Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Cancer Gene Ther ; 15(7): 413-48, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18404164

ABSTRACT

ERBB3, a member of the epidermal growth factor receptor (EGFR) family, is unique in that its tyrosine kinase domain is functionally defective. It is activated by neuregulins, by other ERBB and nonERBB receptors as well as by other kinases, and by novel mechanisms. Downstream it interacts prominently with the phosphoinositol 3-kinase/AKT survival/mitogenic pathway, but also with GRB, SHC, SRC, ABL, rasGAP, SYK and the transcription regulator EBP1. There are likely important but poorly understood roles for nuclear localization and for secreted isoforms. Studies of ERBB3 expression in primary cancers and of its mechanistic contributions in cultured cells have implicated it, with varying degrees of certainty, with causation or sustenance of cancers of the breast, ovary, prostate, certain brain cells, retina, melanocytes, colon, pancreas, stomach, oral cavity and lung. Recent results link high ERBB3 activity with escape from therapy targeting other ERBBs in lung and breast cancers. Thus a wide and centrally important role for ERBB3 in cancer is becoming increasingly apparent. Several approaches for targeting ERBB3 in cancers have been tested or proposed. Small inhibitory RNA (siRNA) to ERBB3 or AKT is showing promise as a therapeutic approach to treatment of lung adenocarcinoma.


Subject(s)
Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Receptor, ErbB-3/metabolism , Humans , Neuregulins/metabolism , Protein Structure, Tertiary/genetics , RNA, Small Interfering/genetics , Receptor, ErbB-3/genetics , Signal Transduction/physiology
2.
Mol Endocrinol ; 15(5): 716-33, 2001 May.
Article in English | MEDLINE | ID: mdl-11328854

ABSTRACT

The current study investigates the activation in vivo and regulation of the expression of components of the p38 mitogen-activated protein kinase (MAPK) pathway during gonadotropin-induced formation and development of the rat corpus luteum, employing a sequential PMSG/human CG (hCG) treatment paradigm. We postulated that the p38 MAPK pathway could serve to promote phosphorylation of key substrates during luteal maturation, since maturing luteal cells, thought to be cAMP-nonresponsive, nevertheless maintain critical phosphoproteins. Both p38 MAPK and its upstream activator MAPK kinase-6 (MKK6) were found to be chronically activated during the luteal maturation phase, with activation detected by 24 h post hCG and maintained through 4 days post hCG. The p38 MAPK downstream protein kinase target termed MAPK-activated protein kinase-3 (MAPKAPK-3) was newly induced at both mRNA and protein levels during luteal formation and maturation, while mRNA and protein expression of the closely related MAPKAPK-2 diminished. Two potential substrates for MAPKAPKs, the small heat shock protein HSP-27 and the cAMP regulatory element binding protein CREB, were monitored in vivo for phosphorylation. HSP-27 phosphorylation was not modulated during luteal maturation. In contrast, we observed sustained luteal-phase CREB phosphorylation in vivo, consistent with upstream MKK6/p38 MAPK activation and MAPKAPK-3 induction. MAPKAPK-3-specific immune complex kinase assays provided direct evidence that MAPKAPK-3 was in an activated state during luteal maturation in vivo. Cellular inhibitor studies indicated that an intact p38 MAPK path was required for CREB phosphorylation in a cellular model of luteinization, as treatment of luteinized granulosa cells with the p38 MAPK inhibitor SB 203580 strongly inhibited CREB phosphorylation. Transient transfection studies provided direct evidence that MAPKAPK-3 was capable of signaling to activate CREB transcriptional activity, as assessed by means of GAL4-CREB fusion protein construct coexpressed with GAL4-luciferase reporter construct. Introduction of wild-type, but not kinase-dead mutant, MAPKAPK-3 cDNA, into a mouse ovarian cell line stimulated GAL4-CREB- dependent transcriptional activity approximately 3-fold. Thus MAPKAPK-3 is indeed uniquely poised to support luteal maturation through the phosphorylation and activation of the nuclear transcription factor CREB.


