Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
J Hematol Oncol ; 9: 38, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27071522

ABSTRACT

BACKGROUND: There is no consensus regarding optimal treatment for peripheral T-cell lymphomas (PTCL), especially in relapsed or refractory cases, which have very poor prognosis and a dismal outcome, with 5-year overall survival of 30 %. METHODS: A multicenter prospective phase II trial was conducted to investigate the role of the combination of gemcitabine plus romidepsin (GEMRO regimen) in relapsed/refractory PTCL, looking for a potential synergistic effect of the two drugs. GEMRO regimen contemplates an induction with romidepsin plus gemcitabine for six 28-day cycles followed by maintenance with romidepsin for patients in at least partial remission. The primary endpoint was the overall response rate (ORR); secondary endpoints were survival, duration of response, and safety of the regimen. RESULTS: The ORR was 30 % (6/20) with 15 % (3) complete response (CR) rate. Two-year overall survival was 50 % and progression-free survival 11.2 %. Grade ≥3 adverse events were represented by thrombocytopenia (60 %), neutropenia (50 %), and anemia (20 %). Two patients are still in CR with median response duration of 18 months. The majority of non-hematological toxicities were mild and transient. No treatment-related death occurred and no toxicity led to treatment interruption. CONCLUSIONS: GEMRO combination regimen shows efficacy data similar to those of single-agent romidepsin with additional hematologic toxicities. Synergy observed in preclinical phase did not turn into ability to improve clinical outcomes. TRIAL REGISTRATION: The trial was registered under EudraCT 2012-001404-38; ClinicalTrials.gov number, NCT01822886 .


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphoma, T-Cell, Peripheral/drug therapy , Adult , Aged , Anemia/chemically induced , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/analogs & derivatives , Depsipeptides/administration & dosage , Depsipeptides/adverse effects , Drug Administration Schedule , Drug Resistance, Neoplasm , Female , Humans , Lymphoma, T-Cell, Peripheral/pathology , Male , Middle Aged , Neoplasm Recurrence, Local , Neutropenia/chemically induced , Prospective Studies , Remission Induction , Survival Analysis , Thrombocytopenia/chemically induced , Treatment Outcome , Young Adult , Gemcitabine
2.
Oncotarget ; 5(9): 2513-28, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24810336

ABSTRACT

Thyroid cancer incidence is rapidly increasing. Papillary Thyroid Carcinoma (PTC), the most frequent hystotype, usually displays good prognosis, but no effective therapeutic options are available for the fraction of progressive PTC patients. BRAF and RET/PTC are the most frequent driving genetic lesions identified in PTC. We developed two complementary in vitro models based on RET/PTC1 oncogene, starting from the hypothesis that miRNAs modulated by a driving PTC-oncogene are likely to have a role in thyroid neoplastic processes. Through this strategy, we identified a panel of deregulated miRNAs. Among these we focused on miR-199a-3p and showed its under-expression in PTC specimens and cell lines. We demonstrated that miR-199a-3p restoration in PTC cells reduces MET and mTOR protein levels, impairs migration and proliferation and, more interesting, induces lethality through an unusual form of cell death similar to methuosis, caused by macropinocytosis dysregulation. Silencing MET or mTOR, both involved in survival pathways, does not recapitulate miR-199a-3p-induced cell lethality, thus suggesting that the cooperative regulation of multiple gene targets is necessary. Integrated analysis of miR-199a-3p targets unveils interesting networks including HGF and macropinocytosis pathways. Overall our results indicate miR-199a-3p as a tumor suppressor miRNA in PTC.


