Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters








Publication year range
1.
Oncogene ; 32(3): 341-51, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-22370637

ABSTRACT

MicroRNAs (miRNAs) constitute a class of small non-coding RNAs that negatively regulate the expression of their target genes. They are involved in many biological processes, including cell proliferation, apoptosis and differentiation, and are considered as promising new therapeutic targets for cancer. However, the identity of miRNAs involved in apoptosis and their respective targets remain largely unknown. Given the elevated complexity of miRNA regulation of gene expression, we performed a functional screening as an alternative strategy to identify those miRNAs that in lung cancer cells may interfere with the apoptotic process. To this aim, we generated a derivative of the non-small cell lung carcinoma A549 cell line in which caspase-8, a critical upstream initiator of apoptosis, can be activated by administration of the small dimerizer drug AP20187. We found a number of miRNAs that may rescue cell viability from caspase-8 activation. They included miRNAs already described as oncogenic such as miR-17, miR-135 and miR-520, but also some miRNAs such as miR-124-1 and miR-34c for which a tumor-suppressive role has instead been described or expected. Among them, miR-34c-5p markedly increased resistance to paclitaxel-induced apoptosis. We demonstrate that Bmf (Bcl-2-modifying factor) is a target of miR-34c-5p, and that its silencing, together with that of c-myc, a known target of miR-34c-5p, contributes to resistance to apoptosis induced by paclitaxel through p53 downregulation.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MicroRNAs/genetics , Paclitaxel/pharmacology , Base Sequence , Caspase 8/metabolism , Cell Line, Tumor , Humans , MicroRNAs/metabolism
2.
J Signal Transduct ; 2012: 735135, 2012.
Article in English | MEDLINE | ID: mdl-22685651

ABSTRACT

Gliomas are the most common primary central nervous system tumors with a dismal prognosis. Despite recent advances in surgery, radiotherapy, and chemotherapy, current treatment regimens have a modest survival benefit. A crucial challenge is to deliver drugs effectively to invasive glioma cells residing in a sanctuary within the central nervous system. New therapies are essential, and oligonucleotide-based approaches, including antisense, microRNAs, small interfering RNAs, and nucleic acid aptamers, may provide a viable strategy. Thanks to their unique characteristics (low size, good affinity for the target, no immunogenicity, chemical structures that can be easily modified to improve their in vivo applications), these molecules may represent a valid alternative to antibodies particularly to overcome challenges presented by the blood-brain barrier. Here we will discuss recent results on the use of oligonucleotides that will hopefully provide new effective treatment for gliomas.

3.
Curr Med Chem ; 18(27): 4152-8, 2011.
Article in English | MEDLINE | ID: mdl-21838690

ABSTRACT

Deregulation of kinase function has been implicated in several important diseases, including cancer, neurological and metabolic disorders. Because of their key role in causing disease, kinases have become one of the most intensively pursued classes of drug targets. To date, several monoclonal antibodies (mAbs) and small-molecule inhibitors have been approved for the treatment of cancer. Aptamers are short structured single stranded RNA or DNA ligands that bind at high affinity to their target molecules and are now emerging as promising molecules to target specific cancer epitopes in clinical diagnosis and therapy. Further, because of their high specificity and low toxicity aptamers will likely reveal among the most promising molecules for in vivo targeted recognition as therapeutics or delivery agents for nanoparticles, small interfering RNAs bioconjugates, chemotherapeutic cargos and molecular imaging probes. In this article, we discuss recent advances in the development of aptamers targeting kinase proteins.


Subject(s)
Aptamers, Nucleotide/metabolism , Protein Kinases/metabolism , Animals , Drug Delivery Systems , Humans , Intracellular Space/enzymology , Intracellular Space/metabolism , Molecular Imaging , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology
4.
Endocrinology ; 144(10): 4298-305, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12959980