Subject(s)
Corpus Luteum/physiology , Gene Expression Regulation, Developmental , Protein Serine-Threonine Kinases/physiology , Animals , Blotting, Western , Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Corpus Luteum/enzymology , Corpus Luteum/growth & development , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activation , Female , Heat-Shock Proteins/metabolism , In Situ Hybridization , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase 3 , MAP Kinase Kinase 6 , Mitogen-Activated Protein Kinase Kinases/biosynthesis , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/biosynthesis , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Transfection , p38 Mitogen-Activated Protein Kinases
3.
Exp Lung Res ; 26(8): 659-71, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11195463

ABSTRACT

Although K-ras is mutated in many human and mouse lung adenocarcinomas, the function of K-ras p21 in lung is not known. We sought evidence for the prevalent hypothesis that K-ras p21 activates raf, which in turn passes the signal through the extracellular signal regulated kinases (Erks) to stimulate cell division, and that this pathway is upregulated when K-ras is mutated. Results from both mouse lung tumors and immortalized cultured E10 and C10 lung type II cells failed to substantiate this hypothesis. Lung tumors did not have more total K-ras p21 or K-ras p21 GTP than normal lung tissue, nor were high levels of these proteins found in tumors with mutant K-ras. Activated K-ras p21-GTP levels did not correlate with proliferating cell nuclear antigen. Special features of tumors with mutant K-ras included small size of carcinomas compared with carcinomas lacking this mutation, and correlation of proliferating cell nuclear antigen with raf-1. In nontransformed type II cells in culture, both total and activated K-ras p21 increased markedly at confluence but not after serum stimulation, whereas both Erk1/2 and the protein kinase Akt were rapidly activated by the serum treatment. Reverse transcriptase-polymerase chain reaction (RT-PCR) assays of K-ras mRNA indicated an increase in confluent and especially in postconfluent cells. Together the findings indicate that normal K-ras p21 activity is associated with growth arrest of lung type II cells, and that the exact contribution of mutated K-ras p21 to tumor development remains to be discovered.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Genes, ras , Lung Neoplasms/genetics , Lung/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma/metabolism , Animals , Cell Line, Transformed , Epithelial Cells/metabolism , Lung Neoplasms/metabolism , Mice , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/metabolism , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
4.
Oncogene ; 17(4): 493-502, 1998 Jul 30.
Article in English | MEDLINE | ID: mdl-9696043

ABSTRACT

Fetal mouse lung before gestation day 17 shows unique sensitivity to causation of rapidly growing tumors by N-ethylnitrosourea (ENU). Since mouse lung tumors present a mutated K-ras oncogene, we hypothesized that this special susceptibility might reflect an unusual vulnerability of the K-ras gene. Of the lung tumors caused by ENU exposure of BALB/c mice on gestation day 14, 8/25 had a codon 12 mutation in K-ras, vs 4/25 in codon 61. Of 15 tumors after day 16 exposure, three had codon 12 and four codon 61 changes. Tumors from day 18 exposure had only codon 61 mutations (11/16), all A:T to G:C changes (CGA). By contrast, codon 12 (GGT) changes included G:C to T:A, to A:T, and to C:G. These results show significant (P<0.01) shift in the sensitivity of particular K-ras codons to ENU mutation, during fetal mouse lung maturation. In a test of a possible relationship to expression of K-ras, K-ras p21 was measured in lungs of fetal mice, and found to increase markedly on day 18 in comparison to days 14 and 16. Both alkylation of DNA and base damage due to reactive oxygen species are postulated as mechanisms for mutation by ENU, whose efficacies vary with state of fetal lung maturation and K-ras expression.


