Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
Clin Genet ; 94(5): 429-437, 2018 11.
Article in English | MEDLINE | ID: mdl-30051459

ABSTRACT

Spondylometaphyseal dysplasia (SMD) is characterized by developmental changes in long bones and vertebrae. It has large phenotypic diversity and multiple genetic causes, including a recent link to novel variants in the extracellular matrix (ECM) protein fibronectin (FN), a regulator of ECM assembly and key link between the ECM and proper cell function. We identified a patient with a unique SMD, similar to SMD with corner fractures. The patient has been followed over 19 years and presents with short stature, genu varum, kyphoscoliosis, and pectus carinatum. Radiography shows metaphyseal changes that resolved over time, vertebral changes, and capitular avascular necrosis. Whole exome sequencing identified a novel heterozygous FN1 variant (p.Cys97Trp). Using mass spectroscopy, mutant FN was detected in plasma and in culture medium of primary dermal fibroblasts isolated from the patient, but mutant protein was much less abundant than wild-type FN. Immunofluorescence and immunoblotting analyses show that mutant fibroblasts assemble significantly lower amounts of FN matrix than wild-type cells, and mutant FN was preferentially retained within the endoplasmic reticulum. This work highlights the importance of FN in skeletal development, and its potential role in the pathogenesis of a subtype of SMD.


Subject(s)
Fibronectins/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Variation , Osteochondrodysplasias/diagnosis , Osteochondrodysplasias/genetics , Alleles , Child , Child, Preschool , Extracellular Matrix Proteins , Fibroblasts/metabolism , Fibronectins/blood , Fibronectins/metabolism , Humans , Immunohistochemistry , Male , Mutation , Osteochondrodysplasias/metabolism , Osteochondrodysplasias/physiopathology , Radiography , Exome Sequencing
2.
Oncogene ; 33(13): 1649-57, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-23624917

ABSTRACT

In the mammary gland, the stromal extracellular matrix (ECM) undergoes dramatic changes during development and in tumorigenesis. For example, normal adult breast tissue is largely devoid of the ECM protein fibronectin (FN) whereas high FN levels have been detected in the stroma of breast tumors. FN is an established marker for epithelial-mesenchymal transition (EMT), which occurs during development and has been linked to cancer. During EMT, epithelial cell adhesion switches from cell-cell contacts to mainly cell-ECM interactions, raising the possibility that FN may have a role in promoting this transition. Using MCF-10A mammary epithelial cells, we show that exposure to exogenous FN induces an EMT response including upregulation of the EMT markers FN, Snail, N-cadherin, vimentin, the matrix metalloprotease MMP2, α-smooth muscle actin and phospho-Smad2, as well as acquisition of cell migratory behavior. FN-induced EMT depends on Src kinase and extracellular signal-regulated kinase/mitogen-activated protein (ERK/MAP) kinase signaling but not on the immediate early gene EGR-1. FN initiates EMT under serum-free conditions; this response is partially reversed by a transforming growth factor (TGF)ß-neutralizing antibody, suggesting that FN enhances the effect of endogenous TGFß. EMT marker expression is upregulated in cells on a fragment of FN containing the integrin-binding domain but not other domains. Differences in gene expression between FN and Matrigel are maintained with addition of a subthreshold level of TGFß1. Together, these results show that cells interacting with FN are primed to respond to TGFß. The ability of FN to induce EMT shows an active role for the stromal ECM in this process and supports the notion that the increased levels of FN observed in breast tumors facilitate tumorigenesis.