ABSTRACT

The Src homology 2-containing tyrosine phosphatase, Shp-2, is a crucial enzyme that mediates intracellular signaling and is implicated in cell proliferation and differentiation. Here we investigated the involvement of the Shp-2 tyrosine phosphatase in determining the downstream signaling pathways initiated by the Ret oncogene, carrying either the cysteine 634 to tyrosine or the methionine 918 to threonine substitutions. These mutations convert the receptor tyrosine kinase, Ret, into a dominant transforming protein and induce constitutive activation of its intrinsic tyrosine kinase activity leading to congenital and sporadic cancers in neuroendocrine organs. Using the PC12, rat pheochromocytoma cell line, as model system, we show that Shp-2 mediates immediate-early gene expression if induced by either of the mutant alleles. Furthermore, we show that Shp-2 activity is required for RetM918T-induced Akt activation. The results indicate that Shp-2 is a downstream mediator of the mutated receptors RetC634Y and RetM918T, thus suggesting that it may act as a limiting factor in Ret-associated endocrine tumors, in the neoplastic syndromes multiple endocrine neoplasia types 2A and 2B.


Subject(s)
Intracellular Membranes/physiology , Mutation/physiology , Oncogene Proteins/genetics , Protein Tyrosine Phosphatases/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/physiology , Animals , Cell Differentiation/physiology , Cell Line , Cell Survival/physiology , Glial Cell Line-Derived Neurotrophic Factor , Intracellular Signaling Peptides and Proteins , Nerve Growth Factors/metabolism , Oncogene Proteins/metabolism , PC12 Cells/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Proto-Oncogene Proteins c-ret , Rats , Receptor Protein-Tyrosine Kinases/metabolism
5.
Gene ; 253(1): 107-15, 2000 Jul 25.
Article in English | MEDLINE | ID: mdl-10925207

ABSTRACT

The dbl oncogene is generated by substitution of the 5' portion of its normal counterpart with an unrelated human sequence. To analyze the genomic structure and transcriptional regulation of the dbl proto-oncogene, we have isolated human genomic clones containing the entire human proto-dbl gene, localized in Xq26. Restriction mapping of a 600kb YAC clone (yWXD311) placed proto-dbl about 50kb telomeric to the coagulation Factor IX gene. The genomic DNA fragment containing the 5' end of proto-dbl was subcloned into plasmid vectors and the nucleotide sequences of exon 1, the flanking intronic region and genomic DNA 5' of the first codon were determined. Sequence analysis of 85119bp from the region revealed the genomic structure of proto-dbl. It contains 25 exons coding for a 4.7kb transcript including large 5'- and 3'- (1218bp and 701bp, respectively) untranslated regions (UTRs). RNase protection and primer extension assays on RNA from medullary thyroid carcinoma (TT) cells, which normally express dbl, revealed a transcription start site 1218bp upstream of the ATG of the first exon. A 1.6kb genomic 5' of the translation start sites drives the expression of a CAT-reporter in transient transfections in the TT cell line, though lacking TATA or CAAT boxes.


Subject(s)
Proto-Oncogene Proteins/genetics , Transcription, Genetic , X Chromosome/genetics , Base Sequence , Chloramphenicol O-Acetyltransferase/genetics , Chloramphenicol O-Acetyltransferase/metabolism , DNA/chemistry , DNA/genetics , Exons , Genes/genetics , Guanine Nucleotide Exchange Factors , Humans , Introns , Promoter Regions, Genetic , Proto-Oncogene Mas , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Analysis, DNA , Tumor Cells, Cultured
6.
Oncology ; 58(4): 311-8, 2000 May.
Article in English | MEDLINE | ID: mdl-10838497

ABSTRACT

Activating mutations of the RET oncogene cause the inheritance of multiple endocrine neoplasia type 2 (MEN2). The RET pre-mRNA is spliced into several transcripts coding for multiple isoforms, including Ret9 and Ret51. When harboring activating mutations in the cytoplasmic region, the Ret51 protein displays a higher in vitro transforming efficiency as compared to the corresponding Ret9 isoform. We investigated whether a more transforming isoform was preferentially expressed in MEN2 tumors as compared to normal tissues or sporadic pheochromocytomas. By quantitative RNases protection assays, we measured the absolute abundance of the 3' splice variants in pheochromocytomas and in normal tissues. The proportion of RET51 transcripts was highly dispersed between tumors and normal tissues. In familial tumors the proportion of RET51 transcripts was significantly larger (48.1%) than in sporadic tumors (36.75%). This result suggests that the preferential expression of the Ret51 protein isoform, even though moderate, is a growth advantage for MEN2 tumors.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Drosophila Proteins , Multiple Endocrine Neoplasia Type 2a/metabolism , Multiple Endocrine Neoplasia Type 2b/metabolism , Pheochromocytoma/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Adrenal Gland Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Germ-Line Mutation , Humans , Isomerism , Pheochromocytoma/genetics , Proto-Oncogene Proteins c-ret , RNA Probes , RNA Splicing
7.
J Biol Chem ; 275(25): 19306-14, 2000 Jun 23.
Article in English | MEDLINE | ID: mdl-10858459