Subject(s)
Carcinogens/pharmacology , Ethylnitrosourea/pharmacology , Genes, ras/drug effects , Lung/embryology , Mutagenesis , Mutagens/pharmacology , Adenocarcinoma, Papillary/chemically induced , Adenoma/chemically induced , Animals , Codon , Gene Expression , Lung Neoplasms/chemically induced , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-raf/genetics , ras Proteins/metabolism
5.
Mol Cell Biol ; 16(12): 6687-97, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8943323

ABSTRACT

Recently we have identified a mitogen-activated protein kinase (MAPK)-activated protein kinase, named 3pK (G. Sithanandam, F. Latif, U. Smola, R. A. Bernal, F.-M. Duh, H. Li, I. Kuzmin, V. Wixler, L. Geil, S. Shresta, P. A. Lloyd, S. Bader, Y. Sekido, K. D. Tartof, V. I. Kashuba, E. R. Zabarovsky, M. Dean, G. Klein, B. Zbar, M. I. Lerman, J. D. Minna, U. R. Rapp, and A. Allikmets, Mol. Cell. Biol. 16:868-876, 1996). In vitro characterization of the kinase revealed that 3pK is activated by ERK. It was further shown that 3pK is phosphorylated in vivo after stimulation of cells with serum. However, the in vivo relevance of this observation in terms of involvement of the Raf/MEK/ERK cascade has not been established. Here we show that 3pK is activated in vivo by the growth inducers serum and tetradecanoyl phorbol acetate in promyelocytic HL60 cells and transiently transfected embryonic kidney 293 cells. Activation of 3pK was Raf dependent and was mediated by the Raf/MEK/ERK kinase cascade. 3pK was also shown to be activated after stress stimulation of cells. In vitro studies with recombinant proteins demonstrate that in addition to ERK, members of other subgroups of the MAPK family, namely, p38RK and Jun-N-terminal kinases/stress-activated protein kinases, were also able to phosphorylate and activate 3pK. Cotransfection experiments as well as the use of a specific inhibitor of p38RK showed that these in vitro upstream activators also function in vivo, identifying 3pK as the first kinase to be activated through all three MAPK cascades. Thus, 3pK is a novel convergence point of different MAPK pathways and could function as an integrative element of signaling in both mitogen and stress responses.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Calcium-Calmodulin-Dependent Protein Kinases/genetics , HL-60 Cells , Heat Stress Disorders , Humans , Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases/genetics , Transfection
6.
Mol Cell Biol ; 16(3): 868-76, 1996 Mar.
Article in English | MEDLINE | ID: mdl-8622688

ABSTRACT

NotI linking clones, localized to the human chromosome 3p21.3 region and homozygously deleted in small cell lung cancer cell lines NCI-H740 and NCI-H1450, were used to search for a putative tumor suppressor gene(s). One of these clones, NL1G210, detected a 2.5-kb mRNA in all examined human tissues, expression being especially high in the heart and skeletal muscle. Two overlapping cDNA clones containing the entire open reading frame were isolated from a human heart cDNA library and fully characterized. Computer analysis and a search of the GenBank database to reveal high sequence identity of the product of this gene to serine-threonine kinases, especially to mitogen-activated protein kinase-activated protein kinase 2, a recently described substrate of mitogen-activated kinases. Sequence identitiy was 72% at the nucleotide level and 75% at the amino acid level, strongly suggesting that this protein is a serine-threonine kinase. Here we demonstrate that the new gene, referred to as 3pK (for chromosome 3p kinase), in fact encodes a mitogen-activated protein kinase-regulated protein serine-threonine kinase with a novel substrate specificity.


