Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Br J Cancer ; 104(2): 272-80, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21179031

ABSTRACT

BACKGROUND: Pralatrexate is a dihydrofolate reductase (DHFR) inhibitor with high affinity for reduced folate carrier 1 (RFC-1) and folylpolyglutamate synthetase (FPGS), resulting in extensive internalization and accumulation in tumour cells. Pralatrexate is approved in the US for the treatment of relapsed or refractory peripheral T-cell lymphoma and is being investigated in various malignancies. Here, we evaluated molecular correlates of sensitivity to pralatrexate and explored combinations with a variety of anticancer agents. METHODS: Antiproliferative effects of pralatrexate were evaluated in 15 human-cancer cell lines using the MTT assay. Gene expression was evaluated using qRT-PCR. RESULTS: Pralatrexate and methotrexate had a similar pattern of cytotoxicity, pralatrexate being more potent. Pralatrexate potentiated the effects of platinum drugs, antimetabolites and EGFR inhibitors. Dose- and time-dependent cytotoxicity of pralatrexate correlated with high mRNA expression of FPGS. Acquired resistance to pralatrexate was associated with decreased RFC-1 expression, whereas methotrexate resistance correlated with increased DHFR expression, suggesting different mechanisms of acquired resistance. CONCLUSION: Pralatrexate was more potent than methotrexate in a panel of solid tumour lines. Our findings support the further clinical development of pralatrexate in combination with certain cytotoxics and targeted therapies, and suggest that RFC-1, FPGS and DHFR may be potential biomarkers of outcome.


Subject(s)
Aminopterin/analogs & derivatives , Antineoplastic Agents/pharmacology , Folic Acid Antagonists/pharmacology , Aminopterin/administration & dosage , Aminopterin/pharmacology , Antineoplastic Agents/administration & dosage , Apoptosis , Blotting, Western , Cell Cycle , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Folic Acid Antagonists/administration & dosage , Humans , Methotrexate/administration & dosage , Methotrexate/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
2.
Neuroscience ; 127(2): 481-96, 2004.
Article in English | MEDLINE | ID: mdl-15262337

ABSTRACT

We investigated the CNS delivery of insulin-like growth factor-I (IGF-I), a 7.65 kDa protein neurotrophic factor, following intranasal administration and the possible pathways and mechanisms underlying transport from the nasal passages to the CNS. Anesthetized adult male Sprague-Dawley rats were given [125I]-IGF-I intranasally or intravenously and then killed by perfusion-fixation within 30 min. Other animals were killed following cisternal puncture and withdrawal of cerebrospinal fluid (CSF) or intranasal administration of unlabeled IGF-I or vehicle. Both gamma counting of microdissected tissue and high resolution phosphor imaging of tissue sections showed that the tissue concentrations and distribution following intranasal administration were consistent with two routes of rapid entry into the CNS: one associated with the peripheral olfactory system connecting the nasal passages with the olfactory bulbs and rostral brain regions (e.g. anterior olfactory nucleus and frontal cortex) and the other associated with the peripheral trigeminal system connecting the nasal passages with brainstem and spinal cord regions. Intranasal administration of [125I]-IGF-I also targeted the deep cervical lymph nodes, consistent with their possible role in lymphatic drainage of both the nasal passages and the CNS. Cisternal CSF did not contain [125I]-IGF-I following intranasal administration. Intravenous [125I]-IGF-I resulted in blood and peripheral tissue exposure similar to that seen following intranasal administration but CNS concentrations were significantly lower. Finally, delivery of IGF-I into the CNS activated IGF-I signaling pathways, confirming some portion of the IGF-I that reached CNS target sites was functionally intact. The results suggest intranasally delivered IGF-I can bypass the blood-brain barrier via olfactory- and trigeminal-associated extracellular pathways to rapidly elicit biological effects at multiple sites within the brain and spinal cord.


