Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Mol Cell Biol ; 19(10): 6754-64, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10490614

ABSTRACT

Fibroblast growth factor receptors (FGFRs) are membrane-spanning tyrosine kinases that have been implicated in a variety of biological processes including mitogenesis, cell migration, development, and differentiation. We identified a unique isoform of FGFR2 expressed as a diffuse band with an unusually large molecular mass. This receptor is modified by glycosaminoglycan at a Ser residue located immediately N terminal to the acidic box, a stretch of acidic amino acids. The acidic box and the glycosaminoglycan modification site are encoded by an alternative exon of the FGFR2 gene. The acidic box appears to play an important role in glycosaminoglycan modification, and the presence of this domain is required for modification by heparan sulfate glycosaminoglycan. Moreover, the presence of the first immunoglobulin-like domain encoded by another alternative exon abrogated the modification. The high-affinity receptor with heparan sulfate modification enhanced receptor autophosphorylation, substrate phosphorylation, and ternary complex factor-independent gene expression. It also sustained mitogen-activated protein kinase activity and increased eventual DNA synthesis, a long-term response to fibroblast growth factor stimulation, at physiological ligand concentrations. We propose a novel regulation mechanism of FGFR2 signal transduction through glycosaminoglycan modification.


Subject(s)
Alternative Splicing , Glycosaminoglycans/metabolism , Immunoglobulins/metabolism , Protein Processing, Post-Translational , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Fibroblast Growth Factor/metabolism , Amino Acid Motifs , Enzyme Activation , Fibroblast Growth Factor 1 , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factors/metabolism , Ligands , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Phosphorylation , Protein Isoforms/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-fos/biosynthesis , Receptor, Fibroblast Growth Factor, Type 2 , Signal Transduction , Transcription Factors/metabolism
2.
Cancer Res ; 61(24): 8703-11, 2001 Dec 15.
Article in English | MEDLINE | ID: mdl-11751388

ABSTRACT

The focus of this study was to develop retinoic acid receptor (RAR) RAR alpha/beta selective agonists with anticancer efficacy and reduced toxicity associated with RAR gamma activity. In these studies, we report the identification and characterization of high-affinity RAR alpha/beta selective agonists with limited RAR gamma activity. These compounds inhibited human tumor cell line proliferation with similar efficacy to that observed for a pan-RAR agonist. However, for most tumor cell lines, the efficacy of these compounds was restricted to the micromolar range. To determine whether the RAR alpha/beta selective agonists could be additive or synergistic with existing agents, we investigated the effects of combining RAR alpha/beta selective agonists with various cytotoxic agents. Our results showed that the alpha/beta selective retinoids dramatically lowered the effective dose of Taxol needed to induce cytotoxicity of a wide range of tumor cell lines. This synergy was specific to tubulin-modifying agents and could not be observed with a variety of other cytotoxic agents of diverse function. Examination of pathways common to Taxol and retinoid signaling revealed that this synergy was related in part to effects on Bcl-2 expression/phosphorylation as well as the activity of the c-Jun NH(2)-terminal kinase and activator protein-1. In contrast, the tubulin polymerization induced by Taxol was not further affected by cotreatment with a variety of retinoid receptor ligands. These observations indicate that potent RAR alpha/beta selective agonists may be of therapeutic benefit in combination with Taxol therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Paclitaxel/pharmacology , Receptors, Retinoic Acid/agonists , Retinoids/pharmacology , Animals , Apoptosis/drug effects , Cell Division/drug effects , Drug Synergism , Growth Inhibitors/administration & dosage , Humans , JNK Mitogen-Activated Protein Kinases , Mice , Mitogen-Activated Protein Kinases/metabolism , Paclitaxel/administration & dosage , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/metabolism , Retinoic Acid Receptor alpha , Retinoids/administration & dosage , Substrate Specificity , Transcription Factor AP-1/metabolism , Transcriptional Activation/drug effects , Tubulin/metabolism , Tumor Cells, Cultured
3.
Oncogene ; 10(10): 2051-5, 1995 May 18.
Article in English | MEDLINE | ID: mdl-7761105

ABSTRACT

A mouse testis cDNA library in lambda pCEV27 eukaryotic expression vector was transfected in NIH3T3 fibroblasts and several transformed foci were identified. A plasmid with high-titered focus forming activity was rescued from one of these transformants. Structural analysis of this cDNA predicted a protein identical to androgen induced growth factor (AIGF), recently identified as the eighth member of the fibroblast growth factor (FGF) family. A 1.6 kilobasepair transcript of the FGF-8 gene was detected in testis but not in other adult tissues analysed. During development, expression of FGF-8 was restricted to embryonic days 9 through 13 suggesting that the growth factor plays a role during a discrete stage of mouse embryogenesis. An exon-containing genomic clone of human FGF-8 was isolated and structural comparisons indicated that the gene structure of this region is highly conserved among the FGF genes. Using a panel of human-rodent somatic cell hybrids, the FGF-8 gene was localized to human chromosome 10.