Subject(s)
Breast Neoplasms/metabolism , Breast/metabolism , Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/physiology , Fibronectins/metabolism , Breast/pathology , Breast Neoplasms/genetics , Cell Differentiation/physiology , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Fibronectins/pharmacology , Humans , Phosphorylation , Signal Transduction , Transforming Growth Factor beta/metabolism , Up-Regulation
4.
J Cell Biol ; 154(5): 1081-8, 2001 Sep 03.
Article in English | MEDLINE | ID: mdl-11535624

ABSTRACT

Fibronectin (FN) assembly into a fibrillar extracellular matrix is a stepwise process requiring participation from multiple FN domains. Fibril formation is regulated in part by segments within the first seven type III repeats (III1-7). To define the specific function(s) of this region, recombinant FNs (recFNs) containing an overlapping set of deletions were tested for the ability to assemble into fibrils. Surprisingly, recFN lacking type III repeat III1 (FNDeltaIII1), which contains a cryptic FN binding site and has been suggested to be essential for fibril assembly, formed a matrix identical in all respects to a native FN matrix. Similarly, displacement of the cell binding domain in repeats III9-10 to a position close to the NH2-terminal assembly domain, as well as a large deletion spanning repeats III4-7, had no effect on assembly. In contrast, two deletions that included repeat III2, DeltaIII1-2 and DeltaIII2-5, caused significant reductions in fibril elongation, although binding of FN to the cell surface and initiation of assembly still proceeded. Using individual repeats in binding assays, we show that III2 but not III1 contains an FN binding site. Thus, these results pinpoint repeat III2 as an important module for FN-FN interactions during fibril growth.


Subject(s)
Extracellular Matrix/metabolism , Fibronectins/metabolism , Animals , Binding Sites , CHO Cells , Cricetinae , Extracellular Matrix/chemistry , Fibronectins/genetics , Humans , Immunoblotting , Microscopy, Fluorescence , Protein Binding , Protein Structure, Tertiary , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
5.
J Biol Chem ; 276(39): 36404-10, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11473126

ABSTRACT

Heterodimeric integrin receptors for extracellular matrix (ECM) play vital roles in bidirectional signaling during tissue development, organization, remodeling, and repair. The beta integrin subunit cytoplasmic domain is essential for transmission of many of these signals and overexpression of an unpaired beta tail in cultured cells inhibits endogenous integrins. Unlike vertebrates, which have at least nine beta subunit genes, the nematode Caenorhabditis elegans expresses only one beta subunit (betapat-3), and a null mutation in this gene causes embryonic lethality. To determine the functions of integrins during larval development and in adult tissues, we have taken a dominant negative approach by expression of an HA-betatail transgene composed of a hemagglutinin (HA) epitope tag extracellular domain connected to the betapat-3 transmembrane and cytoplasmic domains. Expression of this transgene in muscle and gonad, major sites of integrin expression, caused a variety of phenotypes dependent on the level of transgene expression. Abnormalities in body wall and sex muscles led to uncoordinated movement and egg-laying defects. Significant anomalies in migration and pathfinding were caused by tissue-specific expression of HA-betatail in the distal tip cells (DTC), the cells that direct gonad morphogenesis. A pat-3 gene with Tyr to Phe mutations in the cytoplasmic domain was able to rescue pat-3 null animals but also showed DTC migration defects. These results show that betapat-3 plays important roles in post-embryonic organogenesis and tissue function.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Gonads/metabolism , Integrin beta Chains , Integrins/physiology , Muscles/metabolism , Actins/chemistry , Alleles , Amino Acid Sequence , Animals , Animals, Genetically Modified , Cell Movement , Cytoplasm/metabolism , DNA, Complementary/metabolism , Epitopes , Genes, Dominant , Hemagglutinins/chemistry , Integrins/biosynthesis , Integrins/genetics , Molecular Sequence Data , Mutation , Ovulation/genetics , Phenotype , Phenylalanine/chemistry , Protein Structure, Tertiary , Transgenes , Tyrosine/chemistry
6.
J Cell Biol ; 150(4): 913-20, 2000 Aug 21.
Article in English | MEDLINE | ID: mdl-10953015