ABSTRACT

Oncogenic variants of the receptor tyrosine kinase, Ret, cause formation of tumors of neuroendocrine derivation in the multiple endocrine neoplasia type 2 and, thus, likely interfere with antiproliferative and/or differentiative extracellular signals. Here we took advantage of two rat pheochromocytoma-derived cell lines (PC12/MEN2A and PC12/MEN2B) to investigate whether Ret-induced nerve growth factor (NGF) unresponsiveness might involve impairment of ERK signaling. In fact, these cells, stably transfected with distinct forms of the active ret oncogene, fail to block proliferation, even upon NGF stimulation. In these cells we show the presence of both chronic ERKs activity and high expression levels of MKP-3, an ERK-specific phosphatase. Despite the presence of MKP-3, ERK activity can be further stimulated by NGF, but it fails to translocate into the nucleus and consequently to induce immediate-early gene transcription. Because of the presence of MKP-3, our results suggest the existence of a negative regulatory feedback acting on ERKs as a mechanism responsible for the abrogation of NGF-induced terminal differentiation. Indeed, MKP-3 seems to be implicated in the persistence of ERKs in cell cytoplasm. This interpretation is further supported by the observation that in ret-transfected cells, forced expression of an active form of MEK-1 may overcome this block; it restores transcription from the c-fos promoter, induces translocation of ERKs into the nucleus, and inhibits cell proliferation.


Subject(s)
Cell Differentiation/physiology , Cell Nucleus/metabolism , Drosophila Proteins , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factor/physiology , Oncogenes , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Animals , Biological Transport , Gene Expression Regulation/physiology , PC12 Cells , Promoter Regions, Genetic , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-ret , Rats , Signal Transduction , Transfection
8.
J Biol Chem ; 275(25): 19297-305, 2000 Jun 23.
Article in English | MEDLINE | ID: mdl-10748077

ABSTRACT

Specific germline mutations of the receptor tyrosine kinase, Ret, predispose to multiple endocrine neoplasia types 2A and 2B and familial medullary thyroid carcinoma. The mechanisms by which different Ret isoforms (Ret-2A and Ret-2B) cause distinct neoplastic diseases remain largely unknown. On the other hand, forced expression of these mutated versions of Ret induces the rat pheochromocytoma cell line, PC12, to differentiate. Here we used an inducible vector encoding a dominant-negative Ras (Ras p21(N17)) to investigate the contributions of the Ras pathway to the phenotype induced in PC12 cells by the expression of either Ret-2A or Ret-2B mutants. We show that the Ret-induced molecular and morphological changes are both mediated by Ras-dependent pathways. However, even though inhibition of Ras activity was sufficient to revert Ret-induced differentiation, the kinetics of morphological reversion of the Ret-2B- was more rapid than the Ret-2A-transfected cells. Further, we show that in Ret-transfected cells the suc1-associated neurotrophic factor-induced tyrosine phosphorylation target, SNT, is chronically phosphorylated in tyrosine residues, and associates with the Sos substrate. These results indicate the activation of the Ras cascade as an essential pathway triggered by the chronic active Ret mutants in PC12 cells. Moreover, our data indicate SNT as a substrate for both Ret mutants, which might mediate the activation of this cascade.


Subject(s)
Cell Differentiation/genetics , Drosophila Proteins , Oncogene Protein p21(ras)/metabolism , Oncogenes , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Adaptor Proteins, Signal Transducing , Animals , Membrane Proteins/metabolism , Neurons/cytology , Neurons/metabolism , PC12 Cells , Phosphoproteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-ret , Rats , Tyrosine/metabolism
9.
Cancer Res ; 60(5): 1365-70, 2000 Mar 01.
Article in English | MEDLINE | ID: mdl-10728700

