide a molecular basis for an ultraweak PPI in regulating focal adhesion dynamics during integrin signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement , Cell Shape , DNA-Binding Proteins/metabolism , Integrins/metabolism , Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Amino Acid Sequence , Animals , Autoantigens/metabolism , Binding Sites , DNA-Binding Proteins/genetics , Focal Adhesions , HeLa Cells , Humans , LIM Domain Proteins , Magnetic Resonance Spectroscopy , Membrane Proteins , Mice , Mice, Knockout , Molecular Sequence Data , Oncogene Proteins/genetics , Protein Binding , Protein Folding , Protein Interaction Mapping , Sequence Homology, Amino Acid , Signal Transduction , src Homology Domains
15.
J Cell Sci ; 118(Pt 4): 697-710, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15671069

ABSTRACT

Cell-cell junctions are essential for epithelial and endothelial tissue formation and communication between neighboring cells. We report here that migfilin, a recently identified component of cell-extracellular matrix adhesions, is recruited to cell-cell junctions in response to cadherin-mediated cell-cell adhesions. Migfilin is detected at cell-cell junctions in both epithelial and endothelial cells. It forms detergent-resistant, discrete clusters that associate with actin bundles bridging neighboring cells. Immunoelectron microscopic analyses reveal that migfilin is closely associated with beta-catenin, but not desmosomes, at cell-cell junctions. Furthermore, we show that the C-terminal LIM domains, but not its N-terminal domain, mediates migfilin localization to cell-cell junctions. The site mediating the localization of migfilin to cell-cell junctions at least partially overlaps with that mediating the localization of migfilin to cell-ECM adhesions. Finally, siRNA-mediated depletion of migfilin compromised the organization of adherens junctions and weakened cell-cell association. These results identify migfilin as a component of adherens junctions and suggest an important role for migfilin in the organization of the cell-cell adhesion structure.


Subject(s)
Cell Adhesion Molecules/analysis , Cell Adhesion , Actin Cytoskeleton/chemistry , Cadherins/metabolism , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/physiology , Cytoskeletal Proteins/metabolism , Endothelial Cells/chemistry , Epithelial Cells/chemistry , Extracellular Matrix/chemistry , Humans , Intercellular Junctions/chemistry , Intercellular Junctions/ultrastructure , Microscopy, Immunoelectron , Protein Structure, Tertiary , Trans-Activators/metabolism , beta Catenin
16.
J Biol Chem ; 279(40): 41695-705, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15284246

ABSTRACT

Proteins at cell-extracellular matrix adhesions (e.g. focal adhesions) are crucially involved in regulation of cell morphology and survival. We show here that CH-ILKBP/actopaxin/alpha-parvin and affixin/beta-parvin (abbreviated as alpha- and beta-parvin, respectively), two structurally closely related integrin-linked kinase (ILK)-binding focal adhesion proteins, are co-expressed in human cells. Depletion of alpha-parvin dramatically increased the level of beta-parvin, suggesting that beta-parvin is negatively regulated by alpha-parvin in human cells. Loss of PINCH-1 or ILK, to which alpha- and beta-parvin bind, significantly reduced the activation of Rac, a key signaling event that controls lamellipodium formation and cell spreading. We were surprised to find that loss of alpha-parvin, but not that of beta-parvin, markedly stimulated Rac activation and enhanced lamellipodium formation. Overexpression of beta-parvin, however, was insufficient for stimulation of Rac activation or lamellipodium formation, although it was sufficient for promotion of apoptosis, another important cellular process that is regulated by PINCH-1, ILK, and alpha-parvin. In addition, we show that the interactions of ILK with alpha- and beta-parvin are mutually exclusive. Overexpression of beta-parvin or its CH(2) fragment, but not a CH(2) deletion mutant, inhibited the ILK-alpha-parvin complex formation. Finally, we provide evidence suggesting that inhibition of the ILK-alpha-parvin complex is sufficient, although not necessary, for promotion of apoptosis. These results identify Rac as a downstream target of PINCH-1, ILK, and parvin. Furthermore, they demonstrate that alpha- and beta-parvins play distinct roles in mammalian cells and suggest that the formation of the ILK-alpha-parvin complex is crucial for protection of cells from apoptosis.