Subject(s)
Carcinoma, Small Cell/genetics , Genes, Tumor Suppressor , Lung Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Amino Acid Sequence , Base Sequence , Cloning, Molecular , DNA, Complementary/genetics , DNA, Complementary/isolation & purification , Humans , Intracellular Signaling Peptides and Proteins , Molecular Sequence Data , Protein Serine-Threonine Kinases/isolation & purification , Sequence Alignment
7.
EMBO J ; 13(7): 1610-9, 1994 Apr 01.
Article in English | MEDLINE | ID: mdl-8157000

ABSTRACT

Many growth factors whose receptors are protein tyrosine kinases stimulate the MAP kinase pathway by activating first the GTP-binding protein Ras and then the protein kinase p74raf-1. p74raf-1 phosphorylates and activates MAP kinase kinase (MAPKK). To understand the mechanism of activation of MAPKK, we have identified Ser217 and Ser221 of MAPKK1 as the sites phosphorylated by p74raf-1. This represents the first characterization of sites phosphorylated by this proto-oncogene product. Ser217 and Ser221 lie in a region of the catalytic domain where the activating phosphorylation sites of several other protein kinases are located. Among MAPKK family members, this region is the most conserved, suggesting that all members of the family are activated by the phosphorylation of these sites. A 'kinase-dead' MAPKK1 mutant was phosphorylated at the same residues as the wild-type enzyme, establishing that both sites are phosphorylated directly by p74raf-1, and not by autophosphorylation. Only the diphosphorylated form of MAPKK1 (phosphorylated at both Ser217 and Ser221) was detected, even when the stoichiometry of phosphorylation by p74raf-1 was low, indicating that phosphorylation of one of these sites is rate limiting, phosphorylation of the second then occurring extremely rapidly. Ser217 and Ser221 were both phosphorylated in vivo within minutes when PC12 cells were stimulated with nerve growth factor. Analysis of MAPKK1 mutants in which either Ser217 or Ser221 were changed to glutamic acid, and the finding that inactivation of maximally activated MAPKK1 required the dephosphorylation of both serines, shows that phosphorylation of either residue is sufficient for maximal activation.


Subject(s)
Isoenzymes/metabolism , Protein Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Serine/metabolism , Amino Acid Sequence , Animals , DNA Mutational Analysis , Enzyme Activation , Mitogen-Activated Protein Kinase Kinases , Molecular Sequence Data , Nerve Growth Factors/pharmacology , PC12 Cells , Peptide Fragments/metabolism , Phosphorylation , Protein Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-raf , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Species Specificity
8.
Mol Cell Biol ; 13(12): 7645-51, 1993 Dec.
Article in English | MEDLINE | ID: mdl-8246981

ABSTRACT

We have investigated the relationship between hydrolysis of phosphatidylcholine (PC) and activation of the Raf-1 protein kinase in Ras-mediated transduction of mitogenic signals. As previously reported, cotransfection of a PC-specific phospholipase C (PC-PLC) expression plasmid bypassed the block to cell proliferation resulting from expression of the dominant inhibitory mutant Ras N-17. In contrast, PC-PLC failed to bypass the inhibitory effect of dominant negative Raf mutants, suggesting that PC-PLC functions downstream of Ras but upstream of Raf. Consistent with this hypothesis, treatment of quiescent cells with exogenous PC-PLC induced Raf activation, even when normal Ras function was blocked by Ras N-17 expression. Further, activation of Raf in response to mitogenic growth factors was blocked by inhibition of endogenous PC-PLC. Taken together, these results indicate that hydrolysis of PC mediates Raf activation in response to mitogenic growth factors.


Subject(s)
Genes, ras , Phosphatidylcholines/metabolism , Proto-Oncogene Proteins/metabolism , 3T3 Cells , Animals , Cell Division , Enzyme Activation , Hydrolysis , Mice , Mutation , Proto-Oncogene Proteins c-raf , Signal Transduction , Transfection , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism
9.
Mol Cell Biol ; 12(9): 3733-42, 1992 Sep.
Article in English | MEDLINE | ID: mdl-1508179