Subject(s)
Brain/drug effects , Insulin-Like Growth Factor I/administration & dosage , Olfactory Nerve/drug effects , Olfactory Pathways/drug effects , Spinal Cord/drug effects , Trigeminal Nerve/drug effects , Administration, Intranasal , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Brain/cytology , Brain/metabolism , Cerebrospinal Fluid/metabolism , Dose-Response Relationship, Drug , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor I/pharmacokinetics , Iodine Radioisotopes , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Male , Olfactory Nerve/cytology , Olfactory Nerve/metabolism , Olfactory Pathways/cytology , Olfactory Pathways/metabolism , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reaction Time/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Cord/cytology , Spinal Cord/metabolism , Trigeminal Nerve/cytology , Trigeminal Nerve/metabolism
3.
Cell Growth Differ ; 7(4): 543-50, 1996 Apr.
Article in English | MEDLINE | ID: mdl-9052995

ABSTRACT

We have used two approaches to identify possible substrates of the insulin receptor (IR) tyrosine kinase. First, we used a potent tyrosine phosphatase inhibitor, phenylarsine oxide (PAO), which is reported to be specific for the insulin-induced signal transduction route, to augment tyrosine phosphorylation. Second, we used src homology 2 (SH2) domains fused to glutathione S-transferase as high affinity binding agents for tyrosine-phosphorylated proteins. Using the SH2 domain-containing region of p120 GTPase-activating protein and growth factor-bound protein 2, we observed a tyrosine-phosphorylated M(r) 70,000 protein in insulin- plus PAO-treated NIH3T3 cells overexpressing the IR. This M(r) 70,000 protein, which migrated as a doublet on SDS-polyacrylamide gels, efficiently bound to polyuridylic acid-Sepharose but is distinct from similar-size RNA-binding proteins such as p62 (sam68) and heterogeneous nuclear ribonucleoproteins I, K, L, and M. In addition, it differs from other M(r) 70,000 tyrosine-phosphorylated proteins, such as SH2-containing tyrosine phosphatase, raf1, and paxillin. Tyrosine phosphorylation of this protein was hardly observed after epidermal growth factor treatment. This suggests that the M(r) 70,000 protein is a novel and specific substrate for the IR kinase or an insulin-induced tyrosine kinase. The requirement for PAO to identify this tyrosine phosphorylation indicates a high turnover rate of the tyrosine phosphate.


Subject(s)
Insulin/pharmacology , Proteins/genetics , Receptor Protein-Tyrosine Kinases/metabolism , src Homology Domains/physiology , 3T3 Cells , Animals , Arsenicals/pharmacology , Enzyme Inhibitors , GRB10 Adaptor Protein , GTPase-Activating Proteins , Heterogeneous-Nuclear Ribonucleoproteins , Mice , Phosphorylation , Poly U/metabolism , Receptor Protein-Tyrosine Kinases/drug effects , Ribonucleoproteins
4.
Science ; 271(5250): 808-10, 1996 Feb 09.
Article in English | MEDLINE | ID: mdl-8628997

ABSTRACT

Genetic studies indicated that the Drosophila melanogaster protein REAPER (RPR) controls apoptosis during embryo development. Induction of RPR expression in Drosophila Schneider cells rapidly stimulated apoptosis. RPR-mediated apoptosis was blocked by N-benzyloxycarbonyl-Val-Ala-Asp-fluoromethylketone (Z-VAD-fmk), which suggests that an interleukin-1 beta converting enzyme (ICE)-like protease is required for RPR function. RPR-induced apoptosis was associated with increased ceramide production that was also blocked by Z-VAD-fmk, which suggests that ceramide generation requires an ICE-like protease as well. Thus, the intracellular RPR protein uses cell death signaling pathways similar to those used by the vertebrate transmembrane receptors Fas (CD95) and tumor necrosis factor receptor type 1.