Subject(s)
Apoptosis , Carcinoma, Papillary/genetics , Cell Movement , Genes, Tumor Suppressor , MicroRNAs/genetics , Thyroid Neoplasms/genetics , Blotting, Western , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Cell Proliferation , Cells, Cultured , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Pinocytosis , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Thyroid Gland/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
3.
Endocr Relat Cancer ; 20(1): 23-37, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23132790

ABSTRACT

Thyroid carcinomas derived from follicular cells comprise papillary thyroid carcinoma (PTC), follicular thyroid carcinoma, poorly differentiated thyroid carcinoma (PDTC) and undifferentiated anaplastic thyroid carcinoma (ATC). PTC, the most frequent thyroid carcinoma histotype, is associated with gene rearrangements that generate RET/PTC and TRK oncogenes and with BRAF-V600E and RAS gene mutations. These last two genetic lesions are also present in a fraction of PDTCs. The ERK1/2 pathway, downstream of the known oncogenes activated in PTC, has a central role in thyroid carcinogenesis. In this study, we demonstrate that the BRAF-V600E, RET/PTC, and TRK oncogenes upregulate the ERK1/2 pathway's attenuator cytoplasmic dual-phase phosphatase DUSP6/MKP3 in thyroid cells. We also show DUSP6 overexpression at the mRNA and protein levels in all the analysed PTC cell lines. Furthermore, DUSP6 mRNA was significantly higher in PTC and PDTC in comparison with normal thyroid tissues both in expression profile datasets and in patients' surgical samples analysed by real-time RT-PCR. Immunohistochemical and western blot analyses showed that DUSP6 was also overexpressed at the protein level in most PTC and PDTC surgical samples tested, but not in ATC, and revealed a positive correlation trend with ERK1/2 pathway activation. Finally, DUSP6 silencing reduced the neoplastic properties of four PTC cell lines, thus suggesting that DUSP6 may have a pro-tumorigenic role in thyroid carcinogenesis.


Subject(s)
Adenocarcinoma, Follicular/metabolism , Biomarkers, Tumor/genetics , Carcinoma, Papillary/metabolism , Cell Differentiation , Dual Specificity Phosphatase 6/metabolism , Thyroid Neoplasms/metabolism , Adenocarcinoma, Follicular/genetics , Adenocarcinoma, Follicular/pathology , Adult , Aged , Aged, 80 and over , Apoptosis , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Dual Specificity Phosphatase 6/genetics , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Gland/cytology , Thyroid Gland/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology
4.
Endocr Relat Cancer ; 18(4): 519-27, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21690267

ABSTRACT

Activating mutations of RET, a gene encoding two isoforms of a tyrosine kinase receptor physiologically expressed in several neural crest-derived cell lineages, are associated with the inherited forms of medullary thyroid carcinoma (MTC). The identification and characterization of novel RET mutations involved in MTC is valuable, as RET gene testing plays a crucial role in the management of these patients. In an MTC patient, we have identified a germline c.1996A>G transition in heterozygosis leading to K666E substitution. In addition, the conservative S904S (c.2712C>G) and the non-conservative functional G691S (c.2071G>A) polymorphisms have been identified. Through functional studies, we demonstrate for the first time that K666E is a gain-of-function mutation with oncogenic potential, based on its ability to transform NIH3T3 cells. It was not possible to define whether K666E is a de novo or inherited RET variant in the patient, as the family history was negative for MTC, and the carrier status of family members could not be tested. Our results, together with a recent report of co-segregation of the mutation in three MTC families, suggest that K666E is a causative MTC mutation. As we have shown that the same patient allele carries both K666E and G691S variants, the latter known to increase downstream RET signaling, a possible role for the G691S polymorphism has also been investigated. We have demonstrated that, although RET-G691S is not oncogenic per se, it enhances the transforming activity of the RET-K666E mutant, thus suggesting a modifier role for this functional polymorphism.


Subject(s)
Cell Transformation, Neoplastic , Germ-Line Mutation/genetics , Neoplastic Syndromes, Hereditary/genetics , Polymorphism, Genetic/genetics , Proto-Oncogene Proteins c-ret/genetics , Thyroid Neoplasms/genetics , Animals , Blotting, Western , Carcinoma, Medullary/congenital , Cells, Cultured , DNA, Neoplasm/genetics , Female , Heterozygote , Humans , Mice , Middle Aged , Multiple Endocrine Neoplasia Type 2a , NIH 3T3 Cells , Neoplastic Syndromes, Hereditary/metabolism , Neoplastic Syndromes, Hereditary/pathology , Polymerase Chain Reaction , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
5.
PLoS One ; 5(9): e12701, 2010 Sep 22.
Article in English | MEDLINE | ID: mdl-20877637