ABSTRACT

Cell binding to extracellular matrix (ECM) components changes cytoskeletal organization by the activation of Rho family GTPases. Tenascin-C, a developmentally regulated matrix protein, modulates cellular responses to other matrix proteins, such as fibronectin (FN). Here, we report that tenascin-C markedly altered cell phenotype on a three-dimensional fibrin matrix containing FN, resulting in suppression of actin stress fibers and induction of actin-rich filopodia. This distinct morphology was associated with complete suppression of the activation of RhoA, a small GTPase that induces actin stress fiber formation. Enforced activation of RhoA circumvented the effects of tenascin. Effects of active Rho were reversed by a Rho inhibitor C3 transferase. Suppression of GTPase activation allows tenascin-C expression to act as a regulatory switch to reverse the effects of adhesive proteins on Rho function. This represents a novel paradigm for the regulation of cytoskeletal organization by ECM.


Subject(s)
Cell Adhesion/physiology , Cytoskeleton/physiology , Tenascin/metabolism , rho GTP-Binding Proteins/metabolism , 3T3 Cells , Actins/physiology , Animals , Cytoskeleton/ultrastructure , Extracellular Matrix Proteins/physiology , Extracellular Matrix Proteins/ultrastructure , Fibrin/physiology , Fibroblasts/physiology , Fibroblasts/ultrastructure , Fibronectins/physiology , GTP Phosphohydrolases/metabolism , Mice , Rats , Recombinant Proteins/metabolism , Substrate Specificity , cdc42 GTP-Binding Protein/metabolism
7.
Oncogene ; 19(28): 3156-63, 2000 Jun 29.
Article in English | MEDLINE | ID: mdl-10918570

ABSTRACT

Fibronectin extracellular matrix plays a critical role in the microenvironment of cells. Loss of this matrix frequently accompanies oncogenic transformation, allowing changes in cell growth, morphology, and tissue organization. The HT1080 human fibrosarcoma cell line is deficient in formation of fibronectin matrix fibrils but assembly can be induced by the glucocorticoid dexamethasone. Here we show that fibronectin assembly can also be restored by stimulation of alpha5beta1 integrin with activating antibody or with Mn2+ suggesting that integrin activity is reduced in these cells. While dexamethasone promoted actin stress fiber formation, actin filaments remained cortical following Mn2+ treatment showing that the dexamethasone effect is not due solely to cytoskeletal changes. HT1080 cells have one activated allele of N-ras and PD98059 inhibition of signaling from Ras through ERK increased fibronectin matrix accumulation. Conversely, the p38 MAP kinase inhibitor SB203580 blocked induction of matrix and increased ERK phosphorylation. Thus, two MAP kinase pathways contribute to the control of integrin-mediated fibronectin assembly. ERK activity and fibronectin assembly were linked in three different ras-transformed cell lines but not in SV40- or RSV-transformed cells indicating that oncogenic Ras uses a distinct mechanism to down-regulate cell-fibronectin interactions.


Subject(s)
Extracellular Matrix/metabolism , Fibronectins/metabolism , ras Proteins/metabolism , 3T3 Cells , Animals , Cell Line, Transformed , Dexamethasone/pharmacology , Humans , Intracellular Fluid/metabolism , MAP Kinase Signaling System , Mice , Rabbits , Receptors, Fibronectin/biosynthesis , Receptors, Vitronectin/biosynthesis , Tumor Cells, Cultured
8.
Mech Dev ; 92(2): 239-50, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10727862

ABSTRACT

Fibronectin (FN) is an adhesive extracellular matrix component that is essential for vertebrate development. It forms a fibrillar matrix at the cell surface which controls cell morphology, migration, proliferation, and other important cellular processes. To address specific functions of FN matrix structure during early vertebrate development, we introduced normal and mutant recombinant FNs (recFNs) into the blastocoel cavity of embryos of the amphibian Pleurodeles waltl. Here we show that a native recFN FN(A-B-) as well as recFNs with specific mutations in the cell-binding domain, FN(RGD-) and FN(syn-), or in a FN-binding region, FNDeltaIII(1), are assembled into fibrillar matrix. A recFN (FNDeltaIII(1-7)) that forms a structurally distinct matrix in cultured cells was assembled into aggregates at the cell periphery and was able to inhibit assembly of endogenous amphibian FN matrix in a dose-dependent manner. Cell adhesion, spreading, and migration were perturbed in vitro and in vivo on chimeric matrices containing FN(RGD-), FN(syn-), or FNDeltaIII(1-7) co-assembled with amphibian FN. Developmentally, this perturbation resulted in defects in mesoderm patterning and inhibition of gastrulation. These results indicate that FN matrix fibrillar structure and composition are important determinants of cell adhesion and migration during development.