Subject(s)
Apoptosis , Ceramides/metabolism , Cysteine Endopeptidases/metabolism , Drosophila Proteins , Drosophila melanogaster/cytology , Peptides/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Caspase 1 , Cell Line , Ceramides/pharmacology , Copper/pharmacology , Copper Sulfate , Drosophila melanogaster/embryology , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Enzyme Activation , Gene Expression , Molecular Sequence Data , Peptides/genetics , Protease Inhibitors/pharmacology , Signal Transduction , Transfection
5.
Oncogene ; 10(5): 919-25, 1995 Mar 02.
Article in English | MEDLINE | ID: mdl-7898933

ABSTRACT

Association of the p21ras guanine nucleotide exchange factor mSOS with tyrosine-phosphorylated Shc has been implicated in the activation of p21ras. In addition, after growth factor stimulation mSOS becomes phosphorylated as indicated by the appearance of a form of mSOS with reduced electrophoretic mobility. This phosphorylation is delayed with respect to Shc-Grb2-mSOS complex formation and activation of p21ras. To investigate the role of mSOS phosphorylation in further detail we have investigated the effect of phosphorylation on mSOS complex formation and p21ras activation. We found that Shc is associated with the unphosphorylated, faster migrating form of mSOS. Furthermore, although there is a correlation between the amount of complexes formed and the activation of p21ras, there is no such a correlation between mSOS phosphorylation and p21ras activation. In addition, inhibition of mSOS phosphorylation did not affect complex formation of mSOS with tyrosine phosphorylated Shc. Also, induction of mSOS phosphorylation prior to complex formation did not affect EGF-induced association of mSOS with Shc significantly, and Shc still associated predominantly with the faster migrating form of mSOS. From these results we conclude that the unphosphorylated form of mSOS is associated with Shc and that perhaps a phosphorylation-dephosphorylation step is part of the mSOS activation-inactivation cycle.


Subject(s)
Adaptor Proteins, Signal Transducing , Proteins/metabolism , 3T3 Cells , Animals , ErbB Receptors/metabolism , GRB2 Adaptor Protein , Guanine Nucleotide Exchange Factors , Mice , Phosphorylation , Proto-Oncogene Proteins p21(ras)/metabolism , Tyrosine/metabolism , ras Guanine Nucleotide Exchange Factors
7.
J Biol Chem ; 269(52): 33116-22, 1994 Dec 30.
Article in English | MEDLINE | ID: mdl-7806540

ABSTRACT

The activation of p21ras by receptor tyrosine kinases involves the translocation of the growth factor receptor bound protein 2-mammalian son of sevenless protein (Grb2-SOS) complex to the plasma membrane where p21ras is localized. Insulin receptors induce p21ras-GTP formation by two possible mechanisms: tyrosine phosphorylation of insulin receptor substrate 1 (IRS1) and its subsequent association with Grb2, or Shc phosphorylation and its subsequent association with Grb2. We investigated the contribution of the major tyrosine autophosphorylation sites Tyr1158, Tyr1162, and Tyr1163 of the insulin receptor to IRS1.Grb2 and Shc.Grb2 association and the formation of p21ras-GTP. Chinese hamster ovary-derived cell lines were used overexpressing mutant insulin receptors in which the major tyrosine autophosphorylation sites were stepwise replaced by phenylalanines. In cell lines expressing wild type or mutant Y1158F,Y1162,Y1163 (FYY) receptors, insulin stimulated tyrosine phosphorylation of IRS1 and Shc and the formation of IRS1.Grb2 and Shc.Grb2 protein complexes, together with an increase in p21ras-GTP. Cell lines expressing mutant Y1158,Y1162F,Y1163F (YFF) receptors showed insulin-induced tyrosine phosphorylation of Shc, Shc.Grb2 complex formation, and p21ras-GTP formation, whereas tyrosine phosphorylation of IRS1 was strongly decreased and formation of IRS1.Grb2 complexes was undetectable. The activity of FYY and YFF receptors to mediate p21ras-GTP formation correlated with their activity to induce Shc phosphorylation and Shc.Grb2 association. The mutant insulin receptors Y1158F,Y1162F,Y1163 and Y1158F,Y1162F,Y1163F were inactive in inducing any of these responses. We conclude that phosphorylation of Tyr1158 and Tyr1162 of the insulin receptor is linked to distinct post-receptor processes and that YFF receptors generate p21ras-GTP via the Shc.Grb2 pathway rather than one involving IRS1.Grb2 interaction.