ABSTRACT

BACKGROUND: Papillary thyroid carcinoma (PTCs), the most frequent thyroid cancer, is usually not life threatening, but may recur or progress to aggressive forms resistant to conventional therapies. A more detailed understanding of the signaling pathways activated in PTCs may help to identify novel therapeutic approaches against these tumors. The aim of this study is to identify signaling pathways activated in PTCs. METHODOLOGY/PRINCIPAL FINDINGS: We examined coordinated gene expression patterns of ligand/receptor (L/R) pairs using the L/R database DRLP-rev1 and five publicly available thyroid cancer datasets of gene expression on a total of 41 paired PTC/normal thyroid tissues. We identified 26 (up) and 13 (down) L/R pairs coordinately and differentially expressed. The relevance of these L/R pairs was confirmed by performing the same analysis on REarranged during Transfection (RET)/PTC1-infected thyrocytes with respect to normal thyrocytes. TGFA/EGFR emerged as one of the most tightly regulated L/R pair. Furthermore, PTC clinical samples analyzed by real-time RT-PCR expressed EGFR transcript levels similar to those of 5 normal thyroid tissues from patients with pathologies other than thyroid cancer, whereas significantly elevated levels of TGFA transcripts were only present in PTCs. Biochemical analysis of PTC cell lines demonstrated the presence of EGFR on the cell membrane and TGFA in conditioned media. Moreover, conditioned medium of the PTC cell line NIM-1 activated EGFR expressed on HeLa cells, culminating in both ERK and AKT phosphorylation. In NIM-1 cells harboring BRAF mutation, TGFA stimulated proliferation, contributing to PI3K/AKT activation independent of MEK/ERK signaling. CONCLUSIONS/SIGNIFICANCE: We compiled a reliable list of L/R pairs associated with PTC and validated the biological role of one of the emerged L/R pair, the TGFA/EGFR, in this cancer, in vitro. These data provide a better understanding of the factors involved in the biology of PTCs and would be useful in developing combination therapeutic approaches against these cancers.


Subject(s)
Carcinoma, Papillary/metabolism , Computational Biology , ErbB Receptors/metabolism , Signal Transduction , Thyroid Neoplasms/metabolism , Transforming Growth Factor alpha/metabolism , Carcinoma, Papillary/genetics , Carcinoma, Papillary/pathology , Carcinoma, Papillary/physiopathology , Cell Line, Tumor , Cell Proliferation , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Humans , Ligands , Protein Binding , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Thyroid Neoplasms/physiopathology , Transforming Growth Factor alpha/genetics
6.
Clin Endocrinol (Oxf) ; 72(5): 702-8, 2010 May.
Article in English | MEDLINE | ID: mdl-20447069

ABSTRACT

OBJECTIVE: The recent concept that oncogenes responsible for thyroid neoplastic transformation are able to elicit an inflammatory protumourigenic microenvironment raises interest in further studies on papillary thyroid cancer (PTC) associated with thyroid autoimmunity. PATIENTS: The clinical and molecular features, and the expression of inflammation-related genes, were investigated in a large series of PTCs with and without associated thyroiditis (groups A, n = 128 and B, n = 215). RESULTS: The two groups did not show significant differences in clinical and prognostic features, whereas they harboured a significantly different genetic background (P = 0.001), with RET/PTC1 being more represented in PTCs associated with autoimmunity, and BRAF(V600E) in patients with PTC alone. A RET/PTC rearrangement was also found in 41% of non-neoplastic thyroiditis tissues, contralateral to tumours harbouring either RET/PTC or BRAF mutations. The expression of genes encoding CCL20, CXCL8 and l-selectin was significantly higher in PTC specimens (either with RET/PTC, BRAF(V600E) or unknown genetic lesion) compared with normal thyroid samples. On the contrary, thyroiditis showed l-selectin expression levels even higher than PTCs, but CCL20 and CXCL8 levels comparable with normal tissues. CONCLUSIONS: The present data extend the knowledge about the tight relationships among oncogenes, thyroiditis and thyroid cancer. A different genetic background among PTCs with and without associated autoimmunity has been firstly demonstrated. The strong association between RET/PTC1 and thyroiditis points to a critical role of this oncoprotein in the modulation of the autoimmune response. Moreover, preliminary expression studies, indicating enhanced expression of inflammatory molecules in PTCs, suggest a proinflammatory, nonautoimmune relationship between thyroiditis and thyroid cancer.