ABSTRACT

B-Raf, a member of the Raf family of serine/threonine kinases, is expressed primarily in the brain and in the nervous system. In this study, the biochemical properties of the B-Raf protein were investigated in nerve growth factor (NGF)-responsive cell lines and in brain tissues. B-Raf was identified by using phosphopeptide mapping analysis and cDNA analysis as a 95-kDa protein which is primarily localized in the cytosol. NGF rapidly stimulated both serine and threonine phosphorylation in vivo and autophosphorylation activity in vitro of the B-Raf protein. In PC12 cells, B-Raf autokinase activity was induced by both differentiation factors and mitogens, with maximal activity observed after 5 min of factor addition. B-Raf kinase activity was also observed following NGF treatment of SH-SY5Y neuroblastoma cells and in adult mouse brain and hippocampus. Induction of B-Raf kinase activity in NGF-treated PC12 cells required expression of kinase-active trk receptors. Exogenous substrates or a peptide containing the autophosphorylation site became phosphorylated when added to immune complex kinase assays and reduced the in vitro autophosphorylation activity of B-Raf, suggesting that in vitro autophosphorylation sites and exogenous substrates compete for active sites of the B-Raf kinase. Finally, the major in vitro autophosphorylation site of B-Raf was identified as threonine 372 in the conserved region 2 domain. A threonine residue is present at similar positions in all three mammalian Raf family members and may represent a regulatory site for these proteins.


Subject(s)
Protein Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Amino Acid Sequence , Animals , Blotting, Western , Brain/enzymology , Enzyme Activation , Mice , Molecular Sequence Data , Nerve Growth Factors/physiology , PC12 Cells , Peptide Mapping , Phosphorylation , Protein Kinases/chemistry , Protein Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-raf
10.
Oncogene ; 7(4): 795-9, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1565476

ABSTRACT

The B-raf gene was localized to human chromosome region 7p11-7qter by Southern blot analysis of its segregation in a panel of rodent-human hybrids, using a B-raf cDNA clone. Chromosomal in situ hybridization refined the localization to 7q33-36. Analysis of genomic B-raf clones identified a B-raf pseudogene in addition to the active gene. Based on the chromosomal mapping data we conclude that the pseudogene is located near the active gene.


Subject(s)
Chromosomes, Human, Pair 7 , Proto-Oncogene Proteins/genetics , Base Sequence , Blotting, Southern , Chromosome Mapping , Humans , Molecular Sequence Data , Nucleic Acid Hybridization , Protein Kinases/genetics , Proto-Oncogene Proteins c-raf , Proto-Oncogenes , Pseudogenes , Restriction Mapping
11.
J Biol Chem ; 266(35): 23753-60, 1991 Dec 15.
Article in English | MEDLINE | ID: mdl-1748651

ABSTRACT

Treatment of PC12 cells with nerve growth factor does not alter the levels of B-raf mRNA, but does induce rapid phosphorylation of B-raf proteins. Phosphorylation was observed after 1.5 min and reached a maximum by 10-15 min. B-raf protein was phosphorylated almost exclusively on serine residues; no tyrosine phosphorylation was detected. Nerve growth factor-induced phosphorylation was not affected by depletion of protein kinase C or by removal of extracellular calcium but was inhibited by K-252a. Concomitant with the increase in serine phosphorylation, nerve growth factor treatment also increased the serine/threonine kinase activity of B-raf protein within 1-2 min.


Subject(s)
Nerve Growth Factors/pharmacology , Protein Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Amino Acid Sequence , Animals , Antibodies , Carbazoles/pharmacology , Indole Alkaloids , Kinetics , Molecular Sequence Data , Oligopeptides/chemical synthesis , Oligopeptides/immunology , PC12 Cells , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Proto-Oncogene Proteins c-raf , Substrate Specificity
12.
Oncogene ; 5(12): 1775-80, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2284096

ABSTRACT

A 2.2 kb cDNA clone which presumably includes the complete human B-raf coding sequence was isolated from a human testes cDNA library. Sequence analysis showed the presence of all three conserved regions CR1, CR2 and CR3 previously identified in raf protein kinases. The cDNA was expressed in E. coli yielding a B-raf fusion protein which reacts with antibodies specific for B-raf raised against the C-terminal peptide as well as with monoclonal antibodies which map into the kinase domain.