Subject(s)
Adaptor Proteins, Signal Transducing , Guanosine Triphosphate/metabolism , Oncogene Protein p21(ras)/metabolism , Phosphoproteins/metabolism , Proteins/metabolism , Receptor, Insulin/physiology , Animals , CHO Cells , Cricetinae , GRB2 Adaptor Protein , Insulin Receptor Substrate Proteins , Mitogen-Activated Protein Kinase 1 , Mutation , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, Insulin/genetics , Tyrosine/metabolism
8.
Mol Cell Biol ; 14(3): 1575-81, 1994 Mar.
Article in English | MEDLINE | ID: mdl-8114695

ABSTRACT

Shc proteins are phosphorylated on tyrosine residues and associate with growth factor receptor-bound protein 2 (Grb2) upon treatment of cells with epidermal growth factor (EGF) or insulin. We have studied the role of Shc in insulin- and EGF-induced activation of p21ras in NIH 3T3 cells overexpressing human insulin receptors (A14 cells). A14 cells are equally responsive to insulin and EGF with respect to activation of p21ras. Analysis of Shc immunoprecipitates revealed that (i) both insulin and EGF treatment resulted in Shc tyrosine phosphorylation and (ii) Shc antibodies coimmunoprecipitated both Grb2 and mSOS after insulin and EGF treatment. The induction of tyrosine phosphorylation of Shc and the presence of Grb2 and mSOS in Shc immunoprecipitates followed similar time courses, with somewhat higher levels after EGF treatment. In mSOS immunoprecipitates, Shc could be detected as well. Furthermore, Shc immune complexes contained guanine nucleotide exchange activity toward p21ras in vitro. From these results, we conclude that after insulin and EGF treatment, Shc associates with both Grb2 and mSOS and therefore may mediate, at least in part, insulin- and EGF-induced activation of p21ras. In addition, we investigated whether the Grb2-mSOS complex associates with the insulin receptor or with insulin receptor substrate 1 (IRS1). Although we observed association of Grb2 with IRS1, we did not detect complex formation between mSOS and IRS1 in experiments in which the association of mSOS with Shc was readily detectable. Furthermore, whereas EGF treatment resulted in the association of mSOS with the EGF receptor, insulin treatment did not result in the association of mSOS with the insulin receptor. These results indicate that the association of Grb2-nSOS with Shc may be an important event in insulin-induced, mSOS-mediated activation of p21ras.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , ErbB Receptors/physiology , Proteins/physiology , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Cell Line , Epidermal Growth Factor/pharmacology , GRB2 Adaptor Protein , Guanine Nucleotides/metabolism , Membrane Proteins/metabolism , Mice , Shc Signaling Adaptor Proteins , Signal Transduction , Son of Sevenless Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Viral Proteins/metabolism
10.
EMBO J ; 12(11): 4211-20, 1993 Nov.
Article in English | MEDLINE | ID: mdl-8223435