Subject(s)
Autoimmunity , Carcinoma, Papillary/pathology , Inflammation/pathology , Thyroid Neoplasms/pathology , Adolescent , Adult , Aged , Carcinoma, Papillary/genetics , Chemokine CCL20/genetics , Female , Genetic Association Studies , Humans , Inflammation/genetics , Inflammation/immunology , Interleukin-8/genetics , Male , Middle Aged , Mutation , Oncogene Proteins, Fusion/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Neoplasms/genetics , Thyroid Neoplasms/immunology , Thyroiditis/genetics , Thyroiditis/immunology , Thyroiditis/pathology , Young Adult
7.
Neoplasia ; 11(1): 10-21, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19107227

ABSTRACT

Activation of the RET gene by chromosomal rearrangements generating RET/PTC oncogenes is a frequent, early, and causative event in papillary thyroid carcinoma (PTC). We have previously shown that, in human primary thyrocytes, RET/PTC1 induces a transcriptional program including the MET proto-oncogene. In PTCs, beta-catenin is frequently mislocated to the cytoplasm nucleus. We investigated the interplay between Ret/ptc1 signaling and Met in regulating the proinvasive phenotype and beta-catenin localization in cellular models of human PTC. Here, we show that Met protein is expressed and is constitutively active in human thyrocytes exogenously expressing RET/PTC1 as well as a mutant (Y451F) devoid of the main Ret/ptc1 multidocking site. Both in transformed thyrocytes and in the human PTC cell line TPC-1, Ret/ptc1-Y451-dependent signaling and Met cooperated to promote a proinvasive phenotype. Accordingly, gene/functional silencing of either RET/PTC1 or MET abrogated early branching morphogenesis in TPC-1 cells. The same effect was obtained by blocking the common downstream effector Akt. Y451 of Ret/ptc1 was required to promote proliferation and nuclear translocation of beta-catenin, suggesting that these oncogene-driven effects are Met-independent. Pharmacologic inhibition of Ret/ptc1 and Met tyrosine kinases by the multitarget small molecule RPI-1 blocked cell proliferation and invasive ability and dislocated beta-catenin from the nucleus. Altogether, these results support that Ret/ptc1 cross talks with Met at transcriptional and signaling levels and promotes beta-catenin transcriptional activity to drive thyrocyte neoplastic transformation. Such molecular network, promoting disease initiation and acquisition of a proinvasive phenotype, highlights new options to design multitarget therapeutic strategies for PTCs.


Subject(s)
Cell Nucleus/metabolism , Cell Transformation, Neoplastic/genetics , Proto-Oncogene Proteins c-ret/physiology , Proto-Oncogene Proteins/genetics , Receptors, Growth Factor/genetics , Thyroid Gland/pathology , beta Catenin/metabolism , Active Transport, Cell Nucleus , Animals , Cell Adhesion/genetics , Cell Proliferation , Cells, Cultured , Gene Expression Regulation, Neoplastic , Humans , Mice , NIH 3T3 Cells , Neoplasm Invasiveness , Phenotype , Proto-Oncogene Mas , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-met , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Receptors, Growth Factor/metabolism , Receptors, Growth Factor/physiology , Thyroid Gland/metabolism , Up-Regulation/physiology , beta Catenin/physiology
8.
Cancer Lett ; 267(2): 262-70, 2008 Aug 28.
Article in English | MEDLINE | ID: mdl-18502035