ABSTRACT

Pheochromocytomas are tumors originating from chromaffin cells, the large majority of which are sporadic neoplasms. The genetic and molecular events determining their tumorigenesis continue to remain unknown. On the other hand, RET germ-line mutations cause the inheritance of familial tumors in multiple endocrine neoplasia (MEN)-2 diseases, which account for a minority of pheochromocytomas. We investigated the expression of the RET gene in 14 sporadic tumors harboring no activating mutations. A subset of highly RET-expressing tumors (50%) could be distinguished. They showed RET transcript, protein amounts as well as Ret-associated phosphotyrosine levels similar to those measured in MEN-2A-associated pheochromocytomas. We also determined the GDNF and GDNF family receptor alpha (GFRalpha)-1 transcript levels in tumors and in normal tissues. Whereas the GFRalpha-1 transcripts were detected at similar levels in normal tissues and in tumors, GDNF was frequently found expressed in sporadic tumors at levels several times higher than in controls. These results led us to propose the existence of an autocrine or paracrine loop leading to chronic stimulation of the Ret signaling pathway, which could participate in the pathogenesis of a number of sporadic pheochromocytomas.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Drosophila Proteins , Nerve Growth Factors , Pheochromocytoma/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Adrenal Gland Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Pheochromocytoma/genetics , Phosphorylation , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction , Tyrosine
10.
Oncogene ; 16(19): 2435-45, 1998 May 14.
Article in English | MEDLINE | ID: mdl-9627110

ABSTRACT

The RET proto-oncogene encodes a functional receptor tyrosine kinase (Ret) for the Glial cell line Derived Neurotrophic Factor (GDNF). RET is involved in several neoplastic and non-neoplastic human diseases. Oncogenic activation of RET is detected in human papillary thyroid tumours and in multiple endocrine neoplasia type 2 syndromes. Inactivating mutations of RET have been associated to the congenital megacolon, i.e. Hirschprung's disease. In order to identify pathways that are relevant for Ret signalling to the nucleus, we have investigated its ability to induce the c-Jun NH2-terminal protein kinases (JNK). Here we show that triggering the endogenous Ret, expressed in PC12 cells, induces JNK activity; moreover, Ret is able to activate JNK either when transiently transfected in COS-1 cells or when stably expressed in NIH3T3 fibroblasts or in PC Cl 3 epithelial thyroid cells. JNK activation is dependent on the Ret kinase function, as a kinase-deficient RET mutant, associated with Hirschsprung's disease, fails to activate JNK. The pathway leading to the activation of JNK by RET is clearly divergent from that leading to the activation of ERK: substitution of the tyrosine 1062 of Ret, the Shc binding site, for phenylalanine abrogates ERK but not JNK activation. Experiments conducted with dominant negative mutants or with negative regulators demonstrate that JNK activation by Ret is mediated by Rho/Rac related small GTPases and, particularly, by Cdc42.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Drosophila Proteins , Guanine Nucleotide Dissociation Inhibitors , Guanine Nucleotide Exchange Factors , MAP Kinase Kinase 4 , Mitogen-Activated Protein Kinase Kinases , Mitogen-Activated Protein Kinases , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , 3T3 Cells , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cell Line , DNA-Binding Proteins , Enzyme Activation , Epitopes , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , GTPase-Activating Proteins , Glial Cell Line-Derived Neurotrophic Factor Receptors , JNK Mitogen-Activated Protein Kinases , Mice , Mitogen-Activated Protein Kinase 1 , Mutagenesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , PC12 Cells , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Rats , Receptor Protein-Tyrosine Kinases/genetics , Repressor Proteins , rho-Specific Guanine Nucleotide Dissociation Inhibitors
11.
Cancer Res ; 58(4): 823-8, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9485041

ABSTRACT

A subtractive thyroid cDNA library was constructed from two human thyroid carcinoma cell lines originating from an anaplastic carcinoma and a papillary thyroid carcinoma. The library was used to identify genes correlated with the progression to a highly malignant phenotype. The thymosin beta-10 gene was isolated and found to be expressed at much higher levels in the anaplastic cell line than in the papillary cells. The thymosin beta-10 gene was overexpressed in five carcinoma cell lines compared with normal thyroid tissue and normal thyroid primary culture cells. The highest expression occurred in the most malignant cell lines. Thymosin beta-10 gene expression was also increased in surgically removed human thyroid carcinomas and was highest in the anaplastic carcinomas. Thymosin beta-10 gene expression was correlated with the degree of the malignant phenotype also in rat thyroid cells transfected with cellular and viral oncogenes of different tumorigenicity. These results show that thymosin beta-10 overexpression is a general event of thyroid cell neoplastic transformation and suggest that the gene is involved in the progression of thyroid carcinogenesis. Finally, the thymosin beta-10 gene was located on chromosome 2q37 by fluorescence in situ hybridization analysis.