Subject(s)
Antibodies, Monoclonal/immunology , DNA/genetics , Isoenzymes/genetics , Protein Kinases/genetics , Proto-Oncogene Proteins/genetics , Amino Acid Sequence , Antibody Specificity , Escherichia coli , Humans , Immunoblotting , Isoenzymes/immunology , Isoenzymes/metabolism , Male , Molecular Sequence Data , Precipitin Tests , Protein Kinases/immunology , Protein Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf
13.
Mol Cell Biol ; 10(7): 3325-33, 1990 Jul.
Article in English | MEDLINE | ID: mdl-1694010

ABSTRACT

A genomic DNA fragment containing the Raf-1 promoter region was isolated by using a cDNA extension clone. Nucleotide sequencing of genomic DNA clones, primer extension, and S1 nuclease assays have been used to identify the 5' ends of Raf-1 RNAs. Consistent with its ubiquitous expression, the Raf-1 promoter region had features of a housekeeping gene in that it was GC-rich (HTF-like), lacked TATA and CAAT boxes, and contained heterogeneous RNA start sites and four potential binding sites for the transcription factor SP1. In addition, an octamer motif (ATTTCAT), a potential binding site for the octamer family of transcription factors, was located at -734 base pairs. The Raf-1 promoter region drove reporter gene expression 30-fold over the promoterless reporter in Cos 7 cells.


Subject(s)
Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Base Sequence , Blotting, Northern , Blotting, Southern , Cloning, Molecular , Gene Library , Humans , Molecular Sequence Data , Poly A/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-raf , RNA/genetics , RNA Splicing , RNA, Messenger , Restriction Mapping , T-Lymphocytes/enzymology
14.
Crit Rev Oncog ; 2(1): 1-8, 1990.
Article in English | MEDLINE | ID: mdl-2091747

ABSTRACT

There are three active raf genes in man and at least two in Xenopus and Drosophila. The mammalian c- and A-raf genes have 16 coding exons, which span 40 and 20 kb, respectively. B-raf is larger and extends over greater than 46 kb. Human c-raf-1 maps to chromosome 3p25 and A-raf-1 to Xp21. c-raf-1 RNA is present in many tissues, while A-raf and B-raf expression is restricted. A- and c-raf encode cytoplasmic ser/thr protein kinases of 68 and 74 kDa, which contain three conserved regions (CR). CR1 and 2 are in the amino terminal half, CR1 comprises the presumed ligand binding site, and CR3 represents the carboxy terminal kinase domain. All three genes can be artificially activated by deletions, provided CR3 is preserved. However, only c-raf-1 occurs naturally in truncated versions, such as v-raf and v-mil in the acutely transforming retroviruses 3611-MSV and MH2. raf transformation can also be affected by point mutation, suggesting that this mechanism may activate c-raf-1 as an oncogene in carcinogenesis.


Subject(s)
Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Animals , Humans , Neoplasms, Experimental/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-raf
15.
Oncogene ; 4(4): 451-5, 1989 Apr.
Article in English | MEDLINE | ID: mdl-2566144

ABSTRACT

The c-raf-1 oncogene is located at chromosome 3p25, near a region known to be specifically deleted in patients with renal cell carcinoma and small cell lung carcinoma (SCLC). From cytogenetic analyses of SCLC cell lines, we have estimated that one c-raf-1 allele was deleted in approximately 80% of the cases. However, c-raf-1 was generally thought to be distal to the most common deletion in SCLC, 3p14-23. Using restriction site polymorphisms (RFLPs) located within the c-raf-1 locus, we have examined DNA from 84 human lung carcinomas. In an analysis of 11 paired (normal versus tumor) SCLC DNA samples, all five informative cases showed loss of heterozygosity at this locus in the corresponding tumor sample. Analysis of 73 unpaired lung carcinoma DNAs showed that out of 31 non-SCLC samples, 19% were heterozygous for the BglI polymorphism and 25% showed heterozygosity with TaqI. However, all of the 42 SCLC samples were homozygous for both of these RFLPs. This striking loss of heterozygosity at the c-raf-1 locus in SCLC indicates that one allele of c-raf-1 is deleted in SCLC. The kinase activity of the c-raf protein appears to be constitutively activated in these cells. Whether this apparent activation results from genetic or epigenetic events is under investigation.