ABSTRACT

Inflammation has long been suspected to contribute to tumor growth. However, the concept that oncogenes, known for decades as responsible for cell neoplastic transformation, build up an inflammatory pro-tumorigenic microenvironment is emerging only in the last few years. The well known oncogenes RAS and MYC have been causally linked to tumor angiogenesis through different ways. Moreover, in thyroid tumors, where many of the genetic tumor-initiating events have been identified, the oncogenes driving tumorigenesis were proved able to induce an inflammatory program. This minireview will focus on growing evidence implicating the role of intrinsic, oncogene-driven pathways leading to pro-tumoral inflammation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Inflammation/complications , Neoplasms/etiology , Oncogenes , Animals , Humans , Inflammation/genetics , Inflammation/pathology , Mice , Neoplasms/blood supply , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics
9.
Proc Natl Acad Sci U S A ; 102(41): 14825-30, 2005 Oct 11.
Article in English | MEDLINE | ID: mdl-16203990

ABSTRACT

Rearrangements of the RET receptor tyrosine kinase gene generating RET/PTC oncogenes are specific to papillary thyroid carcinoma (PTC), the most frequent thyroid tumor. Here, we show that the RET/PTC1 oncogene, when exogenously expressed in primary normal human thyrocytes, induces the expression of a large set of genes involved in inflammation and tumor invasion, including those encoding chemokines (CCL2, CCL20, CXCL8, and CXCL12), chemokine receptors (CXCR4), cytokines (IL1B, CSF-1, GM-CSF, and G-CSF), matrix-degrading enzymes (metalloproteases and urokinase-type plasminogen activator and its receptor), and adhesion molecules (L-selectin). This effect is strictly dependent on the presence of the RET/PTC1 Tyr-451 (corresponding to RET Tyr-1062 multidocking site). Selected relevant genes (CCL20, CCL2, CXCL8, CXCR4, L-selectin, GM-CSF, IL1B, MMP9, UPA, and SPP1/OPN) were found up-regulated also in clinical samples of PTC, particularly those characterized by RET/PTC activation, local extrathyroid spread, and lymph node metastases, when compared with normal thyroid tissue or follicular thyroid carcinoma. These results, demonstrating that the RET/PTC1 oncogene activates a proinflammatory program, provide a direct link between a transforming human oncogene, inflammation, and malignant behavior.


Subject(s)
Gene Expression Regulation/genetics , Gene Rearrangement/genetics , Inflammation/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Gland/metabolism , Thyroid Neoplasms/metabolism , Blotting, Western , Cell Movement/physiology , Cells, Cultured , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Inflammation/genetics , L-Selectin/metabolism , Metalloproteases/metabolism , Microarray Analysis , Polymerase Chain Reaction , Proto-Oncogene Proteins c-ret/genetics , Receptors, Cytokine/metabolism , Thyroid Gland/cytology , Thyroid Neoplasms/genetics , Urokinase-Type Plasminogen Activator/metabolism
10.
Oncogene ; 23(44): 7297-309, 2004 Sep 23.
Article in English | MEDLINE | ID: mdl-15326489

ABSTRACT

The receptor tyrosine kinase RET is alternatively spliced to yield two main isoforms, RET9 and RET51, which differ in their carboxyl terminal. Activated RET induces different biological responses such as morphological transformation, neurite outgrowth, proliferation, cell migration and branching. The two isoforms have been suggested to have separate intracellular signaling pathways and different roles in mouse development. Here we show that both isoforms are able to induce cell scattering of SK-N-MC neuroepithelioma cell line and branching tubule formation in MDCK cell line. However, the Y1062F mutation, which abrogates the transforming activity of both activated RET isoforms in NIH3T3 cells, does not abolish scattering and branching morphogenesis of RET51, whereas impairs these biological effects of RET9. The GDNF-induced biological effects of RET51 are inhibited by the simultaneous abrogation of both Tyr1062 and Tyr1096 docking sites. Thus, Tyr1096 may substitute the functions of Tyr1062. GRB2 is the only known adaptor protein binding to Tyr1096. Dominant-negative GRB2 expressed in MDCK cells together with RET9 or RET51 significantly reduces branching. Therefore, GRB2 is necessary for RET-mediated branching of MDCK cells.


