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










Publication year range
1.
Oncogene ; 32(16): 2016-26, 2013 Apr 18.
Article in English | MEDLINE | ID: mdl-22710722

ABSTRACT

Metastatic cell migration and invasion are regulated by altered adhesion-mediated signaling to the actin-based cytoskeleton via activated Src-FAK complexes. Src-suppressed C-kinase substrate (SSeCKS, the rodent orthologue of human Gravin/AKAP12), whose expression is downregulated by oncogenic Src and in many human cancers, antagonizes oncogenic Src pathways including those driving neovascularization at metastatic sites, metastatic cell motility and invasiveness. This is likely manifested through its function as a scaffolder of F-actin and signaling proteins such as cyclins, calmodulin, protein kinase C and A. Here we show that in contrast to its ability to inhibit haptotaxis, SSeCKS increased prostate cancer cell adhesion to fibronectin and type I collagen in a FAK-dependent manner, correlating with a relative increase in FAK(poY397) levels. In contrast, SSeCKS suppressed adhesion-induced Src activation (Src(poY416)) and phosphorylation of FAK at Y925, a known Src substrate site. SSeCKS also induced increased cell spreading, cell flattening, integrin ß1 clustering and formation of mature focal adhesion plaques. An in silico analysis identified a Src-binding domain on SSeCKS(aa 153-166) that is homologous to the Src-binding domain of caveolin-1, and this region is required for SSeCKS-Src interaction, for SSeCKS-enhanced Src activity and sequestration to lipid rafts and for SSeCKS-enhanced adhesion of MAT-LyLu and CWR22Rv1 prostate cancer cells. Our data suggest a model in which SSeCKS suppresses oncogenic motility by sequestering Src to caveolin-rich lipid rafts, thereby disengaging Src from FAK-associated adhesion and signaling complexes.


Subject(s)
A Kinase Anchor Proteins/metabolism , Actin Cytoskeleton/metabolism , Caveolin 1/metabolism , Cell Cycle Proteins/metabolism , Focal Adhesion Kinase 1/metabolism , Membrane Microdomains/enzymology , A Kinase Anchor Proteins/genetics , Amino Acid Sequence , Cell Adhesion/physiology , Cell Cycle Proteins/genetics , Cell Movement/physiology , Chemotaxis/physiology , Cytoskeleton/metabolism , Humans , Integrin beta1/metabolism , Molecular Sequence Data , Signal Transduction , src-Family Kinases/metabolism
2.
Oncogene ; 32(10): 1266-73, 2013 Mar 07.
Article in English | MEDLINE | ID: mdl-22525271

ABSTRACT

The Hippo signaling pathway regulates cellular proliferation and survival, thus exerting profound effects on normal cell fate and tumorigenesis. The pivotal effector of this pathway is YAP, a transcriptional co-activator amplified in mouse and human cancers where it promotes epithelial-to-mesenchymal transition and malignant transformation. Here, we report a novel regulatory mechanism for the YAP oncogenic function via direct interaction with non-receptor tyrosine phosphatase 14 (PTPN14) through the WW domain of YAP and the PPxY domain of PTPN14. We also found that YAP is a direct substrate of PTPN14. In addition, luciferase reporter assay showed that the inhibition of the YAP transcriptional co-activator function by PTPN14 is mediated through their protein interactions and may result from an increase in the inactive cytoplasmic form of YAP. Last, knockdown of PTPN14 induces the nuclear retention of YAP and increases the YAP-dependent cell migration. In summary, our results indicate a potential regulatory role of PTPN14 on YAP and demonstrate a novel mechanism in YAP regulation.


Subject(s)
Nuclear Proteins/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Transcription Factors/genetics , Amino Acid Sequence , Cell Cycle Proteins , Cell Growth Processes/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , HEK293 Cells , Humans , Molecular Sequence Data , Nuclear Proteins/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Signal Transduction , Subcellular Fractions/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Transfection
4.
Virology ; 292(1): 1-5, 2002 Jan 05.
Article in English | MEDLINE | ID: mdl-11878902

ABSTRACT

Closed circular (cc) forms of extrachromosomal HIV DNA are detected in patients with high viral loads; however, it is unclear whether these forms remain if virus replication is suppressed to undetectable levels by combination antiretroviral therapy. A nested primer polymerase chain reaction amplification assay was used to detect the presence of ccDNA containing two long terminal repeat sequences (2-LTR) in PBMC of patients with low or undetectable plasma HIV RNA. Fifty percent of patients with plasma RNA levels <50 copies/ml of blood had detectable 2-LTR DNA. Sequencing of the products identified normal LTR--LTR junctions in the minority of cases with the majority containing anomalies including deletions and insertions. The persistence of HIV ccDNA in patients with no detectable plasma RNA could be consistent with ongoing de novo infection of dividing cells or with stability of this form of DNA in nondividing cells.