ABSTRACT

In fibroblasts, stimulation of receptor tyrosine kinases results in the activation of the extracellular signal-regulated kinase 2 (ERK2). The major signalling pathway employed by these receptors involves the activation of p21ras and raf-1 kinase. Here we show that in NIH3T3 and rat-1 fibroblasts, elevation of the intracellular cAMP level results in the inhibition of ERK2 activation induced by PDGF, EGF and insulin treatment. Analysis of various signalling intermediates shows that cAMP interferes at a site downstream of p21ras, but upstream of raf-1 kinase. Inhibition by cAMP depends on both the cAMP concentration and the absolute amount of p21ras molecules bound to GTP, suggesting a mechanism of competitive inhibition. Also TPA-induced, p21ras-independent, activation of raf-1 kinase and ERK2 is inhibited by cAMP. We have used the inhibitory effect of cAMP to investigate whether phosphorylation of mSos, a p21ras nucleotide exchange factor, is dependent on the activity of the raf-1 kinase/ERK2 pathway. We found that phosphorylation of mSos, as monitored by a mobility shift, is delayed with respect to p21ras and ERK2 activation and is inhibited by cAMP in a similar cell type- and concentration-dependent manner as the inactivation of ERK2. These results provide evidence for a model of p21ras-directed signalling towards ERK2 that feeds back on mSos by regulating its phosphorylation status and that can be negatively modulated by protein kinase A and positively modulated by protein kinase C action.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP/pharmacology , Membrane Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Cells, Cultured , Enzyme Activation/drug effects , Growth Substances/pharmacology , Mitogen-Activated Protein Kinase 1 , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf , Rats , Signal Transduction/drug effects , Son of Sevenless Proteins
11.
Oncogene ; 8(10): 2773-80, 1993 Oct.
Article in English | MEDLINE | ID: mdl-8378086

ABSTRACT

We have investigated whether complex formation between the p21ras GTPase-activating protein (GAP) and the phosphotyrosine-containing proteins p62 and p190 is dependent on functional p21ras, to test the hypothesis that binding of p21rasGTP to GAP enables GAP to associate with these phosphoproteins. The formation of p21rasGTP was inhibited by a dominant interfering mutant of p21ras, p21ras(Asn-17), which was introduced with a vaccinia virus expression system. We used NIH3T3 cells in which complex formation between GAP and tyrosine-phosphorylated p62 and p190 can be induced either by v-src transformation, by incubating the cells with the phosphotyrosine phosphatase inhibitor pervanadate or by activation of a growth factor receptor tyrosine kinase. In all cases, expression of p21ras(Asn-17) did not affect the presence or the formation of the GAP-phosphoprotein complexes. To monitor the effectiveness of p21ras inhibition, we measured p21ras-mediated phosphorylation of extracellular signal-regulated kinase 2 (ERK2). In all cases, expression of p21ras(Asn-17) completely blocked signalling to ERK2. From these data we conclude that p21rasGTP formation is not essential for complex formation between GAP and tyrosine-phosphorylated p62 and p190, and thus complex formation does not depend on interaction of GAP with p21rasGTP.


Subject(s)
DNA-Binding Proteins/metabolism , Guanine Nucleotide Exchange Factors , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Transcription Factors, TFII , 3T3 Cells/metabolism , Animals , Cell Line, Transformed , GTPase-Activating Proteins , Mice , Mitogen-Activated Protein Kinase 1 , Phosphorylation , Repressor Proteins , Transcription Factor TFIIH , Vanadates/pharmacology , ras GTPase-Activating Proteins , ras-GRF1
12.
J Biol Chem ; 268(8): 5748-53, 1993 Mar 15.
Article in English | MEDLINE | ID: mdl-8449939

ABSTRACT

The products of the shc gene appear to be substrates for activated oncogenic tyrosine kinases, such as v-Src and v-Fps and activated tyrosine kinase receptors like the epidermal growth factor (EGF) and platelet-derived growth factor (PDGF) receptors. We investigated whether the Shc proteins are targets for the activated insulin receptor tyrosine kinase. Here we show that the 46- and 52-kDa Shc proteins are rapidly phosphorylated upon insulin receptor activation in fibroblasts expressing elevated levels of human insulin receptors. Furthermore, we observed insulin-induced association of a 23-kDa protein with the Shc proteins. These effects on Shc proteins are similar to those observed after EGF and PDGF treatment. In contrast to the observed Shc-EGF receptor association, we did not detect association between the Shc proteins and the insulin receptor. We conclude that the Shc proteins are common elements in a signal transduction pathway that is shared by EGF, PDGF, and insulin.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Insulin/pharmacology , Proteins/metabolism , 3T3 Cells , Animals , CHO Cells , Cricetinae , Epidermal Growth Factor/pharmacology , ErbB Receptors/drug effects , Humans , Mice , Phosphorylation , Receptor, Insulin/metabolism , Serine/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Tyrosine/metabolism
13.
J Biol Chem ; 267(33): 24058-63, 1992 Nov 25.
Article in English | MEDLINE | ID: mdl-1331107