Subject(s)
Embryo, Nonmammalian/cytology , Extracellular Matrix/metabolism , Fibronectins/metabolism , Pleurodeles/embryology , Amino Acid Motifs , Animals , Blastocyst , Cell Movement , Extracellular Matrix/chemistry , Extracellular Matrix/ultrastructure , Fibronectins/genetics , Fibronectins/ultrastructure , Gastrula , Mesoderm , Mutation , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/ultrastructure
9.
J Cell Sci ; 113 ( Pt 8): 1491-8, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10725231

ABSTRACT

Fibronectin (FN) matrix assembly is a multi-step process that involves binding to integrin receptors, FN-FN interactions and connections to the actin cytoskeleton. Ultimately, FN is converted into stable matrix fibrils that are detergent-insoluble. RGD-binding integrins such as alpha5beta1 play a major role in the assembly of fibrillar FN. Here we show that alpha4beta1 binding to the alternatively spliced V (IIICS) region of FN initiates an alternative assembly pathway. Activation of alpha4beta1 with exogenous agents such as Mn(2+) or a beta1-stimulatory antibody TS2/16 was sufficient to induce initiation of FN fibrillogenesis by Ramos B lymphoma cells and by CHO(B2)alpha4 cells. Using recombinant FNs lacking specific sequences, we show that assembly is independent of the RGD sequence but requires the V25/CS-1 segment. Previously, we have characterized an activated recombinant FN (FN III(1-7)) that rapidly forms detergent-insoluble multimers upon binding to alpha5beta1 integrin. Alpha4beta1 also formed FNdeltaIII(1-7) multimers without the aid of exogenous stimulants, suggesting that an activated form of FN can override the need for activation of the integrin. In contrast to assembly by alpha5beta1, actin filaments remained largely cortical and no change in cell growth rate was observed with alpha4beta1-mediated assembly. These results show that binding sites on FN other than the RGD sequence/synergy site and distant from the cell binding domain can promote FN assembly. Thus, there appear to be multiple, integrin-specific mechanisms for assembly of FN matrix.


Subject(s)
Extracellular Matrix/metabolism , Fibronectins/metabolism , Integrins/metabolism , Receptors, Lymphocyte Homing/metabolism , Alternative Splicing , Animals , Binding Sites , CHO Cells , Cricetinae , Fibronectins/genetics , Humans , Integrin alpha4beta1 , Oligopeptides/metabolism , Protein Binding , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
10.
Curr Opin Cell Biol ; 11(5): 622-7, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10508649

ABSTRACT

Fibronectin matrix assembly is a regulated stepwise process. In the past year, analyses of fibronectin domains, integrin and cytoskeletal contributions, and fibril architecture have provided new insights into assembly mechanisms and matrix control of cell functions. Like fibronectin, laminin polymerization is cell-mediated. Thus a common pathway for extracellular matrix assembly is emerging.


Subject(s)
Extracellular Matrix/metabolism , Fibronectins/metabolism , Microfibrils/metabolism , Animals , Basement Membrane/metabolism , Basement Membrane/ultrastructure , CHO Cells , Cell Differentiation , Cell Movement , Cells, Cultured , Cricetinae , Cricetulus , Extracellular Matrix/ultrastructure , Fibronectins/chemistry , Integrins/metabolism , Laminin/metabolism , Motion , Protein Binding , Protein Conformation , Protein Structure, Tertiary
11.
J Biol Chem ; 274(30): 20943-8, 1999 Jul 23.
Article in English | MEDLINE | ID: mdl-10409640