Subject(s)
DNA, Circular/genetics , DNA, Viral/genetics , HIV Infections/virology , HIV Long Terminal Repeat/genetics , Leukocytes, Mononuclear/virology , RNA, Viral/blood , Base Sequence , HIV-1/genetics , HIV-1/physiology , Humans , Molecular Sequence Data , Sequence Analysis, DNA
5.
J Cell Sci ; 114(Pt 12): 2291-300, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11493668

ABSTRACT

The beta1,4-galactosyltransferase family contains at least seven unique gene products, of which beta1,4-galactosyltransferase I (GalT) is the most exhaustively studied. GalT exists in the Golgi complex, similar to many other glycosyltransferases, as well as on the cell surface, where it functions as a signaling receptor for extracellular glycoside ligands. When expressed on the surface, GalT associates with the cytoskeleton and, upon ligand-induced aggregation, induces cell-type specific intracellular signal cascades. In an effort to define the mechanisms by which surface GalT exerts these intracellular effects, we used the yeast two-hybrid system to identify proteins that specifically interact with the GalT cytoplasmic domain. The yeast two-hybrid screen identified two distinct clones (1.12 and 2.52) that showed identity to portions of SSeCKS (Src Suppressed C Kinase Substrate). SSeCKS is a previously defined kinase and cytoskeleton scaffolding protein whose subcellular distribution and functions are remarkably similar to those attributed to GalT. Both SSeCKS and GalT have been localized to the perinuclear/Golgi region as well as to filopodia/lamellipodia. SSeCKS and GalT have been implicated in regulating cell growth, actin filament dynamics, and cell spreading. Interestingly, 1.12 and 2.52-GFP constructs were localized to subcellular domains that correlated with the two purported subcellular distributions for GalT; 2.52 being confined to the Golgi, whereas 1.12 localized primarily to filopodia. Coimmunoprecipitation assays demonstrate stable binding between the GalT cytoplasmic domain and the 1.12 and 2.52 domains of SSeCKS in appropriately transfected cells. Similar assays demonstrate binding between the endogenous GalT and SSeCKS proteins also. Coimmunoprecipitation assays were performed in both directions and produced similar results (i.e. using either anti-GalT domain or anti-SSeCKS domain antibodies as the precipitating reagent). A functional interaction between the GalT cytoplasmic domain and SSeCKS was illustrated by the ability of either the 1.12 or 2.52 SSeCKS domain to restore a normal adhesive phenotype in cells overexpressing the TL-GFP dominant negative construct. TL-GFP is composed of the GalT cytoplasmic and transmembrane domains fused to GFP, and leads to a loss of cell adhesion on laminin by displacing the endogenous GalT from its cytoskeleton binding sites. This is the first reported interaction between a glycosyltransferase and a scaffolding protein, and suggests that SSeCKS serve to integrate the various functions ascribed to the GalT cytoplasmic domain.


Subject(s)
Cell Cycle Proteins , Cytoskeleton/metabolism , Mitogens/metabolism , N-Acetyllactosamine Synthase/chemistry , N-Acetyllactosamine Synthase/metabolism , A Kinase Anchor Proteins , Amino Acid Sequence , Animals , Blotting, Western , Cell Adhesion , Cell Extracts , Cell Line , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Microscopy, Fluorescence , Mitogens/chemistry , Mitogens/genetics , Molecular Sequence Data , N-Acetyllactosamine Synthase/genetics , Phenotype , Precipitin Tests , Protein Binding , Sequence Homology, Amino Acid , Signal Transduction , Solubility , Two-Hybrid System Techniques , Yeasts
6.
Cancer Res ; 61(14): 5644-51, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454719

ABSTRACT

The molecular mechanisms leading to prostate cancer remain poorly understood, especially concerning the progression to the metastatic form. SSeCKS, a major protein kinase C substrate with tumor suppressor activity, is likely the rodent orthologue of human Gravin/AKAP12, a scaffolding protein for protein kinases A and C. Gravin was mapped as a single-copy gene to 6q24-25.2, a hotspot for deletion in advanced prostate cancer, and therefore, we investigated the role of SSeCKS/Gravin in prostate oncogenesis. SSeCKS/Gravin protein was detected in untransformed rat and human prostate epithelial cell lines EP12 and PZ-HPV-7, respectively, and in human prostatic epithelium, especially basal epithelial cells. In contrast, SSeCKS/Gravin protein and RNA levels were severely reduced in human (PC-3, PPC-1, LNCaP, DU145, and TSU) and rat Dunning (AT3.1 and MatLyLu) prostate cancer cell lines. The regulated reexpression of SSeCKS in MatLyLu cells induced filopodia-like projections and a decrease in anchorage-independent growth. In nude mice, SSeCKS reexpression slightly decreased primary-site tumor growth but severely decreased the formation of lung metastases. Primary-site tumors that progressed lost regulated SSeCKS reexpression. SSeCKS/Gravin expression was detected in benign human prostatic lesions and well-differentiated carcinomas but not in undifferentiated lesions with Gleason sums > or =6. Our data suggest a role for the loss of SSeCKS/Gravin in the metastatic progression of human prostate cancer.