ABSTRACT

We investigated the involvement of the p21ras-GTPase activating protein (GAP) in insulin-induced signal transduction. In cells overexpressing the insulin receptor, we did not observe association between GAP and the insulin receptor after insulin treatment nor the phosphorylation of GAP on tyrosine residues. However, after insulin treatment in the presence of the phosphotyrosine phosphatase inhibitor phenylarsine oxide (PAO), 5-10% of GAP was found to be associated with the insulin receptor, and, in addition, a fraction of total GAP was phosphorylated on tyrosine. Using in vitro binding we showed that the N-terminal part of GAP containing the src-homology domains 2 and 3 (SH2-SH3-SH2 region) is involved in binding to the autophosphorylated insulin receptor beta-chain. In vitro binding between GAP and the autophosphorylated insulin receptor occurred independently of PAO pretreatment. These results suggest that GAP can transiently interact with the insulin receptor after insulin treatment, and this interaction is arrested after PAO pretreatment.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Receptor, Insulin/metabolism , 3T3 Cells , Animals , Arsenicals/pharmacology , GTPase-Activating Proteins , Humans , Immunoblotting , Macromolecular Substances , Mice , Molecular Weight , Peptide Mapping , Phosphopeptides/isolation & purification , Protein Tyrosine Phosphatases/antagonists & inhibitors , Proteins/isolation & purification , Receptor, Insulin/genetics , Receptor, Insulin/isolation & purification , Transfection , Trypsin
14.
Oncogene ; 7(2): 389-94, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1549356

ABSTRACT

p21ras GAP is phosphorylated on tyrosine residues and associates with 62 kDa and 190 kDa tyrosine phosphorylated proteins in v-src-transformed fibroblasts. We were interested in identifying the tyrosine kinase responsible for phosphorylation of GAP and the two associated proteins. Here, we report that GAP-immunoprecipitates from v-src transformed cells contain a tyrosine kinase activity that phosphorylates GAP, p62 and p190. Tryptic peptide analysis indicated that the sites phosphorylated in vitro and in vivo are indistinguishable, suggesting that the precipitated kinase could be responsible for tyrosine phosphorylation of GAP in vivo. The GAP-associated kinase activity might be due to v-src itself, because pp60v-src is able to associate with GAP in vitro and GAP can be phosphorylated by pp60v-src immunecomplexes.


Subject(s)
Oncogene Protein pp60(v-src)/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , 3T3 Cells , Animals , Cell Transformation, Viral , GTPase-Activating Proteins , Macromolecular Substances , Mice , Peptide Mapping , Phosphoproteins/metabolism , Phosphorylation , Recombinant Proteins/metabolism
15.
Virology ; 176(2): 362-8, 1990 Jun.
Article in English | MEDLINE | ID: mdl-2140628

ABSTRACT

Primary baby rat kidney cells, primary human embryonic retinoblast cells, established NIH3T3 and established normal rat kidney (NRK) cells, expressing E1A and/or E1B gene regions of adenovirus 5 (Ad5) or Ad12, were investigated for susceptibility to the cytotoxic/cytostatic action of Tumor Necrosis Factor (TNF). In the primary cells and in the NRK cells, there was no correlation between TNF sensitivity and E1 gene expression; neither did sensitivity to TNF correlate with the oncogenicity of the Ad serotype. In contrast, the expression of Ad E1 gene regions in NIH3T3 cells was found to enhance TNF sensitivity of this cell line. Differences in E1A expression levels between cell types cannot explain this discrepancy regarding modulation of TNF sensitivity by E1A.