ABSTRACT

Retraction of the blood clot by nucleated cells contributes both to hemostasis and to tissue remodeling. Although plasma fibronectin (FN) is a key component of the clot, its role in clot retraction is unclear. In this report, we demonstrate that the incorporation of FN into fibrin matrices significantly improves clot retraction by nucleated cells expressing the integrin alpha(5)beta(1). Further, we show that FN-fibrin clots support increased cell spreading when compared with fibrin matrices. To determine the structural requirements for FN in this process, recombinant FN monomers deficient in ligand binding or fibrin cross-linking were incorporated into fibrin clots. We show that recombinant FN monomers support clot retraction by Chinese hamster ovary cells expressing the integrin alpha(5)beta(1). This process depends on both the Arg-Gly-Asp (RGD) and the synergy cell-binding sites and on covalent FN-fibrin binding, demonstrating that cross-linking within the clot is important for cell-FN interactions. These data show that alpha(5)beta(1) can bind to FN within a clot to promote clot retraction and support cell shape change. This provides strong evidence that alpha(5)beta(1)-FN interactions may contribute to the cellular events required for wound contraction.


Subject(s)
Blood Coagulation , Fibrin/metabolism , Receptors, Fibronectin/metabolism , Animals , Binding Sites , CHO Cells , Cricetinae , Cross-Linking Reagents , Fibrin/chemistry , Ligands , Protein Binding , Receptors, Fibronectin/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Transfection
12.
Biochem J ; 341 ( Pt 3): 555-61, 1999 Aug 01.
Article in English | MEDLINE | ID: mdl-10417317

ABSTRACT

Fibronectin is an extracellular-matrix glycoprotein encoded by a single gene, but with significant protein heterogeneity introduced through alternative RNA splicing and post-translational modifications. The (V+C)(-) splice variant, in which nucleotides encoding protein segments III-15 and I-10 are deleted along with the entire variable region, is unique in that expression is restricted to cartilaginous tissues. All known fibronectin splice variants retain the two C-terminal cysteine residues essential for dimerization, but cellular and/or structural constraints appear to influence homo- and heterodimerization patterns. Dimerization patterns of the (V+C)(-) isoform were studied under native conditions within canine articular cartilage and experimentally in COS-7, NIH-3T3 and CHO-K1 cell cultures. In all systems, (V+C)(-) fibronectin secretion was predominantly in a homodimeric configuration. Lower levels of (V+C)(-) monomers were also present. Heterodimers of (V+C)(-) with V(+),C(+) (V120) isoforms were not detected. Heterodimers of (V+C)(-) with V(-),C(+) (V0) subunits were detected only at low levels. Functional properties may differ significantly among monomers, homodimers and heterodimers. The unique dimerization pattern of (V+C)(-) fibronectin is consistent with this isoform having specialized functional properties in situ that are important for either the structural organization and biomechanical properties of cartilage matrix or regulation of a chondrocytic phenotype.


Subject(s)
Cartilage, Articular/metabolism , Fibronectins/metabolism , Protein Isoforms/metabolism , Animals , Cell Line , Dimerization , Dogs , Fibronectins/chemistry , Protein Isoforms/chemistry , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
13.
J Surg Res ; 83(1): 27-31, 1999 May 01.
Article in English | MEDLINE | ID: mdl-10210638

ABSTRACT

BACKGROUND: Integrins are heterodimeric transmembrane glycoproteins that mediate cell interactions with the extracellular matrix. In vivo, integrin affinity can be modulated by intracellular signaling events. This can be simulated by a point mutation (D723R) in the cytoplasmic tail of the beta3 integrin subunit which results in constitutive activation. The effects of beta3 integrin activation on the function of alphavbeta3, an integrin which is important to the adhesive events of multiple cell types, were addressed using Chinese hamster ovary cells expressing either the wild-type alphavbeta3 integrin or the mutant alphavbeta3(D723R). The interactions of these cell lines with fibrin matrices were compared. METHODS: Receptor expression levels were confirmed by FACS analyses using a monoclonal anti-alphavbeta3 antibody. Cell attachment to fibrin-coated dishes was determined after 1 h by fixation and crystal violet staining followed by elution of the dye and OD measurement. Fibrin clot retraction was measured by culturing cells in fibrin clots for 24 h. The clots were detached from the dish and the surface area was calculated at individual time points. RESULTS: CHO alphavbeta3(D723R) cells displayed a greater than twofold increase in attachment to fibrinogen or to fibrin matrices when compared to wild-type transfectants. Further, CHO alphavbeta3(D723R) cell retraction of fibrin matrices was significantly greater at nearly all time points. CONCLUSION: Activation of the beta3 integrin subunit significantly improves the interaction of alphavbeta3 with fibrin and may play a role in the integrin-mediated signaling events which occur following vascular injury.


Subject(s)
Antigens, CD/physiology , Fibrin/metabolism , Platelet Membrane Glycoproteins/physiology , Receptors, Vitronectin/physiology , Animals , CHO Cells , Cell Adhesion , Clot Retraction , Cricetinae , Fibrinogen/metabolism , Humans , Integrin beta3 , Mutagenesis , Receptors, Vitronectin/genetics
14.
Biochem J ; 338 ( Pt 2): 375-86, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10024513

ABSTRACT

Fibronectin (Fn) binds to fibrin in clots by covalent and non-covalent interactions. The N- and C-termini of Fn each contain one non-covalent fibrin-binding site, which are composed of type 1 (F1) structural repeats. We have previously localized the N-terminal site to the fourth and fifth F1 repeats (4F1.5F1). In the current studies, using proteolytic and recombinant proteins representing both the N- and C-terminal fibrin-binding regions, we localized and characterized the C-terminal fibrin-binding site, compared the relative fibrin-binding activities of both sites and determined the contribution of each site to the fibrin-binding activity of intact Fn. By fibrin-affinity chromatography, a protein composed of the 10F1 repeat through to the C-terminus of Fn (10F1-COOH), expressed in COS-1 cells, and 10F1-12F1, produced in Saccharomyces cerevisiae, displayed fibrin-binding activity. However, since 10F1 and 10F1.11F1 were not active, the presence of 12F1 is required for fibrin binding. A proteolytic fragment of 14.4 kDa, beginning 14 residues N-terminal to 10F1, was isolated from the fibrin-affinity matrix. Radio-iodinated 14.4 kDa fibrin-binding peptide/protein (FBP) demonstrated a dose-dependent and saturable binding to fibrin-coated wells that was both competitively inhibited and reversed by unlabelled 14.4 kDa FBP. Comparison of the fibrin-binding affinities of proteolytic FBPs from the N-terminus (25.9 kDa FBP), the C-terminus (14.4 kDa) and intact Fn by ELISA yielded estimated Kd values of 216, 18 and 2.1 nM, respectively. The higher fibrin-binding affinity of the N-terminus was substantiated by the ability of both a recombinant 4F1.5F1 and a monoclonal antibody (mAb) to this site to maximally inhibit biotinylated Fn binding to fibrin by 80%, and by blocking the 90% inhibitory activity of a polyclonal anti-Fn, by absorption with the 25.9 kDa FBP. We propose that whereas the N-terminal site appears to contribute to most of the binding activity of native Fn to fibrin, the specific binding of the C-terminal site may strengthen this interaction.


Subject(s)
Fibrin/metabolism , Fibronectins/metabolism , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Chromatography, Affinity , DNA Primers , Fibronectins/chemistry , Humans , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptide Fragments/metabolism , Protein Binding , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
15.
Curr Biol ; 8(23): 1285-8, 1998 Nov 19.
Article in English | MEDLINE | ID: mdl-9822581

ABSTRACT

The basement membrane is a specialized extracellular matrix located at epithelial-mesenchymal boundaries that supports cell adhesion, migration, and proliferation; it is highly conserved between invertebrates and vertebrates [1,2]. One of its component proteins, SPARC (osteonectin/BM-40), binds calcium and collagens, and can modulate cell-matrix interactions, so altering cell shape, growth, and differentiation [3,5]. The tissue distribution of a secreted fusion protein containing SPARC and green fluorescent protein (GFP) was analyzed in Caenorhabditis elegans. The protein localized to most basement membranes along body wall and sex muscles, and was also deposited around the pharynx and the gonad, in the spermatheca and at the distal tip cells. The contributions of SPARC to C. elegans development were determined using RNA interference, which accurately phenocopies loss-of-function defects [6-8]. A reduction in the amount of SPARC protein resulted in embryonic or larval lethality in a significant proportion of progeny. Those that survived developed a 'clear' phenotype characterized by a lack of gut granules, which made the animals appear transparent, plus small size, and sterility or reduced fecundity. No significant morphological abnormalities were observed, indicating that SPARC plays a regulatory rather than structural role in modulating cell-matrix interactions during normal development and reproduction.


Subject(s)
Caenorhabditis elegans/physiology , Osteonectin/physiology , Animals , Animals, Genetically Modified , Basement Membrane/physiology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Extracellular Matrix/physiology , Green Fluorescent Proteins , Luminescent Proteins , Mutation , Osteonectin/genetics , Recombinant Fusion Proteins
16.
J Biol Chem ; 273(40): 25533-6, 1998 Oct 02.
Article in English | MEDLINE | ID: mdl-9748212

ABSTRACT

Developmental patterning and differentiation, maintenance of parenchymal cell function, and the size, shape, and invasiveness of tumors are all orchestrated by cell interactions with the extracellular matrix. Here we show that the fibrillar structure of fibronectin (FN) matrix encodes essential regulatory cues and controls cell proliferation and signaling through changes in matrix architecture. A matrix assembled from native FN stimulated cell growth. In contrast, a mutant FN (FNDeltaIII1-7) that contains all known cell binding motifs but forms a structurally distinct matrix inhibited progression from G0/G1 into S phase. Furthermore, FNDeltaIII1-7 suppressed the stimulatory capacity of native FN and induced different levels of tyrosine phosphorylation of pp125(FAK). The differential effects on cell growth were ablated by blocking formation of matrix fibrils. Thus, modification of matrix architecture provides a novel approach to control cell proliferation.


Subject(s)
Cell Cycle/physiology , Fibronectins/chemistry , Proteins , Bromodeoxyuridine/metabolism , Cell Adhesion/physiology , Cell Adhesion Molecules/metabolism , Cell Division/physiology , Cell Line , Extracellular Matrix/physiology , Fibronectins/genetics , Fluorescent Antibody Technique , Focal Adhesion Protein-Tyrosine Kinases , Histocytochemistry , Interphase/physiology , Mutation/genetics , Phosphoproteins/metabolism , Phosphorylation , Phosphotyrosine/analysis , Protein-Tyrosine Kinases/metabolism , Retinoblastoma-Like Protein p130 , S Phase/physiology , Signal Transduction/physiology
17.
Cancer Res ; 58(16): 3586-9, 1998 Aug 15.
Article in English | MEDLINE | ID: mdl-9721865

ABSTRACT

The synthetic glucocorticoid dexamethasone markedly decreases the invasiveness of HT-1080 human fibrosarcoma cells. We show here that dexamethasone treatment of HT-1080 cell aggregates more than doubles their cohesivity from 3.9 to 9.7 dyne/cm. Western blot analysis shows a corresponding increase in cadherin expression. This was accompanied by an increase in the rate of calcium-dependent aggregation. Dexamethasone-treated aggregates spread to form a monolayer in Matrigel spreading assays, but the cells remained much more contiguous than their untreated counterparts. Invasion-suppression by dexamethasone may therefore be due, at least in part, to a previously unsuspected increase in cadherin-mediated cohesion.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Cadherins/drug effects , Cell Adhesion/drug effects , Dexamethasone/pharmacology , Neoplasm Proteins/drug effects , Cadherins/metabolism , Calcium/pharmacology , Collagen , Drug Combinations , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Humans , Laminin , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Proteoglycans , Tumor Cells, Cultured/drug effects , Up-Regulation
18.
J Biol Chem ; 273(4): 2073-7, 1998 Jan 23.
Article in English | MEDLINE | ID: mdl-9442046

ABSTRACT

Tenascin-C is a large, multimeric extracellular matrix protein that is found in a variety of tissues and can have profound effects on cell adhesion. It is secreted from cells as a hexamer of six identical chains called a hexabrachion. Disulfide bonding among tenascin subunits mediates intracellular assembly into hexamers. The amino-terminal assembly domain consists of heptad repeats and at least six cysteine residues (Cys-64, -111, -113, -140, -146, -147) that could be involved in multimerization. We have now determined the requirements for these cysteine residues during hexamer assembly. Our results show that only Cys-64 is required to form the hexameric structure. Mutation of Cys-64 to glycine resulted in release of trimer intermediates, which probably form via the heptad repeats, but no hexamers were secreted. In contrast, individual or pairs of mutations of each of the other cysteines had no effect on tenascin hexamer formation, and inclusion of any other cysteine mutations along with C64G did not further disrupt the multimer pattern. However, when all six cysteines were mutated, monomers were the major extracellular form. Together, these results show that trimers are an intermediate of tenascin-C assembly and that Cys-64 is essential for formation of hexabrachions.


Subject(s)
Cysteine/metabolism , Tenascin/metabolism , Amino Acid Substitution , Animals , COS Cells , Cells, Cultured , Cysteine/genetics , Disulfides/analysis , Mice , Mutagenesis, Site-Directed , Protein Conformation , Structure-Activity Relationship , Tenascin/genetics , Transfection
19.
Ann N Y Acad Sci ; 857: 143-54, 1998 Oct 23.
Article in English | MEDLINE | ID: mdl-9917839

ABSTRACT

Changes in extracellular matrix (ECM) structure and composition, such as occur during morphogenesis, can have important regulatory effects on cell behavior. Two fibronectin (FN)-based systems have been developed to dissect how cells respond to different types of ECM. One system mimics the provisional matrix of the wound and is composed of FN cross-linked into a fibrin clot matrix. Unlike cells on FN alone, cells on an FN-fibrin matrix are smaller with cortical distribution of actin filaments and membrane ruffles. Addition of the ECM protein tenascin to the FN-fibrin matrix induces a different cell morphology. Thus, matrix composition can have profound effects on cell phenotype. Cells also interact with FN while assembling it into a fibrillar matrix. Using recombinant FNs, a domain that is required for normal progression of FN fibril formation has been identified. During assembly of this recombinant matrix, formation of actin stress fibers and focal adhesions is delayed, demonstrating that changes in FN matrix structure can affect intracellular organization and activation of signaling pathways.


Subject(s)
Cell Physiological Phenomena , Extracellular Matrix/physiology , Fibronectins/physiology , Actins/physiology , Animals , Fibrin/physiology , Morphogenesis , Signal Transduction , Tenascin/physiology
20.
Trends Cardiovasc Med ; 8(8): 357-62, 1998 Nov.
Article in English | MEDLINE | ID: mdl-14987550

ABSTRACT

At sites of tissue injury or inflammation, extravasation of plasma proteins leads to the formation of a complex fibrillar matrix composed primarily of fibrin and plasma fibronectin (pFN). This protein meshwork serves not only to reestablish the integrity of the vascular system but also to provide a scaffold for cell migration and subsequent wound repair. The interactions between cell surface receptors and this provisional extracellular matrix (ECM) provide important cues that can modulate the cellular response at the injury site, leading to alterations in cell growth and gene expression. Key determinants of this response may lie in the structure and composition of this "injury-associated" ECM.

SELECTION OF CITATIONS
SEARCH DETAIL
...