Subject(s)
Chromosomes, Human, Pair 8/genetics , DNA, Complementary/chemistry , Fibroblast Growth Factors , Growth Substances/isolation & purification , Neoplasm Proteins/isolation & purification , RNA, Messenger/isolation & purification , 3T3 Cells , Animals , Base Sequence , Chromosome Mapping , Fibroblast Growth Factor 8 , Growth Substances/chemistry , Growth Substances/genetics , Humans , Male , Mice , Molecular Sequence Data , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Open Reading Frames , Testis/chemistry , Testis/embryology
4.
Oncogene ; 18(33): 4742-55, 1999 Aug 19.
Article in English | MEDLINE | ID: mdl-10467422

ABSTRACT

A search for transforming genes expressed in brain led to the identification of a novel isoform of Ost, an exchange factor for RhoA and Cdc42. In addition to the Dbl-homology (DH) and pleckstrin-homology (PH) domains identified in the original Ost, this isoform contained a SH3 domain and a novel HIV-Tat related (TR) domain. The presence or absence of these domains in Ost defined multiple isoforms of the protein. RT - PCR and in situ hybridization analysis revealed that these isoforms were generated by tissue-specific and developmentally restricted alternative splicing events. Whereas deletion of the N-terminus activated the transforming properties of Ost, the presence of the SH3 domain reduced the transforming activity of the protein. This inhibition was relieved by the presence of a TR domain, which contained a potential SH3 ligand sequence. The transforming activity of all Ost isoforms was inhibited by dominant negative forms of the Rho family proteins. Expression of Ost isoforms potently induced the formation of actin stress fibers and filopodia as well as JNK activity and AP1- and SRF-regulated transcriptional pathways. Ost transfectants also displayed elevated levels of cyclins A and D1, suggesting that the de-regulation of these cyclins is linked to Ost-mediated transformation.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , GTP-Binding Proteins/metabolism , Nerve Tissue Proteins/metabolism , Oncogene Proteins/metabolism , Protein Isoforms/metabolism , Proteins/metabolism , 3T3 Cells , Age Factors , Amino Acid Sequence , Animals , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cloning, Molecular , Cytoskeleton , DNA-Binding Proteins/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Guanine Nucleotide Exchange Factors , Mice , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nuclear Proteins/metabolism , Oncogene Proteins/genetics , Protein Isoforms/genetics , Proteins/genetics , Pseudopodia , Rats , Sequence Homology, Amino Acid , Serum Response Factor , Signal Transduction , Tissue Distribution , Transcription Factor AP-1/metabolism , cdc42 GTP-Binding Protein , rhoA GTP-Binding Protein , src Homology Domains
5.
Oncogene ; 15(22): 2675-85, 1997 Nov 27.
Article in English | MEDLINE | ID: mdl-9400994

ABSTRACT

Members of the Ras subfamily of GTP-binding proteins, including Ras (H-, K-, and N-), TC21, and R-ras have been shown to display transforming activity, and activating lesions have been detected in human tumors. We have identified an additional member of the Ras gene family which shows significant sequence similarity to the human TC21 gene. This novel human ras-related gene, R-ras3, encodes for a protein of 209 amino acids, and shows approximately 60-75% sequence identity in the N-terminal catalytic domain with members of the Ras subfamily of GTP-binding proteins. An activating mutation corresponding to the leucine 61 oncogenic lesion of the ras oncogenes when introduced into R-ras3, activates its transforming potential. R-ras3 weakly stimulates the mitogen-activated protein kinase (MAPK) activity, but this effect is greatly potentiated by the co-expression of c-raf-1. By the yeast two-hybrid system, R-ras3 interacts only weakly with known Ras effectors, such as Raf and RalGDS, but not with RglII. In addition, R-ras3 displays modest stimulatory effects on trans-activation from different nuclear response elements which bind transcription factors, such as SRF, ETS/TCF, Jun/Fos, and NF-kappaB/Rel. Interestingly, Northern blot analysis of total RNA isolated from various tissues revealed that the 3.8 kilobasepair (kb) transcript of R-ras3 is highly restricted to the brain and heart. The close evolutionary conservation between R-ras3 and Ras family members, in contrast to the significant differences in its biological activities and the pattern of tissue expression, raise the possibility that R-ras3 may control novel cellular functions previously not described for other GTP-binding proteins.