Subject(s)
Carcinoma, Papillary/metabolism , Carcinoma/metabolism , Thymosin/metabolism , Thyroid Neoplasms/metabolism , Animals , Carcinoma/genetics , Carcinoma, Papillary/genetics , Chromosomes, Human, Pair 2 , Gene Expression , Gene Library , Humans , Rats , Rats, Inbred F344 , Thymosin/genetics , Thyroid Gland/metabolism , Thyroid Neoplasms/genetics , Transfection , Tumor Cells, Cultured
12.
Cancer Res ; 57(3): 391-5, 1997 Feb 01.
Article in English | MEDLINE | ID: mdl-9012462

ABSTRACT

Distinct point mutations of RET, a tyrosine-kinase receptor encoding gene, are responsible for the inheritance of multiple endocrine neoplasia type 2 syndromes (MEN2A and MEN2B) and familial medullary thyroid carcinoma (FMTC). In particular, MEN2A is a more complex and aggressive disease than FMTC, being characterized by pheochromocytomas and parathyroid alterations, in addition to medullary thyroid carcinomas. The mutations associated with MEN2A and FMTC affect one of five cysteine residues mapping in the extracellular domain of the Ret protein. However, recent studies have indicated that MEN2A and FMTC disease phenotypes correlate with the position of mutations in RET. Mutations of Cys-634 are more frequent in families with MEN2A, whereas Cys-620 mutations are very rarely found in MEN2A patients and, in contrast, are frequently found in FMTC patients. We have reported previously that mutations of Cys-634 constitutively activate the RET transforming potential by causing a disulfide bridge-mediated homodimerization. Here, we report that the mutation Cys-620 --> Tyr is able to cause a constitutive dimerization of Ret, with consequent activation of its kinase and transforming activities, to a lower extent than mutation of Cys-634. We suggest that the difference in ability to activate RET shown by mutations associated with FMTC and MEN2A represents the molecular basis of the phenotypic diversity between the two syndromes.


Subject(s)
Drosophila Proteins , Gene Expression Regulation, Neoplastic , Multiple Endocrine Neoplasia Type 2a/genetics , Point Mutation , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Receptor Protein-Tyrosine Kinases/genetics , 3T3 Cells , Animals , Mice , Molecular Weight , PC12 Cells , Phenotype , Proto-Oncogene Proteins c-ret , Rats
13.
J Biol Chem ; 271(46): 29497-501, 1996 Nov 15.
Article in English | MEDLINE | ID: mdl-8910618

ABSTRACT

Inherited activating mutations of Ret, a receptor tyrosine kinase, predispose to multiple endocrine neoplasia (MEN) types 2A and 2B and familial medullary thyroid carcinoma. To investigate the effects induced by acute stimulation of Ret, we transfected both PC12 and NIH 3T3 cells with a molecular construct in which the ligand-binding domain of the epidermal growth factor receptor was fused to the catalytic domain of Ret. Acute stimulation of the chimeric receptor induced PC12 cells to express a neuronal-like phenotype. Moreover, we introduced the dominant mutation, responsible for the multiple endocrine neoplasia type 2B, in the catalytic domain of the Ret chimera. Expression of the mutant chimera, in the absence of ligand stimulation, induces the PC12 cells to acquire a flat morphology with short neuritic processes and transforms the NIH 3T3 cells. Stimulation of the mutant chimera with epidermal growth factor causes a drastic overgrowth of long neuritic processes, with the induction of the suc1-associated protein tyrosine phosphorylation in PC12 cells and higher transforming efficiency in NIH 3T3 cells. These data indicate that the gain-of-function MEN2B mutation does not abrogate ligand responsiveness of Ret and suggest that the presence of Ret ligand could play a role in the pathogenesis of the MEN2B syndrome.