Subject(s)
Carcinoma, Small Cell/genetics , Chromosome Mapping , Heterozygote , Lung Neoplasms/genetics , Proto-Oncogenes , DNA/analysis , Humans , Polymorphism, Restriction Fragment Length , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins c-raf
16.
J Virol ; 62(3): 932-43, 1988 Mar.
Article in English | MEDLINE | ID: mdl-2828688

ABSTRACT

The molecular basis has been determined for differences in infectivity and XC phenotype of endogenous ecotropic murine leukemia virus of the low-leukemia mouse strain C3H/He, its relative in the high-leukemia mouse strain AKR, and highly infectious, XC-positive C3H virus variants selected in vitro. Endogenous ecotropic type C virus induced by iododeoxyuridine from the nontransformed C3H/10T1/2 cell line is XC negative and replication deficient. In contrast, viruses produced late after iododeoxyuridine induction in chemically transformed C3H/10T1/2 cells (MCA5) are XC positive and infectious. XC-negative viruses can be converted to XC-positive viruses by being grown in certain transformed cell lines. We have cloned the endogenous ecotropic provirus of C3H/He from MCA5 cells, which is XC negative and replication deficient, as well as two XC-positive C3H proviruses derived by in vitro conversion. Fragment exchange between the XC-negative molecular clone p110 and the XC-positive AKR virus clone p623 revealed that the defect in p110 lies 3' of the SalI site located in the pol region. Nucleotide sequencing established that the C3H p110 provirus was integrated within the R region of an endogenous VL30 long terminal repeat (LTR) in reverse orientation and that the virus differed from the infectious AKR p623 provirus by a point mutation, substituting Lys for Arg at the potential precursor cleavage site for gp70 and p15E. In vitro-converted XC-positive C3H proviral clones 3211 and 4211 are identical to XC-negative C3H p110, except that they have Arg at this site and the normal cleavage site is thus regenerated in these clones. The XC-negative C3H p110 was blocked in processing of Pr85env, whereas clones 3211 and 4211 had normal cleavage of the env precursor into gp70. Both the XC-negative C3H provirus and the in vitro-converted XC-positive C3H proviruses had a single copy of a 99-base-pair enhancer element in the LTR, whereas two copies of this sequence are present in the AKR proviral LTR. Substitution of Arg for Lys at the envelope precursor processing site of C3H p110 by site-directed mutagenesis is sufficient by itself to convert the virus to the XC-positive replication-competent phenotype. Thus, we have established that a single point mutation at the processing site of the envelope precursor protein Pr85 is responsible for the difference in the infectivity and XC phenotype of endogenous ecotropic murine leukemia virus from C3H/He and AKR mice and that the basis for in vitro conversion is a mutation at this site.


Subject(s)
Cell Fusion , Leukemia Virus, Murine/genetics , Mice, Inbred C3H/microbiology , Retroviridae/genetics , Viral Envelope Proteins/genetics , Virus Replication , AKR murine leukemia virus/genetics , Amino Acid Sequence , Animals , Base Sequence , Cell Line , Cell Transformation, Neoplastic/microbiology , DNA, Recombinant , Leukemia Virus, Murine/pathogenicity , Leukemia Virus, Murine/physiology , Mice , Molecular Sequence Data , Phenotype , Protein Processing, Post-Translational , Retroviridae/pathogenicity , Retroviridae/physiology , Viral Envelope Proteins/biosynthesis , Viral Envelope Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...