Subject(s)
ras Proteins/genetics , ras Proteins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Transformation, Neoplastic/genetics , Cloning, Molecular , DNA, Complementary , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Humans , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid , Tissue Distribution
6.
Oncogene ; 18(40): 5497-505, 1999 Sep 30.
Article in English | MEDLINE | ID: mdl-10523826

ABSTRACT

Parathyroid adenomas are benign tumors in the parathyroid glands, whose pathogenesis is largely unknown. We utilized an expression cDNA cloning strategy to identify oncogenes activated in parathyroid adenomas. An expression cDNA library was prepared directly from a clinical sample of parathyroid adenoma tissue, transfected into NIH3T3 cells, and foci of morphologically transformed cells were isolated. Following plasmid rescue, we identified cDNAs for the keratinocyte growth factor receptor at a high frequency. Interestingly, approximately half of the clones encoded a variant receptor containing an altered C-terminus. Analysis of the transforming activity of the variant receptor revealed that the altered C-terminus up-regulated the transforming activity in a ligand-independent manner. The higher transforming activity was not accompanied by increase of dimerization or overall autophosphorylation of the receptor. However, tyrosine phosphorylation of downstream receptor substrates, including Shc isoforms and possibly FRS2, are increased in the transfectants expressing the parathyroid tumor-derived receptor. Genomic analysis showed that a previously unidentified exon was used to form the novel isoform. This alternative splicing appears to occur preferentially in parathyroid adenomas.


Subject(s)
Adenoma/genetics , Neoplasm Proteins/genetics , Parathyroid Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Fibroblast Growth Factor , Receptors, Growth Factor/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cell Transformation, Neoplastic/genetics , Cloning, Molecular , DNA, Complementary/genetics , Dimerization , Enzyme Activation , Gene Library , Humans , Hyperparathyroidism/etiology , Hyperparathyroidism, Secondary/etiology , Hyperplasia , Kidney Failure, Chronic/complications , Mice , Molecular Sequence Data , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Oncogenes , Parathyroid Glands/pathology , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Processing, Post-Translational , RNA Splicing , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Growth Factor/chemistry , Receptors, Growth Factor/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Transfection
7.
Oncogene ; 15(6): 727-35, 1997 Aug 07.
Article in English | MEDLINE | ID: mdl-9264413

ABSTRACT

The heterotrimeric G-protein, G alpha12, together with the closely-related G alpha13, are members of the G12 class of alpha-subunits important in mediating the signaling from seven transmembrane domain-spanning receptors. Recent evidence implicating both G alpha12 and G alpha13 in the activation of signaling pathways involving members of the RHO gene family led us to examine the role of Rac1, RhoA and Cdc42Hs in the transforming properties of G alpha12. Asparagine 17 (Asn 17) dominant inhibitory mutants of Rac1, and to a lesser extent RhoA, block focus forming ability of the GTPase-deficient mutant of G alpha12 (G alpha12 Leu 229) in NIH3T3 cells. In turn, wild-type G alpha12 cooperates well with Rac1 Val 12 but not with RhoA Leu 63 mutant in transforming NIH3T3 cells. Interestingly, the morphology of foci induced by G alpha12 and RhoA mutants are strikingly similar and is distinct from those displayed by Rac1 Val 12 mutant. The fact that G alpha12's ability to induce mitogenesis in NIH3T3 cells is not significantly perturbed by C3 ribosyltransferase suggested that RhoA does not play a major role in G alpha12-induced mitogenic events. Activated mutant of Rac1 has previously been demonstrated to stimulate the activity of the stress-induced c-Jun N-terminal kinase/stress-activated protein kinases (JNK/SAPKs). Transient co-transfection of Rac1 Val 12 mutant with the wild-type G alpha12 in COS7 cells leads to the further activation of an exogenously expressed hemagglutinin(HA)-tagged JNK. Furthermore, the cooperation between G alpha12 and Rac1 in cellular transformation is correlated with their ability to stimulate transcription from c-fos serum response element (SRE).


Subject(s)
GTP-Binding Protein alpha Subunits, Gi-Go , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/physiology , JNK Mitogen-Activated Protein Kinases , Mitogen-Activated Protein Kinase Kinases , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Transformation, Genetic , 3T3 Cells , Animals , Asparagine/genetics , COS Cells , Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , GTP-Binding Protein alpha Subunit, Gi2 , Hemagglutinins/metabolism , Leucine/genetics , MAP Kinase Kinase 4 , Mice , Mutagenesis , Plasmids , Poly(ADP-ribose) Polymerases/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction , Valine/genetics , cdc42 GTP-Binding Protein , rac GTP-Binding Proteins , rhoA GTP-Binding Protein
8.
Oncogene ; 15(7): 817-26, 1997 Aug 14.
Article in English | MEDLINE | ID: mdl-9266968