Subject(s)
Drosophila Proteins , Multiple Endocrine Neoplasia Type 2b/genetics , Mutation , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Animals , Cell Differentiation/genetics , Ligands , Mice , Neurites , PC12 Cells , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Rats , Receptor Protein-Tyrosine Kinases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
14.
Science ; 273(5282): 1719-22, 1996 Sep 20.
Article in English | MEDLINE | ID: mdl-8781236

ABSTRACT

Anandamide is an endogenous ligand for central cannabinoid receptors and is released after neuronal depolarization. Anandamide increased protein tyrosine phosphorylation in rat hippocampal slices and neurons in culture. The action of anandamide resulted from the inhibition of adenylyl cyclase and cyclic adenosine 3', 5'-monophosphate-dependent protein kinase. One of the proteins phosphorylated in response to anandamide was an isoform of pp125-focal adhesion kinase (FAK+) expressed preferentially in neurons. Focal adhesion kinase is a tyrosine kinase involved in the interactions between the integrins and actin-based cytoskeleton. Thus, anandamide may exert neurotrophic effects and play a role in synaptic plasticity.


Subject(s)
Arachidonic Acids/pharmacology , Cell Adhesion Molecules/metabolism , Hippocampus/enzymology , Neurons/enzymology , Protein-Tyrosine Kinases/metabolism , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/metabolism , Amino Acid Sequence , Animals , Arachidonic Acid/pharmacology , Cell Line , Cells, Cultured , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Endocannabinoids , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Hippocampus/drug effects , In Vitro Techniques , Molecular Sequence Data , Neuronal Plasticity/drug effects , Neurons/drug effects , Phosphorylation , Phosphotyrosine/metabolism , Polyunsaturated Alkamides , Prosencephalon , Rats , Receptors, Cannabinoid , Receptors, Drug/metabolism
15.
Cell Growth Differ ; 7(8): 1081-6, 1996 Aug.
Article in English | MEDLINE | ID: mdl-8853904

ABSTRACT

The product of the RET proto-oncogene is a protein belonging to the receptor-like tyrosine kinase superfamily. RET is expressed in several neural crest-derived cell lineages and has been implicated in the correct development of the peripheral nervous system. To gain further insight into RET function, we investigated the presence of active RET in adult rat tissues. We show, by immunoblotting, that the products of the RET proto-oncogene (p155ret) are present in specific regions of adult rat brain, including the cerebellum, striatum, brainstem, hypothalamus, hippocampus, and olfactory bulb. Moreover, in the cerebellum, p155ret is phosphorylated in tyrosine residues, thus indicating that this brain structure contains p155ret in an activated state. Finally, the presence of RET in motoneurons prompted us to analyze the effects of hypoglossal nerve section on its expression. We observed a dramatic increase in p155ret in the motoneuron nuclei, thus suggesting that RET tyrosine kinase plays a role in the neuronal response to axotomy and/or during nerve regeneration.


Subject(s)
Brain/metabolism , Drosophila Proteins , Nerve Regeneration/physiology , Proto-Oncogene Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Animals , Animals, Newborn , Antibodies, Monoclonal , Brain Mapping , Cerebellum/metabolism , Ganglia, Spinal/metabolism , Hippocampus/metabolism , Hypoglossal Nerve/metabolism , Immunohistochemistry , Male , Protein-Tyrosine Kinases/analysis , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-ret , Purkinje Cells/metabolism , Rats , Rats, Sprague-Dawley , Receptor Protein-Tyrosine Kinases/physiology , Time Factors
16.
Proc Natl Acad Sci U S A ; 93(15): 7933-7, 1996 Jul 23.
Article in English | MEDLINE | ID: mdl-8755580

ABSTRACT

Germ-line missense mutations of the receptor-like tyrosine kinase ret are the causative genetic event of the multiple endocrine neoplasia (MEN) type 2A and type 2B syndromes and of the familial medullary thyroid carcinoma. We have used the rat pheochromocytoma cell line, PC12, as a model system to investigate the mechanism or mechanisms by which expression of activated ret alleles contributes to the neoplastic phenotype in neuroendocrine cells. Here we show that stable expression of ret mutants (MEN2A and MEN2B alleles) in PC12 cells causes a dramatic conversion from a round to a flat morphology, accompanied by the induction of genes belonging to the early as well as the delayed response to nerve growth factor. However, in the transfected PC12 cells, the continuous expression of neuronal specific genes is not associated with the suppression of cell proliferation. Furthermore, expression of ret mutants renders PC12 cells unresponsive to nerve growth factor-induced inhibition of proliferation. These results suggest that induction of an aberrant pattern of differentiation, accompanied by unresponsiveness to growth-inhibitory physiological signals, may be part of the mechanism of action of activated ret alleles in the pathogenesis of neuroendocrine tumors associated with MEN2 syndromes.