Subject(s)
Actinin/physiology , Apoptosis , Cell Surface Extensions , Focal Adhesions/metabolism , Adaptor Proteins, Signal Transducing , Cell Size , Cell Survival , DNA-Binding Proteins , Gene Expression Regulation , HeLa Cells , Humans , LIM Domain Proteins , Membrane Proteins , Microfilament Proteins , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Transfection , rac GTP-Binding Proteins/metabolism
17.
Cell ; 113(1): 37-47, 2003 Apr 04.
Article in English | MEDLINE | ID: mdl-12679033

ABSTRACT

Cell-extracellular matrix adhesion is an important determinant of cell morphology. We show here that migfilin, a LIM-containing protein, localizes to cell-matrix adhesions, associates with actin filaments, and is essential for cell shape modulation. Migfilin interacts with the cell-matrix adhesion protein Mig-2 (mitogen inducible gene-2), a mammalian homolog of UNC-112, and the actin binding protein filamin through its C- and N-terminal domains, respectively. Loss of Mig-2 or migfilin impairs cell shape modulation. Mig-2 recruits migfilin to cell-matrix adhesions, while the interaction with filamin mediates the association of migfilin with actin filaments. Migfilin therefore functions as an important scaffold at cell-matrix adhesions. Together, Mig-2, migfilin and filamin define a connection between cell matrix adhesions and the actin cytoskeleton and participate in the orchestration of actin assembly and cell shape modulation.


Subject(s)
Actin Cytoskeleton/metabolism , Caenorhabditis elegans Proteins/metabolism , Cell Adhesion Molecules/isolation & purification , Contractile Proteins/metabolism , Eukaryotic Cells/metabolism , Extracellular Matrix/metabolism , Focal Adhesions/metabolism , Microfilament Proteins/metabolism , rac GTP-Binding Proteins/metabolism , Amino Acid Sequence/genetics , Animals , Base Sequence/genetics , CHO Cells , Caenorhabditis elegans Proteins/genetics , Cell Adhesion Molecules/genetics , Cricetinae , Cytoskeletal Proteins , Cytoskeleton/metabolism , DNA, Complementary/analysis , DNA, Complementary/genetics , Filamins , Focal Adhesions/genetics , Humans , Mice , Molecular Sequence Data , Rats , Tumor Cells, Cultured , rac GTP-Binding Proteins/genetics
18.
J Cell Sci ; 115(Pt 24): 4777-86, 2002 Dec 15.
Article in English | MEDLINE | ID: mdl-12432066

ABSTRACT

PINCH, integrin-linked kinase (ILK) and calponin homology-containing ILK-binding protein (CH-ILKBP) form a ternary complex that plays crucial roles at cell-extracellular matrix adhesion sites. To understand the mechanism underlying the complex formation and recruitment to cell-adhesion sites we have undertaken a combined structural, mutational and cell biological analysis. Three-dimensional structure-based point mutations identified specific PINCH and ILK sites that mediate the complex formation. Analyses of the binding defective point mutants revealed that the assembly of the PINCH-ILK-CH-ILKBP complex is essential for their localization to cell-extracellular matrix adhesion sites. The formation of the PINCH-ILK-CH-ILKBP complex precedes integrin-mediated cell adhesion and spreading. Furthermore, inhibition of protein kinase C, but not that of actin polymerization, inhibited the PINCH-ILK-CH-ILKBP complex formation, suggesting that the PINCH-ILK-CH-ILKBP complex likely serves as a downstream effector of protein kinase C in the cellular control of focal adhesion assembly. Finally, we provide evidence that the formation of the PINCH-ILK-CH-ILKBP complex, while necessary, is not sufficient for ILK localization to cell-extracellular matrix adhesion sites. These results provide new insights into the molecular mechanism underlying the assembly and regulation of cell-matrix adhesion structures.


Subject(s)
Cell Adhesion/physiology , DNA-Binding Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Adaptor Proteins, Signal Transducing , Animals , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Humans , LIM Domain Proteins , Membrane Proteins , Mice , Mutagenesis, Site-Directed , Protein Binding , Protein Serine-Threonine Kinases/metabolism
19.
Int J Biochem Cell Biol ; 34(7): 791-805, 2002 Jul.
Article in English | MEDLINE | ID: mdl-11950595

ABSTRACT

Nck-2 is a ubiquitously expressed adaptor protein comprising primarily three N-terminal SH3 domains and one C-terminal SH2 domain. We report here that Nck-2 interacts with focal adhesion kinase (FAK), a cytoplasmic protein tyrosine kinase critically involved in the cellular control of motility. Using a mutational strategy, we have found that the formation of the Nck-2-FAK complex is mediated by interactions involving multiple SH2 and SH3 domains of Nck-2. The Nck-2 SH2 domain-mediated interaction with FAK is dependent on phosphorylation of Tyr397, a site that is involved in the regulation of cell motility. A fraction of Nck-2 co-localizes with FAK at cell periphery in spreading cells. Furthermore, overexpression of Nck-2 modestly decreased cell motility, whereas overexpression of a mutant form of Nck-2 containing the SH2 domain but lacking the SH3 domains significantly promoted cell motility. These results identify a novel interaction between Nck-2 and FAK and suggest a role of Nck-2 in the modulation of cell motility.


Subject(s)
Carrier Proteins/physiology , Cell Movement/physiology , Protein-Tyrosine Kinases/physiology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Fibronectins/metabolism , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Macromolecular Substances , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases/chemistry , Pseudopodia/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Up-Regulation , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...