ABSTRACT

To assess the effect(s) of the C-terminal domain on FGFR2 function, we engineered a series of mutant FGFR2 cDNAs encoding deletions in the C-terminus of the receptor and compared their growth properties in NIH3T3 fibroblasts. In contrast to FGFR2-WT, receptors with C-terminal truncations induced ligand-independent transformation of NIH3T3 cells and transfectants expressing these mutant receptors efficiently formed colonies in semisolid medium. Introduction of point mutations (Y to F) into the C-terminus of FGFR2 at positions 813, 784 or 780 revealed that these mutant receptors also displayed activities similar to that of C-terminally truncated receptors. C-terminally altered FGF receptors did not show an increase in the basal level of receptor phosphorylation compared to that of FGFR2-WT suggesting that elevated receptor phosphorylation does not underlie the transforming activity of these receptors. Interestingly, expression of transforming FGFR2 derivatives, unlike H-Ras transformed cells, did not result in the activation of the mitogen-activated protein kinases (MAPKs), p42/ERK2 and p44/ERK1, indicating that this pathway is not constitutively active in FGFR2-transformed cells. Finally, we report the overexpression of FGFR2 mRNA and protein in several human tumor cell lines suggesting activation of the receptor in these tumors.


Subject(s)
Mitogen-Activated Protein Kinases , Peptide Fragments/physiology , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Fibroblast Growth Factor/physiology , Tyrosine/physiology , 3T3 Cells , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Colony-Forming Units Assay , Humans , Mice , Mitogen-Activated Protein Kinase 1 , Mitogen-Activated Protein Kinase 3 , Molecular Sequence Data , Peptide Fragments/genetics , Phosphorylation , Point Mutation , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Sequence Deletion , Transfection , Tumor Cells, Cultured , Tyrosine/genetics
9.
Oncogene ; 12(4): 775-84, 1996 Feb 15.
Article in English | MEDLINE | ID: mdl-8632899

ABSTRACT

The p53-regulated p21Waf1 protein is a universal inhibitor of cyclin-dependent kinases (CDKs). To study the potential tumor-suppressive properties of CDK inhibitors, the ability of p21Waf1 to interfere with oncogene-mediated cellular transformation was analysed in the NIH3T3 cell system. Cotransfection of waf1 together with activated ras or several other oncogenes into NIH3T3 cells potently inhibited the formation of transformed foci in a dose-dependent manner. Expression of the CDK-binding N-terminal half of p21Waf1 (N-p21Waf1) was necessary and sufficient to inhibit Ras-induced focus formation. In contrast, expression of the C-terminal domain (C-p21Waf1) had no effect on Ras-induced focus formation. Immunofluorescence analysis revealed that ectopically expressed p21Waf1 and C-p21Waf1 were localized in the nucleus, while N-p21Waf1 was found in the cytoplasm, with the tendency to accumulate around the nuclear membrane. Surprisingly, stable NIH3T3 transfectants expressing ectopic p21Waf1 grew at the same rate and displayed similar cell cycle distribution as NIH3T3 cells transfected with the same vector containing no insert. However, ectopic p21Waf1 expression did inhibit Ras-mediated anchorage-independent colony formation, indicating that p21Waf1 can selectively interfere with oncogene-mediated transformation without affecting NIH3T3 cell growth, at least at the levels of p21Waf1 expression achieved in these experiments. Transient transfection of waf1 into NIH3T3 cells inhibited Ras-induced transcription from a E2F-responsive element but not from a serum-responsive element, indicating that p21Waf1 acts downstream of early transcriptional events induced by Ras but upstream of E2F-controlled gene transcription. These results provide evidence that p21Waf1 potently suppresses oncogene-mediated cellular transformation of NIH3T3 cells and that it may do so by inhibiting E2F-driven transcription of S phase genes.


Subject(s)
Cell Transformation, Neoplastic , Cyclins/biosynthesis , Enzyme Inhibitors/metabolism , Genes, ras , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cell Adhesion , Cell Division , Cloning, Molecular , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , Cyclins/metabolism , DNA Primers , Epitopes/analysis , Gene Expression , Genes, p53 , Kinetics , Mice , Molecular Sequence Data , Plasmids , Polymerase Chain Reaction , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Restriction Mapping , TATA Box , Transcription, Genetic , Transfection
10.
Oncogene ; 20(55): 7965-75, 2001 Nov 29.
Article in English | MEDLINE | ID: mdl-11753679

ABSTRACT

The tumor necrosis factor (TNF) receptor family are ligand-regulated transmembrane proteins that mediate apoptosis as well as activation of the transcription factor NF-kappaB. Exogenous expression of DR6, a recently identified member of the TNF receptor family, induced apoptosis in untransformed or tumor-derived cells and the apoptotic function of DR6 was inhibited by co-expression of Bcl-2, Bcl-x(L) or the inhibitor-of-apoptosis (IAP) family member, survivin. Expression of a dominant negative mutant of FADD failed to protect from DR6-mediated apoptosis indicating that unlike TNFR1 and Fas, DR6 induced apoptosis via a FADD-independent mechanism. Despite the ability of exogenous DR6 expression to induce apoptosis, DR6 mRNA and protein were found to be elevated in prostate tumor cell lines and in advanced stages of prostate cancer. Analysis of several anti-apoptotic proteins revealed that Bcl-x(L) levels and serine 32 phosphorylation of IkappaB, the natural inhibitor of NF-kappaB, were similarly elevated in cells expressing high levels of DR6, suggesting that NF-kappaB-regulated survival proteins may protect from DR6-induced apoptosis and that DR6 is a target of NF-kappaB regulation. Treatment of LnCAP cells with TNF-alpha resulted in increases in both DR6 mRNA and protein levels, and this induction was suppressed by inhibitors of NF-kappaB. Similarly, treatment of cells expressing high levels of DR6 with indomethacin and ibuprofen, compounds also known to perturb NF-kappaB function, resulted in a dose-dependent decrease in DR6 protein and mRNA levels. These results demonstrate that TNF-alpha signaling induces the expression of a member of its own receptor family through activation of NF-kappaB.