Subject(s)
Cell Differentiation , Drosophila Proteins , Immediate-Early Proteins , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2b/genetics , Neurons/cytology , Proto-Oncogene Proteins/physiology , Proto-Oncogenes , Receptor Protein-Tyrosine Kinases/physiology , Adrenal Gland Neoplasms , Animals , Chloramphenicol O-Acetyltransferase/biosynthesis , DNA-Binding Proteins/biosynthesis , Early Growth Response Protein 1 , Humans , Multiple Endocrine Neoplasia Type 2a/pathology , Multiple Endocrine Neoplasia Type 2b/pathology , PC12 Cells , Pheochromocytoma , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins c-ret , Rats , Receptor Protein-Tyrosine Kinases/biosynthesis , Recombinant Fusion Proteins/biosynthesis , Transcription Factors/biosynthesis , Transfection , Zinc Fingers
17.
EMBO J ; 15(11): 2717-25, 1996 Jun 03.
Article in English | MEDLINE | ID: mdl-8654369

ABSTRACT

The RET proto-oncogene encodes a receptor with tyrosine kinase activity (RET) that is involved in several neoplastic and non-neoplastic diseases. Oncogenic activation of RET, achieved by different mechanisms, is detected in a sizeable fraction of human thyroid tumors, as well as in multiple endocrine neoplasia types 2A and 2B (MEN2A and MEN2B) and familial medullary thyroid carcinoma tumoral syndromes. Germline mutations of RET have also been associated with a non-neoplastic disease, the congenital colonic aganglionosis, i.e. Hirschsprung's disease (HSCR). To analyse the impact of HSCR mutations on RET function, we have introduced into wild-type RET and activated RET(MEN2A) and RET(MEN2B) alleles three missense mutations associated with HSCR. Here we show that the three mutations caused a loss of function of RET when assayed in two model cell systems, NIH 3T3 and PC12 cells. The effect of different HSCR mutations was due to different molecular mechanisms. The HSCR972 (Arg972-->Gly) mutation, mapping in the intracytoplasmic region of RET, impaired its tyrosine kinase activity, while two extracellular mutations, HSCR32 (Ser32-->Leu) and HSCR393 (Phe393-->Leu), inhibited the biological activity of RET by impairing the correct maturation of the RET protein and its transport to the cell surface.


Subject(s)
Drosophila Proteins , Hirschsprung Disease/genetics , Immediate-Early Proteins , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , 3T3 Cells , Animals , Base Sequence , DNA Primers/chemistry , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Gene Expression Regulation, Neoplastic , Humans , Mice , Molecular Sequence Data , Nerve Growth Factors , Neuropeptides , PC12 Cells , Point Mutation , Proteins/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret , RNA, Messenger/genetics , Rats , Structure-Activity Relationship , Transcription Factors/genetics
18.
Oncol Res ; 8(2): 85-93, 1996.
Article in English | MEDLINE | ID: mdl-8859779

ABSTRACT

We have previously generated transgenic mice bearing a molecular construct obtained by fusing the rat thyroglobulin promoter with the human Kirsten ras oncogene (KRAS). These mice showed thyroid abnormalities, although at very low incidence and after a long latency period. A six-month thyrotropin (TSH) stimulation of thyroid glands, followed by a two-month suspension, induced a significant increase in the number of lesions in transgenic mice as compared with a nontransgenic control group. Our goal was to follow the progression and the reversion of the tumorigenesis process in relationship with the levels of expression of the KRAS in this experimental model. In situ hybridization was used to detect expression of KRAS mRNA in sections of thyroids of the various groups of mice. A positive hybridization was observed in follicular cells of TSH-stimulated transgenic mice, whereas no expression could be appreciated in control nontransgenic mice. A positive signal was also observed in thyroid glands excised from transgenic mice after the 2-month suspension of treatment; however, the number of expressing cells was decreased compared with transgenic mice killed immediately after 6 months of a goitrogen regimen. Finally, every cell in the single thyroid carcinoma observed after the two-month suspension was positive for the transgene mRNA. This study further strengthens the role of the expression of mutated KRAS in the early stages of thyroid follicular cell transformation and indicates that when the expression of the mutated KRAS becomes independent of exogenous TSH stimulation, this event coincides with a further progression towards tumorigenesis.