Subject(s)
Cell Cycle Proteins , Mitogens/genetics , Prostatic Neoplasms/genetics , A Kinase Anchor Proteins , Animals , Biomarkers, Tumor , Cell Differentiation , Cell Division/drug effects , Chromosome Banding , Chromosome Mapping , Chromosomes, Human, Pair 6/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Nude , Mitogens/physiology , Molecular Weight , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Transplantation , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Prostatic Neoplasms/pathology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Proteins/genetics , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Rats , Tetracycline/pharmacology , Transplantation, Heterologous , Tumor Cells, Cultured
7.
Oncogene ; 20(2): 141-6, 2001 Jan 11.
Article in English | MEDLINE | ID: mdl-11313943

ABSTRACT

Line 10T1/2 mouse fibroblast overexpressing the v-Jun oncoprotein were morphologically altered, grew into multilayered foci in culture and formed colonies when suspended in agar. The growth rate of the v-Jun-transformed 10T1/2 cells was not changed significantly from that of the untransformed parental cells, but the saturation density of the transformed cultures exceeded that of normal controls by a factor of 2. mRNA extracted from v-Jun-transformed 10T1/2 cells was analysed for differential gene expression with DNA micro-array technology. One of the targets downregulated by v-Jun was identified as SSeCKS (Src-suppressed C kinase substrate). Re-expression of SSeCKS in v-Jun-transformed fibroblasts reversed the transformed phenotype of the cells. Their ability to form foci was reduced to background levels, the number and size of agar colonies was lowered by a factor of 10 and the saturation density was significantly diminished. However, expression of SSeCKS had little effect on the morphology of v-Jun-transformed 10T1/2 cells. These data suggest that the SSeCKS protein has growth-attenuating properties. Down-regulation of SSeCKS may be essential for Jun-induced transformation.


Subject(s)
Cell Cycle Proteins , Fibroblasts/pathology , Mitogens/metabolism , Oncogene Protein p65(gag-jun)/metabolism , A Kinase Anchor Proteins , Animals , Cell Division/genetics , Cell Transformation, Neoplastic , Cells, Cultured , Down-Regulation , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic , Mice , Mitogens/genetics , Oligonucleotide Array Sequence Analysis , Oncogene Protein p65(gag-jun)/genetics , Retroviridae/genetics , Tumor Stem Cell Assay
9.
Mol Cell Biol ; 20(19): 7259-72, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10982843

ABSTRACT

SSeCKS, first isolated as a G(1)-->S inhibitor that is downregulated in src- and ras-transformed cells, is a major cytoskeleton-associated PKC substrate with tumor suppressor and kinase-scaffolding activities. Previous attempts at constitutive expression resulted in cell variants with truncated ectopic SSeCKS products. Here, we show that tetracycline-regulated SSeCKS expression in NIH 3T3 cells induces G(1) arrest marked by extracellular signal-regulated kinase 2-dependent decreases in cyclin D1 expression and pRb phosphorylation. Unexpectedly, the forced reexpression of cyclin D1 failed to rescue SSeCKS-induced G(1) arrest. Confocal microscopy analysis revealed cytoplasmic colocalization of cyclin D1 with SSeCKS. Because the SSeCKS gene encodes two potential cyclin-binding motifs (CY) flanking major in vivo protein kinase C (PKC) phosphorylation sites (Ser(507/515)), we addressed whether SSeCKS encodes a phosphorylation-dependent cyclin scaffolding function. Bacterially expressed SSeCKS-CY bound cyclins D1 and E, whereas K-->S mutations within either CY motif ablated binding. Activation of PKC in vivo caused a rapid translocation of cyclin D1 to the nucleus. Cell permeable, penetratin-linked peptides encoding wild-type SSeCKS-CY, but not K-->S or phospho-Ser(507/515) variants, released cyclin D1 from its cytoplasmic sequestration and induced higher saturation density in cyclin D1-overexpressor cells or rat embryo fibroblasts. Our data suggest that SSeCKS controls G(1)-->S progression by regulating the expression and localization of cyclin D1. These data suggest that downregulation of SSeCKS in tumor cells removes gating checkpoints for saturation density, an effect that may promote contact independence.


Subject(s)
CDC2-CDC28 Kinases , Cell Compartmentation/physiology , Cell Cycle Proteins , Contact Inhibition/physiology , Cyclins/metabolism , G1 Phase/physiology , Gene Expression Regulation/physiology , Genes, Tumor Suppressor , Mitogens/physiology , Protein Kinase C/metabolism , S Phase/physiology , 3T3 Cells/cytology , 3T3 Cells/metabolism , A Kinase Anchor Proteins , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Animals , Cell Division , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/genetics , Cyclin D , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinases/metabolism , Cytoplasm/metabolism , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/metabolism , MAP Kinase Signaling System , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogens/genetics , Models, Biological , Molecular Sequence Data , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Rats , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
10.
Histochem J ; 32(1): 13-26, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10805381

ABSTRACT

SSeCKS is a major protein kinase C substrate which has tumour suppressor activity in models of src- and ras-induced oncogenic transformation. The mitogenic regulatory activity of SSeCKS is likely manifested by its ability to bind key signalling proteins such as protein kinases C and A and calmodulin, and to control actin-based cytoskeletal architecture. Rat SSeCKS shares extensive homology with human Gravin, an autoantigen in myasthenia gravis that encodes kinase scaffolding functions and whose expression pattern in fibroblasts and nerves suggests a role in cell motility. Here, we analyse the expression of SSeCKS and Gravin in rodent and human fibroblast and epithelial cell lines using antibodies specific or crossreactive for SSeCKS or Gravin. SSeCKS expression was then analysed in developing mouse embryos and in adult tissues. In the foetal mouse, early SSeCKS protein expression (E10-11) is focused in the loose mesenchyme, luminal surface of the neural tube, notochord, early heart and pericardium, urogenital ridge, and dorsal and ventral sections of limb buds. In later stages (E12-14), SSeCKS is widely expressed in mesenchymal cells but is absent in the spinal ganglia. By E15, SSeCKS expression is ubiquitous, although the staining pattern varies from being striated within smooth muscle sarcomeres to filamentous in mesenchymal and select epithelial cells. In the adult mouse, SSeCKS staining is relatively ubiquitous, with highest expression in the gonads, smooth and cardiac muscle, lung, brain and heart. High expression is also detected in fibroblasts and nerve fibres as well as in more specialized cells such as glomerular mesangial cells and testicular Sertoli cells. SSeCKS expression in the rat testes correlates with the induction of puberty, and in mature mouse spermatozoa, SSeCKS is found in peripheral acrosome membranes and in a helix-like winding pattern within the midsection. Periodic enrichments of SSeCKS are found in sperm midsections and in developing axons, suggesting a role in architectural infrastructure. As with Gravin, high SSeCKS expression is absent in most epithelial cells; however, in contrast to Gravin, SSeCKS is expressed in Purkinje cells, cardiac muscle, macrophages and hepatic stellate cells, indicating overlapping yet distinct patterns of tissue expression in the SSeCKS/Gravin family. The data suggest roles for SSeCKS in the control of cytoskeletal and tissue architecture, formation of migratory processes and cell migration during embryogenesis.


Subject(s)
Cell Cycle Proteins , Cell Movement/physiology , Cytoskeleton/metabolism , Embryonic and Fetal Development/physiology , Genes, Tumor Suppressor , Mitogens/physiology , Protein Kinase C/metabolism , A Kinase Anchor Proteins , Animals , Blotting, Northern , Cells, Cultured , DNA Primers/chemistry , Epithelial Cells/metabolism , Female , Fibroblasts/metabolism , Gene Expression , Humans , Immunoenzyme Techniques , Male , Mice , Mice, Inbred BALB C , Mitogens/genetics , Proteins/genetics , Proteins/metabolism , RNA, Messenger/biosynthesis , Rats , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity
11.
J Cell Sci ; 112 ( Pt 3): 361-70, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9885289

ABSTRACT

Activation of protein kinase C is a key signal transduction event in mesangial cell dedifferentiation and proliferation, yet little is known about downstream substrates or their roles in normal or diseased glomeruli. SSeCKS, a novel protein kinase C substrate originally isolated as a src-suppressed negative mitogenic regulator in fibroblasts, controls actin-based cytoskeletal architecture and scaffolds key signaling kinases such as protein kinase C and protein kinase A. Based on the morphologic similarity between SSeCKS-overexpressing fibroblasts and stellate mesangial cells, we hypothesized that SSeCKS might play a role in mesangial cell morphology in a protein kinase C-dependent manner. Immunoblotting, in situ staining and northern blotting detected abundant expression of SSeCKS in human and rodent mesangial cells and glomerular parietal cells but not in renal tubular epithelia. Immunofluorescence analysis showed enrichment of SSeCKS in mesangial cell podosomes and along a cytoskeletal network distinct from F-actin. Activation of protein kinase C by phorbol ester resulted in a rapid serine phosphorylation of SSeCKS and its subsequent translocation to perinuclear sites, coincident with the retraction of stellate processes. These effects were blocked by concentrations of bis-indolylmaleimide that selectively inhibit protein kinase C. Finally, ablation of SSeCKS expression using retroviral anti-sense vectors induced (1) an elongated, fibroblastic cell morphology, (2) production of thick, longitudinal stress fibers and (3) repositioning of vinculin-associated focal complexes away from the cell edges. These data suggest a role for SSeCKS as a downstream mediator of protein kinase C-controlled, actin-based mesangial cell cytoskeletal architecture.


Subject(s)
Actins/metabolism , Cell Cycle Proteins , Glomerular Mesangium/embryology , Glomerular Mesangium/metabolism , Mitogens/analysis , Mitogens/metabolism , Protein Kinase C/metabolism , A Kinase Anchor Proteins , Actins/ultrastructure , Animals , Cells, Cultured , Cytoskeleton/metabolism , Epithelial Cells/metabolism , Fluorescent Antibody Technique , Humans , Immunohistochemistry , Kidney/metabolism , Kidney/ultrastructure , Mice , Phorbol Esters/metabolism , Phosphorylation , Rats , Tissue Distribution
13.
Cell Motil Cytoskeleton ; 41(1): 1-17, 1998.
Article in English | MEDLINE | ID: mdl-9744295

ABSTRACT

Activation of protein kinase C (PKC) in many cell types results in cytoskeletal reorganization associated with cell proliferation. We previously described a new cell cycle-regulated myristylated PKC substrate, SSeCKS (pronounced essex), that interacts with the actin cytoskeleton [Lin et al., 1995, 1996]. SSeCKS shares significant homology with Gravin, which encodes kinase scaffolding functions for PKC and PKA [Nauert et al., 1997]. This article describes the cellular effects of ectopically expressing SSeCKS in untransformed NIH3T3 fibroblasts. Because the constitutive overexpression of SSeCKS is toxic [Lin et al., 1995], we developed cell lines with tetracycline (tet)-regulated SSeCKS expression. The induction of SSeCKS (removal of tet) caused significant cell flattening and the elaboration of an SSeCKS-associated cortical cytoskeletal matrix resistant to Triton X-100 extraction. Flattened cells were growth-arrested and marked by the formation of cellular projections and the temporary loss of actin stress fibers and vinculin-associated adhesion plaques. SSeCKS overexpression did not affect steady-state levels of actin, vinculin, or focal adhesion kinase (FAK) but did increase integrin-independent FAK tyrosine phosphorylation. Stress fiber loss was coincident with induced SSeCKS expression, strongly suggesting a direct effect. Cytochalasin, and to a lesser extent nocodazole, inhibited SSeCKS-induced cell flattening, however, only cytochalasin affected the shape of pre-flattened cells, suggesting a greater dependence on microfilaments, rather than microtubules. By contrast, only nocodazole caused retraction of the filopodia-like processes. These data indicate a role for SSeCKS in modulating both cytoskeletal and signaling pathways. Thus, we propose to expand SSeCKS scaffolding functions to include the ability to control actin-based cytoskeletal architecture, as well as mitogenic signal pathways.


Subject(s)
Actin Cytoskeleton , Cell Cycle Proteins , Mitogens/physiology , 3T3 Cells , A Kinase Anchor Proteins , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/physiology , Animals , Calcium-Calmodulin-Dependent Protein Kinases/analysis , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Division , Cell Size , Cytochalasin D/pharmacology , Enzyme Activation , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Gene Expression Regulation/drug effects , Mice , Microfilament Proteins/analysis , Microtubules/drug effects , Mitogen-Activated Protein Kinase 1 , Mitogens/genetics , Mitogens/metabolism , Nocodazole/pharmacology , Octoxynol , Protein Kinase C , Protein-Tyrosine Kinases/metabolism , Pseudopodia , Tensins , Tetracycline/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
14.
Mol Cell Biochem ; 175(1-2): 233-41, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9350056