Subject(s)
NF-kappa B/metabolism , Receptors, Tumor Necrosis Factor/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Chromosome Mapping , Chromosomes, Human, Pair 6/genetics , Gene Expression Regulation, Neoplastic/drug effects , HeLa Cells , Humans , I-kappa B Proteins/metabolism , Ibuprofen/pharmacology , Indomethacin/pharmacology , Male , NF-kappa B/antagonists & inhibitors , Phosphorylation/drug effects , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Tumor Cells, Cultured , Up-Regulation/drug effects , bcl-X Protein
11.
Oncogene ; 12(11): 2291-300, 1996 Jun 06.
Article in English | MEDLINE | ID: mdl-8649768

ABSTRACT

We compared structure, expression and functional properties of two hSos1 cDNA isoforms (IsfI and Isf II) isolated, respectively, from human fetal brain and adult skeletal muscle libraries. IsfI and IsfII nucleotide sequences differ only by the presence in IsfII of an inframe 45 hp insertion located near the first proline-rich motif required for Grb2 binding. Some human tissues express only one isoform whereas others express different proportions of both in fetal and adult stages. In vitro binding assays and in vivo functional studies showed that MI exhibits significantly higher Grb2 binding affinity and biological activity than IsfI. These results suggest that functionally different hSos1 isoforms, with differential tissue expression and distribution, play important regulatory roles in the mechanisms controlling Ras activation in different tissues and/or developmental stages.


Subject(s)
Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Brain Chemistry , DNA, Complementary/chemistry , Muscle, Skeletal/chemistry , Peptide Fragments/chemistry , Proteins/chemistry , Proteins/metabolism , Adult , Base Sequence , Fetus , GRB2 Adaptor Protein , Gene Expression Regulation , Genes, ras/genetics , Glutathione Transferase/metabolism , Guanine Nucleotide Exchange Factors , Humans , Molecular Sequence Data , Peptide Fragments/metabolism , Protein Structure, Secondary , Recombinant Fusion Proteins/metabolism , Transcription, Genetic , Transfection , Yeasts/enzymology , beta-Galactosidase/biosynthesis , ras Guanine Nucleotide Exchange Factors
12.
Oncogene ; 13(4): 731-7, 1996 Aug 15.
Article in English | MEDLINE | ID: mdl-8761294

ABSTRACT

In an effort to determine the role of protein kinase C-delta (PKC-delta) in cellular transformation mediated by the sis proto-oncogene, we cotransfected expression vectors containing cDNAs that encode for c-sis with an ATP binding mutant of PKC-delta (PKC-delta K376R) or wild type PKC-delta (PKC-delta WT) into NIH3T3 cells. Our results showed that expression of PKC-delta K376R severely impaired Sis-induced focus formation, whereas cotransfection of PKC-delta WT cDNA had no effect on Sis-mediated transformation. Consistent with this result, PKC-delta K376R expression also inhibited PDGF-BB-mediated anchorage-independent colony formation. While cotransfection of a vector containing a dominant negative mutant of ras (N17 ras) cDNA potently inhibited Sis-induced transformation, the expression of PKC-delta K376R did not block transformation mediated by v-H-Ras or v-Raf. In addition, PDGF-BB-induced Raf and mitogen-activated protein kinase activation, which are known to be downstream molecules in the Ras cascade, were not affected by the expression of PKC-delta K376R, indicating that PKC-delta and Ras are segregated in mediating Sis-induced transformation. Interestingly, expression of PKC-delta K376R strongly reduced TPA responsive element (TRE) transactivation induced by PDGF stimulation, suggesting that activation of TRE-containing genes, which may be involved in Sis-mediated transformation, are negatively regulated by expression of PKC-delta K376R.