Subject(s)
Adenoma/metabolism , Carcinoma/metabolism , Gene Expression , Genes, ras , RNA, Messenger/metabolism , Thyroid Gland , Thyroid Neoplasms/metabolism , ras Proteins/metabolism , Adenoma/chemically induced , Animals , Carcinoma/chemically induced , Humans , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Transgenic , Thyroglobulin/genetics , Thyroglobulin/metabolism , Thyroid Gland/drug effects , Thyroid Neoplasms/chemically induced , Thyrotropin/pharmacology , ras Proteins/genetics
19.
Biochem Biophys Res Commun ; 216(3): 762-70, 1995 Nov 22.
Article in English | MEDLINE | ID: mdl-7488191

ABSTRACT

The dbl oncogene, generated by the truncation of the amino-terminal portion of the proto-oncogene sequence, encodes a guanine-nucleotide-releasing factor. The transforming activity of this oncogene has never been demonstrated in vivo or in vitro except in the NIH 3T3 mouse fibroblast cell line. The expression of the proto-dbl transcript is confined to tissues and tumors of neuroectodermal derivation. Therefore, to study the transforming activity of the dbl oncogene in vivo, we have generated transgenic mice that express this oncogene in neuroepithelial tissues. Mice carrying the dbl oncogene did not develop a tumor. Successively, to establish whether dbl interacts with the tumor suppressor gene p53 in tumorigenesis, we have used a p53 deficient mouse strain. The results reported here indicate that dbl is capable of causing tumor formation in vivo when its expression is driven in an appropriate cellular and genetic environment.


Subject(s)
Gene Expression , Neoplasms, Experimental/genetics , Phosphopyruvate Hydratase/genetics , Promoter Regions, Genetic , Retroviridae Proteins, Oncogenic/genetics , Tumor Suppressor Protein p53/genetics , Animals , Base Sequence , Blotting, Northern , Blotting, Southern , Crosses, Genetic , DNA Probes , Guanine Nucleotide Exchange Factors , Heterozygote , Homozygote , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Molecular Sequence Data , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/physiology
20.
Cell Growth Differ ; 6(11): 1387-94, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8562477

ABSTRACT

Expression of the RET proto-oncogene, a cell surface receptor for an as yet unknown ligand, is associated with tumors, tissues, and cell lines of neural crest origin. Accumulating evidence suggests that RET activity is involved in the process of neuronal differentiation. Moreover, induction of phenotypic differentiation of neuroblastoma cell lines is associated with the rapid accumulation of RET transcripts. To verify the role of RET in neuronal differentiation, we introduced into the human neuroblastoma cell line SK-N-BE four versions of the RET oncogene, activated by different mechanisms: RET/PTC1 and RET/PTC3, which are activated by rearrangement with heterologous genes; and two activated RET mutants, which carry the single amino acid substitution found associated to the inheritance of the multiple endocrine neoplasia type 2A (retMEN2A allele) and type2B (retMEN2B allele), respectively. We demonstrate that, after transfection with the RET oncogenes, SK-N-BE cells display a reduced growth rate and acquire a neurite-bearing phenotype accompanied by enhanced expression of the axonal growth-associated protein, GAP-43, and the high molecular weight neurofilament, NF200. These results indicate that, when activated, RET is able to cause growth inhibition and to promote neuronal differentiation of neuroblastoma cells.


Subject(s)
Drosophila Proteins , Neuroblastoma , Oncogenes/physiology , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Blotting, Northern , Cell Differentiation/physiology , Cell Division/physiology , Fluorescent Antibody Technique , Humans , Mutation/physiology , Phenotype , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , RNA, Messenger/analysis , Receptor Protein-Tyrosine Kinases/genetics , Tretinoin/pharmacology , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/physiology
SELECTION OF CITATIONS
SEARCH DETAIL