ABSTRACT

We recently identified a novel myristylated protein kinase C (PKC) substrate, named SSeCKS (pronounced essex), whose transcription is suppressed > 15 fold in src- or ras-transformed rodent fibroblasts, but not in raf-transformed cells [1, 2]. SSeCKS associates with and controls the elaboration of a cortical cytoskeletal matrix in response to phorbol esters [2], and overexpression of SSeCKS causes growth arrest of untransformed NIH3T3 cells [3]. Our preliminary data suggested that SSeCKS functions as a negative mitogenic regulator by controlling cytoskeletal architecture and that serine phosphorylation of SSeCKS by kinases such as PKC alters its interaction with cytoskeletal matrices and its ability to control mitogenesis. Here, we determine the effects of culture confluency, growth arrest and serum response on the steady-state abundance of SSeCKS RNA and protein and on the relative level of phosphoserine-free SSeCKS. SSeCKS transcription is initially induced by serum factors and by contact-inhibited growth rather than by cell-cycle arrest induced by serum starvation, hydroxyurea or nocodazole, and following serum-induced G1/S progression, SSeCKS transcription is suppressed. SSeCKS protein is hyperphosphorylated on serine residues during G1/S progression but not during the G2/M phase. Finally, we show that the induction of SSeCKS protein expression by contact inhibition is independent of SSeCKS' serum responsiveness. These data suggest that SSeCKS expression and function can be controlled at either the transcriptional or post-translational level in response to serum factors and culture confluency. The data strengthen the notion that SSeCKS plays an important, yet transient, role in cell cycle progression from G0 to G1 that differs from its role in controlling contact-inhibited growth.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Cycle/physiology , Fibroblasts/metabolism , Mitogens/metabolism , Protein Kinase C/metabolism , Protein Processing, Post-Translational , A Kinase Anchor Proteins , Animals , Blotting, Northern , Cell Communication , Cell Culture Techniques , Cell Cycle Proteins/genetics , Flow Cytometry , Gene Expression , Immunoblotting , Immunoenzyme Techniques , Mice , Mitogens/genetics , Phosphorylation , Phosphoserine , RNA/metabolism , Rats , Serine/metabolism
15.
Cancer Res ; 57(11): 2304-12, 1997 Jun 01.
Article in English | MEDLINE | ID: mdl-9187136

ABSTRACT

SSeCKS (pronounced essex) encodes a major protein kinase C substrate, the expression of which is down-regulated in src- and ras-transformed rodent fibroblasts but not in raf-transformed rodent fibroblasts (X. Lin et al., Mol. Cell. Biol., 15: 2754-2762, 1995). Using a panel of ras-transformed or revertant Rat-6 cells that exhibit selective parameters of transformation, we show that down-regulation of SSeCKS correlates with anchorage-independent growth. Cotransfection of NIH3T3 fibroblasts with an SSeCKS expression plasmid decreased 6-30-fold the ability of a v-src expressor plasmid to induce colonies in soft agar. To differentiate between possible tumor suppressive or growth-inhibitory effects of SSeCKS, we developed conditionally transformed cell lines (expressing ts72v-src) with tetracycline-regulated SSeCKS expression. SSeCKS suppressed the ability of v-src to induce increased cellular refractility, focus formation, soft agar colony formation, in vitro invasiveness in Matrigel, and growth in low serum (0.5%) but did not inhibit cell proliferation in high serum (10%) at the permissive (35 degrees C) temperature for src kinase activity. However, at the nonpermissive (39.5 degrees C) temperature, SSeCKS induced growth arrest. SSeCKS expression did not affect: (a) the protein level, in vivo or in vitro kinase activity of ts72src; (b) the activity of jun NH2-terminal kinase; and (c) the level of mitogen-activated protein kinase (extracellular signal-regulated kinase 2) protein. However, extracellular signal-regulated kinase 2 activity was induced 5-10-fold by SSeCKS in the presence of active src. SSeCKS reversed the ability of v-src to decrease the formation of vinculin-associated adhesion plaques, actin-based stress fibers, and filopodia structures. These data suggest a tumor suppressive role for SSeCKS via the control of cytoskeletal architecture and cell signaling.


Subject(s)
Cell Cycle Proteins , Cell Transformation, Neoplastic , Mitogens/metabolism , Oncogene Protein pp60(v-src)/metabolism , 3T3 Cells , A Kinase Anchor Proteins , Animals , Apoptosis , Blotting, Western , Cell Division , Down-Regulation , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic , Mice , Mitogens/genetics , Neoplastic Stem Cells , Oncogene Protein pp60(v-src)/genetics , Phalloidine/metabolism , Protein Kinase C/metabolism , Transcription, Genetic , Tumor Cells, Cultured , Vinculin/metabolism
16.
J Biol Chem ; 271(45): 28430-8, 1996 Nov 08.
Article in English | MEDLINE | ID: mdl-8910468

ABSTRACT

We previously identified a novel src- and ras-suppressed gene, 322, encoding a mitogenic regulatory function (Lin, X., Nelson, P. J., Frankfort, B., Tombler, E., Johnson, R., and Gelman, I. H. (1995) Mol. Cell. Biol. 15, 2754-2762). Here, we characterize the 322 gene product as an in vivo and in vitro substrate of protein kinase C (PKC). Hence, we named this product SSeCKS (pronounced essex) for Src Suppressed C Kinase Substrate. Rabbit polyclonal sera raised against glutathione S-transferase (GST)-SSeCKS recognized a myristylated 280/290-kDa doublet in Rat-6 fibroblasts. SSeCKS levels in src- and ras-transformed Rat-6 cells were 15- and 8-fold less, respectively, than those in untransformed cells. Short-term addition of phorbol ester resulted in a 5-fold increase in SSeCKS phosphorylation which was inhibited by bis-indolylmaleimide. In vitro phosphorylation of GST-SSeCKS by purified rabbit brain PKC-alpha was enhanced by phosphatidylserine and blocked by excess PKC pseudosubstrate inhibitor peptide. GST-SSeCKS bound purified PKC-alpha or PKC from Rat-6 lysates in a phosphatidylserine-dependent manner. Four SSeCKS domains containing Lys/Arg-rich motifs similar to the PKC phosphorylation site in MARCKS were phosphorylated in vitro by PKC. Immunofluorescence analysis showed SSeCKS present throughout the cytoplasm with enrichment in podosomes and at the cell edge. Short-term addition of phorbol esters caused the movement of SSeCKS from plasma membrane sites to the perinucleus coincident with a loss of actin stress fibers. These data suggest a role for SSeCKS in the control of cellular cytoskeletal architecture.