Subject(s)
Adenosine Triphosphate/metabolism , Isoenzymes/metabolism , Platelet-Derived Growth Factor/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins/metabolism , 3T3 Cells , Animals , Base Sequence , Cell Transformation, Neoplastic/genetics , DNA Primers , Enzyme Induction , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Luciferases/biosynthesis , Mice , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Protein Binding , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C-delta , Proto-Oncogene Proteins c-sis , Receptor, Platelet-Derived Growth Factor beta , Receptors, Platelet-Derived Growth Factor/metabolism , ras Proteins/metabolism
13.
Oncogene ; 19(5): 640-8, 2000 Feb 03.
Article in English | MEDLINE | ID: mdl-10698509

ABSTRACT

Hepatocyte growth-factor (HGF) is a potent, widely produced, pleiotropic mediator of mesenchymal-epithelial interaction. In a study of changes in gene expression initiated by HGF in Balb/MK keratinocytes, we observed the induction of Neu-differentiation factor (NDF) mRNA (also known as heregulin, or HRG). Further characterization of the regulation of NDF expression in Balb/MK keratinocytes revealed potent induction by keratinocyte growth factor (KGF) and epidermal growth factor (EGF), but not by HGF/NK2, an alternative HGF isoform with motogenic but not mitogenic or morphogenic activities. Sustained treatment (8 h) of Balb/MK cells with KGF stimulated secretion of mature NDF protein into the culture medium, and Balb/ MK cells treated with purified recombinant NDF protein showed increased DNA synthesis. We also found evidence of NDF induction in two models of tissue repair in mice: in full-thickness skin wounds, following locally increased KGF production, and in kidney after partial hepatectomy, following elevation of circulating HGF levels. These results reveal that mesenchymally-derived HGF and KGF can activate autocrine NDF signaling in their epithelial targets, and suggest that this mechanism contributes to the coordination of stages of wound repair, and possibly development, where these growth factors act in concert to direct epithelial proliferation, morphogenesis and differentiation.


Subject(s)
Fibroblast Growth Factors , Growth Substances/physiology , Hepatocyte Growth Factor/physiology , Neuregulin-1/biosynthesis , Amino Acid Sequence , Animals , Cell Line , Fibroblast Growth Factor 10 , Fibroblast Growth Factor 7 , Keratinocytes/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Skin/metabolism , Skin/pathology , Wound Healing/physiology
14.
FEBS Lett ; 473(3): 285-91, 2000 May 19.
Article in English | MEDLINE | ID: mdl-10818227

ABSTRACT

A RIP-like protein, RIP3, has recently been reported that contains an N-terminal kinase domain and a novel C-terminal domain that promotes apoptosis. These experiments further characterize RIP3-mediated apoptosis and NF-kappaB activation. Northern blots indicate that rip3 mRNA displays a restricted pattern of expression including regions of the adult central nervous system. The rip3 gene was localized by fluorescent in situ hybridization to human chromosome 14q11.2, a region frequently altered in several types of neoplasia. RIP3-mediated apoptosis was inhibited by Bcl-2, Bcl-x(L), dominant-negative FADD, as well as the general caspase inhibitor Z-VAD. Further dissection of caspase involvement in RIP3-induced apoptosis indicated inhibition by the more specific inhibitors Z-DEVD (caspase-3, -6, -7, -8, and -10) and Z-VDVAD (caspase-2). However, caspase-1, -6, -8 and -9 inhibitors had little or no effect on RIP3-mediated apoptosis. Mutational analysis of RIP3 revealed that the C-terminus of RIP3 contributed to its apoptotic activity. This region is similar, but distinct, to the death domain found in many pro-apoptotic receptors and adapter proteins, including FAS, FADD, TNFR1, and RIP. Furthermore, point mutations of RIP3 at amino acids conserved among death domains, abrogated its apoptotic activity. RIP3 was localized by immunofluorescence to the mitochondrion and may play a key role in the mitochondrial disruptions often associated with apoptosis.


Subject(s)
Apoptosis/physiology , Mitochondria/chemistry , NF-kappa B/metabolism , Protein Kinases/metabolism , Amino Acid Sequence , Blotting, Northern , Caspase Inhibitors , Caspases/metabolism , Cell Nucleus/metabolism , Chromosomes, Human, Pair 14 , HeLa Cells , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Protein Structure, Tertiary , Receptor-Interacting Protein Serine-Threonine Kinases , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
15.
Free Radic Biol Med ; 28(1): 39-45, 2000 Jan 01.
Article in English | MEDLINE | ID: mdl-10656289

ABSTRACT

Homocysteine (Hcy) exerts either promoting or suppressive effects on mitogenesis in a cell type-specific manner. Hcy elicits proliferation of vascular smooth muscle cells, but is rather inhibitory to growth of endothelial cells and NIH/3T3 cells. In NIH/3T3 cells, we found that physiologically relevant concentrations (20-100 microM) of Hcy inhibit the activity of activating protein-1 (AP-1) transcription factor, although it is capable of eliciting immediate-early signaling events. Hcy induced p44/42 mitogen-activated protein kinase (MAPK) phosphorylation in control cells, but not in dominant negative p21ras transfected cells, indicating induction of the Ras-MAPK pathway. Hcy also induced the activity of serum response factor and expression of c-fos and c-jun genes. Despite the activation of these upstream events, Hcy potently inhibited AP-1 activity. Oxidized forms of Hcy (Hcy thiolactone, homocystine) were less effective in affecting AP-1. Hcy-mediated inhibition of AP-1 activity was not observed in A7r5 vascular smooth muscle cells. These results demonstrate that Hcy exerts cell type- and redox-specific inhibition of AP-1 dependent biological events.