Subject(s)
Adenoviruses, Human/genetics , Gene Expression , Oncogene Proteins, Viral/genetics , Tumor Necrosis Factor-alpha/toxicity , Adenovirus Early Proteins , Animals , Cell Line , Cell Survival , Cells, Cultured , Genes, Viral , Humans , Rats , Transfection
16.
Nucleic Acids Res ; 18(1): 23-34, 1990 Jan 11.
Article in English | MEDLINE | ID: mdl-2106664

ABSTRACT

We have cloned the immediate-early serum-reponsive JE gene from the rat in order to study the regulation of this gene. We show that sequences of the JE promoter region confer serum-inducibility to a reporter gene. Analysis of the promoter in transient assays reveals that: i) the -141/-88 region is required for the response to the phorbol ester TPA, ii) the -70/-38 region is essential for basal activity. This latter region harbors the sequence TGACTCC, which resembles the consensus site for AP-1 binding, TGACTCA. DNA-protein binding assays indicate that the JE AP-1 site and the consensus AP-1 site have an overlapping but not identical binding spectrum for AP-1 proteins. Our data suggest that the inability of some AP-1 sites to respond to TPA is caused by subtle differences in affinity for AP-1 proteins.


Subject(s)
DNA-Binding Proteins/physiology , Glycoproteins/genetics , Promoter Regions, Genetic , Transcription Factors/physiology , Amino Acid Sequence , Animals , Base Sequence , Chemokine CCL2 , Cloning, Molecular , Gene Expression Regulation/drug effects , Molecular Sequence Data , Proto-Oncogene Proteins c-jun , RNA Splicing , Rats , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic
17.
J Virol ; 63(3): 1470-3, 1989 Mar.
Article in English | MEDLINE | ID: mdl-2521677

ABSTRACT

The expression of the growth factor-inducible JE gene was severely reduced in cells expressing region E1A of human adenoviruses. This reduction was not caused by a decreased stability of JE mRNA but by a reduced transcription rate as determined by run-on transcription experiments. Nevertheless, JE mRNA could still be induced by growth factors, suggesting that intracellular signaling is not blocked but rather that the basal level of JE transcription is repressed by E1A.


Subject(s)
Gene Expression Regulation , Glycoproteins/genetics , Oncogene Proteins, Viral/genetics , Transcription, Genetic , Adenovirus Early Proteins , Animals , Cell Transformation, Viral , Chemokine CCL2 , Gene Expression Regulation/drug effects , Growth Substances/pharmacology , RNA, Messenger/metabolism , Rats , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic/drug effects
18.
Histochemistry ; 84(4-6): 363-70, 1986.
Article in English | MEDLINE | ID: mdl-3522496

ABSTRACT

Fluorescein (Fl) and tetramethyl rhodamine (Rh) were evaluated as possible candidates for a double hapten sandwich system in enzyme immunohistology. Monoclonal antibodies were raised against Fl and Rh. Their fine-specificity was tested with a competition-like assay. A pair of Mab's was selected for immunohistology in which they functioned as a bridge between Fl/Rh conjugated antibodies and Fl/Rh labeled peroxidase and alkaline phosphatase, respectively. The binding of fluorescein labeled antibodies could be successfully demonstrated in histological slides. A large variability in the efficacy of staining was observed in the case of rhodamine labeled antibodies. The phenomenon is explained by assuming that tetramethyl rhodamine isothiocyanate reacts preferentially with lysine residues near to, or embedded in, hydrophobic regions in a protein. This condition may reduce the accessibility of the Rh moiety for anti-Rh antibodies.


Subject(s)
Antibodies, Monoclonal , Fluoresceins/immunology , Immunoenzyme Techniques , Rhodamines/immunology , Xanthenes/immunology , Alkaline Phosphatase , Animals , Antibody Specificity , Fluorescein , Haptens , Horseradish Peroxidase , Humans , Mice , Palatine Tonsil/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...