Subject(s)
Cell Cycle Proteins , Cytoskeleton/chemistry , Mitogens/metabolism , Protein Kinase C/metabolism , 3T3 Cells , A Kinase Anchor Proteins , Amino Acid Sequence , Animals , Binding Sites , Cattle , Cell Line, Transformed , Chickens , Glutathione Transferase/metabolism , Hot Temperature , Mice , Mitogens/chemistry , Molecular Sequence Data , Molecular Weight , Open Reading Frames , Phosphorylation , Protein Denaturation , Rabbits , Rats , Recombinant Fusion Proteins/metabolism
17.
Mol Cell Biol ; 15(5): 2754-62, 1995 May.
Article in English | MEDLINE | ID: mdl-7739556

ABSTRACT

In an attempt to isolate novel regulatory and/or tumor suppressor genes, we identified cDNAs whose abundance is low in NIH 3T3 cells and further decreased following the expression of the activated oncogene, v-src. The transcription of one such gene, 322, is suppressed at least 15-fold in src-, ras-, and fos-transformed cells and 3-fold in myc-transformed cells but is unaffected in raf-, mos-, or neu-transformed cells. Activation of a ts-v-src allele in confluent 3Y1 fibroblasts resulted in an initial increase in 322 mRNA levels after 1 to 2 h followed by a rapid decrease to suppressed levels after 4 to 8 h. Morphological transformation was not detected until 12 h later, indicating that the accumulation of 322 transcripts is regulated by v-src and not as a consequence of transformation. Addition of fetal calf serum to starved subconfluent NIH 3T3 or 3Y1 fibroblasts resulted in a similar biphasic regulation of 322, indicating that 322 transcription is responsive to mitogenic factors. Sequence analysis of a putative full-length 322 cDNA clone (5.4 kb) identified a large open reading frame (ORF) encoding a 148.1-kDa product. In vitro transcription and translation of the 322 cDNA from a T7 promoter resulted in a 207-kDa product whose electrophoretic mobility on a sodium dodecyl sulfate-polyacrylamide gel electrophoresis gel was unaffected by digestion with endoglycosidase F. The discrepancy in predicted versus measured molecular weights may result from the high percentage of acidic residues (roughly 20% Glu or Asp) in the 322 ORF product. Comparison of the 322 cDNA ORF with sequences in data banks indicates that this gene is novel. The 322 ORF product contains a potential Cys-1-His-3 Zn finger, at least five nuclear localization signals of the adenovirus E1a motif K(R/K)X(R/K), and alternating acidic and basic domains. Overexpression of the 322 cells resulted in the selection of rapidly growing cells which had lost the transduced 322 cDNA. Thus, 322 represent a novel src- and ras-regulated gene which encodes a potential regulator of mitogenesis and/or tumor suppressor.


Subject(s)
Genes, Regulator , Genes, Tumor Suppressor , Mitosis/genetics , Suppression, Genetic , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary/genetics , Genes, ras , Genes, src , Mice , Molecular Sequence Data , Protein Biosynthesis , Rats , Transcription, Genetic , Transformation, Genetic
18.
Biochem Biophys Res Commun ; 206(3): 916-26, 1995 Jan 26.
Article in English | MEDLINE | ID: mdl-7832805

ABSTRACT

Our aim is to identify cellular genes whose transcriptional suppression by the v-src oncogene contributes directly and specifically to the transformed and tumorigenic phenotype. We used a modified PCR-based subtractive hybridization technique to isolate 9 cDNAs whose abundance in NIH3T3 fibroblasts is decreased 3-15-fold following transformation by the activated oncogene, v-src. Sequence analysis reveals that 3 cDNAs are unlike those in GenBank. The remaining 6 cDNAs are indentical or highly similar to rat helix-destabilizing protein gene (hnRNP A1), mouse CTLA-2 alpha cysteine protease, rat cytochrome c oxidase (COX) VIc subunit, mouse Type I collagen, human gravin and a partial human cDNA (clone A7C09) isolated by random automated sequencing. Northern blot analysis indicates that the basal level of transcripts in untransformed NIH3T3 of all the genes except mouse Type I collagen was at least 10-fold lower than that of HMG Co-A Reductase, which is abundantly transcribed. These data suggest that the down-regulation of some or all of these genes contributes to v-src-induced changes in mitogenic control or cell morphology.


Subject(s)
DNA-Binding Proteins , Genes, src , Ribonucleoproteins , Suppression, Genetic , Thymus Hormones , Transcription, Genetic , Viral Proteins , 3T3 Cells , A Kinase Anchor Proteins , Animals , Base Sequence , Cell Cycle Proteins , Collagen/genetics , Cysteine Endopeptidases/genetics , DNA Helicases/genetics , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Electron Transport Complex IV/genetics , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B , Humans , Mice , Molecular Sequence Data , Nucleic Acid Hybridization , Polymerase Chain Reaction , Proteins/genetics , Sequence Analysis, DNA , Sequence Homology
19.
Oncogene ; 8(11): 2995-3004, 1993 Nov.
Article in English | MEDLINE | ID: mdl-7692370

ABSTRACT

We previously reported the development of a src-specific tumor regression system in chickens in which preinfection with rASV1702, a mutant of Rous sarcoma virus (RSV) encoding non-myristylated src product with a novel N-terminal domain, results in the immune suppression of challenge tumors induced by RSV. In order to adapt this system to the mouse, we have developed NIH3T3 and Balb/c3T3 (B3T3) cell lines that express 1702src, v-src, c-src, and other src variants, either by transfection or by infection with packaged recombinant Moloney virus (MLV) vectors. The sequence of the 1702 src cDNA, produced by reverse transcription-polymerase chain reaction (RT-PCR), confirmed the previously suggested 1702src N-terminal domain structure, fusing six amino acids from Pr76gag and 39 amino acids of env signal peptide sequence to Ala-76 of src. These cell lines were characterized for src expression and activity, cell compartmentalization of src product, tyrosine phosphorylation substrate specificity and transforming and tumorigenic potential. Based on these parameters, murine cell lines expressing 1702src were characteristically similar to chicken embryo fibroblasts (CEFs) infected with rASV1702. Finally, B3T3/1702src expressors, or membrane fractions of these cells, induced src-specific tumor protection in syngeneic mice against v-src-transformed tumor challenges. Splenic lymphocytes isolated from Balb/c mice inoculated with B3T3/1702src cells showed in vitro cytotoxicity against B3T3/v-src cells but not against untransformed B3T3 cells. Antibodies specific for Lyt2, mouse CD3 and H-2Dd blocked this cytotoxicity, whereas those specific for L3T4 did not, suggesting MHC class I-restricted, CD8-mediated cell killing. These data indicate that Balb/c3T3-expressed 1702src induces a cellular anti-tumor immunity based on src-specific tumor rejection antigens.


Subject(s)
Cell Transformation, Neoplastic , Genes, src , Neoplasms, Experimental/immunology , T-Lymphocytes, Cytotoxic/immunology , 3T3 Cells , Animals , Base Sequence , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neoplasms, Experimental/etiology , Phosphotyrosine , Tyrosine/analogs & derivatives , Tyrosine/metabolism
20.
J Virol ; 67(2): 632-42, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8380459

ABSTRACT

Analysis of the promoter for the herpes simplex virus (HSV) immediate-early (alpha) gene alpha 0 in a short-term transient expression assay revealed that a SacI-to-NcoI fragment from -786 to +148 relative to the cap site directed the synthesis of chloramphenicol acetyltransferase when the fragment was present in either orientation. Although the constitutive levels of promoter activity were similar with either orientation, the reverse-orientation promoter was not induced in response to infection with HSV. Analysis of sequences composing the putative promoter in the opposite orientation revealed the presence of important regulatory elements associated with alpha promoters. These include an alpha-trans-inducing factor (alpha-TIF)-like response element, a high-affinity ICP4-binding site, numerous Sp1-binding sites, and a TATA box. Sequences contained within this region formed specific DNA-protein complexes in extracts from mock-infected and HSV-infected HeLa cells. Transient expression assays revealed that this sequence was positively regulated by the alpha 0 and alpha-TIF genes but negatively regulated by alpha 4. Finally, nuclear run-on transcription assays revealed that this promoter is active in its correct genomic context during the course of virus infection. We suggest that the promoter is a hybrid between an alpha and beta promoter because it exhibits maximal expression at 8 h postinfection and is expressed in the presence of cycloheximide.


Subject(s)
Immediate-Early Proteins , Promoter Regions, Genetic/genetics , Repetitive Sequences, Nucleic Acid/genetics , Simplexvirus/genetics , Base Sequence , Chromosome Mapping , DNA, Viral/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , Molecular Sequence Data , Sequence Homology, Nucleic Acid , Simplexvirus/growth & development , Transcription, Genetic , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...