Subject(s)
Gene Expression Regulation/drug effects , Homocysteine/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , 3T3 Cells/drug effects , 3T3 Cells/metabolism , Animals , Cell Division/drug effects , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Enzyme Activation/drug effects , Genes, Immediate-Early/drug effects , Genes, fos/drug effects , Genes, jun/drug effects , Genes, ras , JNK Mitogen-Activated Protein Kinases , MAP Kinase Signaling System/drug effects , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Organ Specificity , Oxidation-Reduction , Reverse Transcriptase Polymerase Chain Reaction , Serum Response Factor , Transfection
16.
DNA Cell Biol ; 11(8): 593-603, 1992 Oct.
Article in English | MEDLINE | ID: mdl-1388731

ABSTRACT

The product of the choline acetyltransferase (ChAT) gene is the enzyme that synthesizes the neurotransmitter acetylcholine. A 14.4-kb portion of the human ChAT gene contains 7 exons, which are estimated to comprise approximately one-third of the human protein coding sequence by comparison with porcine ChAT mRNA. Two of the exons were used to identify polyadenylated human ChAT gene transcripts on Northern blots. An exon with 84% identity to the region of porcine ChAT mRNA that codes for the amino terminus of the corresponding protein detected 6,000- and 2,300-nucleotide mRNAs in RNA isolated from human CHP134 neuroblastoma cells. Only the 2,300-nucleotide mRNA was detected by a second probe containing an exon with 96% identity to porcine ChAT mRNA in the domain that encodes amino acids 204-263 of the predicted porcine ChAT protein. Further evidence that two species of human mRNA are produced from the human ChAT gene was obtained from nuclease protection assays using an antisense RNA probe prepared from a human ChAT cDNA clone. Total RNA isolated from either CHP134 cells or adult human nucleus basalis protected 525- and 400-nucleotide fragments of this probe, confirming the presence of two species of RNA that differ by the inclusion of an internal exon. cDNA clones of each of these transcripts have been isolated. Their sequences suggest that the 2,300-nucleotide mRNA encodes enzymatically active human ChAT, while translation of the 6,000-nucleotide mRNA would be terminated prematurely by a shift in the reading frame. These results indicate that a complex pattern of transcription produces two mRNAs with different coding potentials from the human ChAT gene.


Subject(s)
Choline O-Acetyltransferase/genetics , RNA, Messenger/genetics , Blotting, Northern , Cloning, Molecular , DNA/genetics , DNA Probes , Exons , Frameshift Mutation , Gene Expression , Genes , Humans , Molecular Sequence Data , RNA Probes , RNA, Antisense , Restriction Mapping , Sequence Analysis, RNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Transcription, Genetic
17.
DNA Cell Biol ; 20(7): 383-90, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11506702

ABSTRACT

The ost protooncogene encodes a guanine nucleotide exchange factor for the Rho family of small GTPases, RhoA and Cdc42. The N-terminal domain of Ost (Ost-N) appears to negatively regulate the oncogenic activity of the protein, as deletion of this domain drastically increases its transforming activity in NIH 3T3 cells. Using a yeast two-hybrid system, we identified five genes encoding proteins that can interact with Ost-N. One of them, designated OSTIP2 (Ost interacting protein 2), encoded a previously uncharacterized protein. The OSTIP2 product is highly expressed in skeletal muscle as a 1.2-kb transcript. Full-length OSTIP2 cDNA contained an ORF of 193 amino acids. Transcription-coupled translation of OSTIP2 cDNA in reticulocyte lysates revealed a protein product of 20 kDa, which corresponded to the predicted size of the protein. Bacterially expressed glutathione S-transferase (GST)-Ostip2 fusion protein efficiently associated in vitro with baculovirus-expressed Ost. Interestingly, expression of Ostip2 in NIH 3T3 cells efficiently induced foci of morphologically transformed cells. Moreover, inoculation of athymic (nude) mice with OSTIP2 transfectants strongly induced tumor formation. These results suggest that Ostip2 is a novel oncoprotein that can interact with the Rho exchange factor Ost.