Subject(s)
Adaptor Proteins, Signal Transducing , Epithelial Cells/cytology , Neurons/cytology , Oncogene Proteins/metabolism , Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Tyrosine , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cell Division/drug effects , Cell Line , Cell Movement/physiology , Cloning, Molecular , DNA Primers , Dogs , Epithelial Cells/drug effects , Epithelial Cells/physiology , GRB2 Adaptor Protein , Glial Cell Line-Derived Neurotrophic Factor , Kidney , Mice , Mitogen-Activated Protein Kinases/metabolism , Nerve Growth Factors/pharmacology , Neurites/ultrastructure , Neurons/drug effects , Neurons/physiology , Oncogene Proteins/genetics , Protein Isoforms/metabolism , Proteins/genetics , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/genetics , Recombinant Fusion Proteins/metabolism , p38 Mitogen-Activated Protein Kinases
11.
Mol Endocrinol ; 18(4): 1004-17, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14715928

ABSTRACT

Gain-of-function mutations of ret receptor tyrosine kinase, the signaling receptor for glial cell line-derived neurotrophic factor, cause sporadic thyroid and adrenal malignancies as well as endocrine cancer syndromes, such as multiple endocrine neoplasia types 2A and 2B (MEN 2A and MEN 2B) and familial medullary thyroid carcinoma. Loss-of-function mutations of ret cause Hirschsprung's disease (HSCR) or colonic aganglionosis. In 20-30% of families with a mutation at residues 609, 611, 618, or 620 of RET, MEN 2A and familial medullary thyroid carcinoma cosegregate with HSCR. These mutations constitutively activate RET due to aberrant disulfide homodimerization and diminish the level of RET at the plasma membrane. It is not known how these mutations simultaneously lead to both gain- and loss-of-function RET-associated diseases. We provide an explanation for the dual phenotypic Janus mutation at Cys620 of RET. In Madin-Darby canine kidney (MDCK) cells, the Janus mutation impairs the glial cell line-derived neurotrophic factor-induced effects of RET on cell migration, differentiation, and survival but simultaneously promotes rapid cell proliferation.


Subject(s)
Hirschsprung Disease/genetics , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2b/genetics , Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Animals , Dimerization , Dogs , Glial Cell Line-Derived Neurotrophic Factor , Hirschsprung Disease/metabolism , Multiple Endocrine Neoplasia Type 2a/metabolism , Multiple Endocrine Neoplasia Type 2b/metabolism , Nerve Growth Factors/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/metabolism
13.
Biochem Biophys Res Commun ; 296(3): 515-22, 2002 Aug 23.
Article in English | MEDLINE | ID: mdl-12176011

ABSTRACT

The receptor tyrosine kinase RET, with a known role in embryonic development and in human pathologies, is alternatively spliced to yield at least two functional isoforms, which differ only in their carboxyl terminal. Enigma protein is a member of the PDZ-LIM family and is known to interact with the short isoform of RET/PTC2, a chimeric oncoprotein isolated from papillary thyroid carcinoma. Here, we show that Enigma also interacts in intact cells with the short isoform of RET-wt and of its pathologic mutants associated to MEN2 syndromes, RET-C634R and RET-M918T. In contrast, Enigma binds all the corresponding RET long isoforms very poorly and colocalizes with short but not long RET/PTC2 isoforms. The RET docking tyrosine for Enigma is the last but one before the divergence between the two isoforms and we demonstrated that short-isoform-specific amino acid residues +2 to +4 to this tyrosine are required for the interaction of RET/PTC2 with Enigma.


Subject(s)
Carrier Proteins/metabolism , Drosophila Proteins , Intracellular Signaling Peptides and Proteins , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Carrier Proteins/analysis , Cell Line , Cytoskeletal Proteins , LIM Domain Proteins , Mice , Molecular Sequence Data , Precipitin Tests , Protein Isoforms/analysis , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases/chemistry , Sequence Alignment
SELECTION OF CITATIONS
SEARCH DETAIL
...