Subject(s)
Cell Transformation, Neoplastic , Guanine Nucleotide Exchange Factors/metabolism , Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , DNA, Complementary , HeLa Cells , Humans , Mice , Mice, Nude , Molecular Sequence Data , Oncogene Proteins/metabolism , Saccharomyces cerevisiae , Two-Hybrid System Techniques , rhoA GTP-Binding Protein/metabolism
18.
Neurosci Lett ; 140(2): 185-8, 1992 Jun 22.
Article in English | MEDLINE | ID: mdl-1380145

ABSTRACT

Nerve growth factor (NGF) increases the activity of choline acetyltransferase (ChAT), the synthetic enzyme for acetylcholine, in rat basal forebrain neurons both in vivo and in vitro. In poly(A)+ RNA isolated from cultures prepared from the embryonic (E15) rat basal forebrain, radiolabeled probes from the human ChAT gene detected a 3,700 nt and a less abundant 2,300 nt transcript. After growth in the presence of NGF, the abundance of both mRNAs was increased approximately twofold, paralleling the increase in ChAT enzyme activity. In vivo, the human ChAT probes detected a single 3,700 nt form of ChAT mRNA in both embryonic and adult rat basal forebrain. These results suggest that the NGF-mediated increase in ChAT activity in basal forebrain cultures is regulated at the transcriptional level.


Subject(s)
Choline O-Acetyltransferase/genetics , Gene Expression Regulation, Enzymologic/drug effects , Nerve Growth Factors/pharmacology , Prosencephalon/enzymology , Acetylcholine/biosynthesis , Animals , Blotting, Northern , Organ Culture Techniques , Poly A/genetics , Poly A/metabolism , RNA/genetics , RNA/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
19.
Cell Death Differ ; 21(2): 234-46, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24037089

ABSTRACT

We previously reported that STAT1 expression is frequently abrogated in human estrogen receptor-α-positive (ERα(+)) breast cancers and mice lacking STAT1 spontaneously develop ERα(+) mammary tumors. However, the precise mechanism by which STAT1 suppresses mammary gland tumorigenesis has not been fully elucidated. Here we show that STAT1-deficient mammary epithelial cells (MECs) display persistent prolactin receptor (PrlR) signaling, resulting in activation of JAK2, STAT3 and STAT5A/5B, expansion of CD61(+) luminal progenitor cells and development of ERα(+) mammary tumors. A failure to upregulate SOCS1, a STAT1-induced inhibitor of JAK2, leads to unopposed oncogenic PrlR signaling in STAT1(-/-) MECs. Prophylactic use of a pharmacological JAK2 inhibitor restrains the proportion of luminal progenitors and prevents disease induction. Systemic inhibition of activated JAK2 induces tumor cell death and produces therapeutic regression of pre-existing endocrine-sensitive and refractory mammary tumors. Thus, STAT1 suppresses tumor formation in mammary glands by preventing the natural developmental function of a growth factor signaling pathway from becoming pro-oncogenic. In addition, targeted inhibition of JAK2 may have significant therapeutic potential in controlling ERα(+) breast cancer in humans.


Subject(s)
Estrogen Receptor alpha/metabolism , Janus Kinase 2/metabolism , Mammary Neoplasms, Animal/metabolism , Neoplastic Stem Cells/metabolism , STAT1 Transcription Factor/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Female , Heterocyclic Compounds, 3-Ring/pharmacology , Janus Kinase 2/antagonists & inhibitors , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/genetics , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , STAT1 Transcription Factor/deficiency , Signal Transduction/drug effects , Suppressor of Cytokine Signaling 1 Protein
20.
Leukemia ; 26(2): 280-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22015772

ABSTRACT

We report the characterization of BMS-911543, a potent and selective small-molecule inhibitor of the Janus kinase (JAK) family member, JAK2. Functionally, BMS-911543 displayed potent anti-proliferative and pharmacodynamic (PD) effects in cell lines dependent upon JAK2 signaling, and had little activity in cell types dependent upon other pathways, such as JAK1 and JAK3. BMS-911543 also displayed anti-proliferative responses in colony growth assays using primary progenitor cells isolated from patients with JAK2(V617F)-positive myeloproliferative neoplasms (MPNs). Similar to these in vitro observations, BMS-911543 was also highly active in in vivo models of JAK2 signaling, with sustained pathway suppression being observed after a single oral dose. At low dose levels active in JAK2-dependent PD models, no effects were observed in an in vivo model of immunosuppression monitoring antigen-induced IgG and IgM production. Expression profiling of JAK2(V617F)-expressing cells treated with diverse JAK2 inhibitors revealed a shared set of transcriptional changes underlying pharmacological effects of JAK2 inhibition, including many STAT1-regulated genes and STAT1 itself. Collectively, our results highlight BMS-911543 as a functionally selective JAK2 inhibitor and support the therapeutic rationale for its further characterization in patients with MPN or in other disorders characterized by constitutively active JAK2 signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Heterocyclic Compounds, 3-Ring/pharmacology , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/chemistry , Blotting, Western , Cell Proliferation/drug effects , Gene Expression Profiling , Heterocyclic Compounds, 3-Ring/chemistry , Humans , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/pathology , Protein Kinase Inhibitors/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL