Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Orphanet J Rare Dis ; 10: 25, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25887804

ABSTRACT

BACKGROUND: Hereditary medullary thyroid carcinoma (MTC) is caused by germ-line gain of function mutations in the RET proto-oncogene, and a phenotypic variability among carriers of the same mutation has been reported. We recently observed this phenomenon in a large familial MTC (FMTC) family carrying the RET-S891A mutation. Among genetic modifiers affecting RET-driven MTC, a role has been hypothesized for RET-G691S non-synonymous polymorphism, though the issue remains controversial. Aim of this study was to define the in vitro contribution of RET-G691S to the oncogenic potential of the RET-S891A, previously shown to harbour low transforming activity. METHODS: The RET-S891A and RET-G691S/S891A mutants were generated by site-directed mutagenesis, transiently transfected in HEK293T cells and stably expressed in NIH3T3 cells. Their oncogenic potential was defined by assessing the migration ability by wound healing assay and the anchorage-independent growth by soft agar assay in NIH3T3 cells stably expressing either the single or the double mutants. Two RET-S891A families were characterised for the presence of RET-G691S. RESULTS: The functional studies demonstrated that RET-G691S/S891A double mutant displays a higher oncogenic potential than RET-S891A single mutant, assessed by focus formation and migration ability. Moreover, among the 25 RET-S891A carriers, a trend towards an earlier age of diagnosis was found in the MTC patients harboring RET-S891A in association with RET-G691S. CONCLUSIONS: We demonstrate that the RET-G691S non-synonymous polymorphism enhances in vitro the oncogenic activity of RET-S891A. Moreover, an effect on the phenotype was observed in the RET-G691S/S891A patients, thus suggesting that the analysis of this polymorphism could contribute to the decision on the more appropriate clinical and follow-up management.


Subject(s)
Carcinoma, Medullary/congenital , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2a/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Animals , Carcinoma, Medullary/genetics , Carcinoma, Medullary/metabolism , Carcinoma, Medullary/pathology , Gene Expression Regulation/physiology , Genomics , HEK293 Cells , Humans , Mice , Multiple Endocrine Neoplasia Type 2a/pathology , Mutation , NIH 3T3 Cells , Penetrance , Polymorphism, Genetic , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret/genetics , Thyroid Neoplasms/pathology
2.
J Transl Med ; 9: 158, 2011 Sep 23.
Article in English | MEDLINE | ID: mdl-21943129

ABSTRACT

BACKGROUND: Gastrointestinal stromal tumors (GISTs) are the most frequent mesenchymal tumors to develop in the digestive tract. These tumors are highly resistant to conventional chemotherapy and only the introduction of imatinib mesylate has improved the prognosis of patients. However, Response Evaluation Criteria in Solid Tumors are inappropriate for assessing tumor response, and the histological/pathological response to imatinib is variable, heterogeneous, and does not associate with clinical response. The effects of imatinib on responding GISTs are still being explored, and few studies correlate the clinical response with the histological response after pharmacological treatment. Recently, apoptosis and autophagy were suggested as possible alternative mechanisms of pharmacological response. METHODS: Here, we used a proteomic approach, combined with other analyses, to identify some molecular stromal components related to the response/behavior of resected, high-risk GISTs after neoadiuvant imatinib therapy. RESULTS: Our proteomic results indicate an elevated concentration of Stem Cell Growth Factor (SCGF), a hematopoietic growth factor having a role in the development of erythroid and myeloid progenitors, in imatinib-responsive tumor areas. SCGFα expression was detected by mass spectrometry, immunohistochemistry and/or western blot and attributed to acellular matrix of areas scored negative for KIT (CD117). RT-PCR results indicated that GIST samples did not express SCGF transcripts. The recently reported demonstration by Gundacker et al. 1 of the secretion of SCGF in mature pro-inflammatory dendritic cells would indicate a potential importance of SCGF in tissue inflammatory response. Accordingly, inflammatory infiltrates were detected in imatinib-affected areas and the CD68-positivity of the SCGF-positive and KIT-negative areas suggested previous infiltration of monocytes/macrophages into these regions. Thus, chronic inflammation subsequent to imatinib treatment may determine monocyte/macrophage recruitment in imatinib-damaged areas; these areas also feature prominent tumor-cell loss that is replaced by dense hyalinization and fibrosis. CONCLUSIONS: Our studies highlight a possible role of SCGFα in imatinib-induced changes of GIST structure, consistent with a therapeutic response.


Subject(s)
Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/metabolism , Hematopoietic Cell Growth Factors/metabolism , Lectins, C-Type/metabolism , Piperazines/therapeutic use , Proteomics/methods , Pyrimidines/therapeutic use , Benzamides , Blotting, Western , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Gene Expression Regulation, Neoplastic , Glycosylation , Hematopoietic Cell Growth Factors/genetics , Humans , Imatinib Mesylate , Immunohistochemistry , Lectins, C-Type/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stromal Cells/metabolism , Stromal Cells/pathology
3.
Biochem Pharmacol ; 82(7): 778-88, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21741956

ABSTRACT

Emerging evidence suggests that Ret oncoproteins expressed in medullary thyroid cancer (MTC) might evade the pro-apoptotic function of the dependence receptor proto-Ret by directly impacting the apoptosis machinery. Identification of the molecular determinants of the interplay between Ret signaling and apoptosis might provide a relevant contribution to the optimization of Ret-targeted therapies. Here, we describe the cross-talk between Ret-M918T oncogenic mutant responsible for type 2B multiple endocrine syndrome (MEN2B), and components of death receptor-mediated extrinsic apoptosis pathway. In the human MEN2B-type MTC cell line MZ-CRC-1 expressing Ret-M918T, Ret was found associated with Fap-1, known as inhibitor of the CD95 death receptor trafficking to the cell membrane, and with procaspase-8, the initiator pro-form caspase in the extrinsic apoptosis pathway. Cell treatment with the anti-tumor Ret kinase inhibitor RPI-1 inhibited tyrosine phosphorylation of procaspase-8, likely inducing its local activation, followed by downregulation of both Ret and Fap-1, and translocation of CD95 into lipid rafts. According to the resulting increase of CD95 cell surface expression, the CD95 agonist antibody CH11 enhanced RPI-1-induced cell growth inhibition and apoptosis. RET RNA interference downregulated Fap-1 protein in MZ-CRC-1 cells, whereas exogenous RET-M918T upregulated Fap-1 in HEK293 cells. Overall, these data indicate that the Ret oncoprotein exerts opposing controls on Fap-1 and CD95, increasing Fap-1 expression and decreasing CD95 cell surface expression. The functional interplay of the Ret mutant with the extrinsic apoptosis pathway provides a mechanism possibly contributing to MTC malignant phenotype and a rational basis for novel therapeutic strategies combining Ret inhibitors and CD95 agonists.


Subject(s)
Apoptosis , Carcinoma, Medullary/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 13/physiology , Proto-Oncogene Proteins c-ret/physiology , Thyroid Neoplasms/pathology , fas Receptor/physiology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Medullary/metabolism , Carcinoma, Neuroendocrine , Caspase 8/metabolism , Cell Line, Tumor , Enzyme Activation , Female , Gene Expression Regulation , Humans , Indoles/pharmacology , Mice , Mice, SCID , NIH 3T3 Cells , Neoplasm Transplantation , Phosphorylation , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Thyroid Neoplasms/metabolism , Transplantation, Heterologous , Tumor Burden
4.
J Proteome Res ; 10(9): 4196-207, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21751813

ABSTRACT

In principle, targeted therapies have optimal activity against a specific subset of tumors that depend upon the targeted molecule or pathway for growth, survival, or metastasis. Consequently, it is important in drug development and clinical practice to have predictive biomarkers that can reliably identify patients who will benefit from a given therapy. We analyzed tumor cell-line secretomes (conditioned cell media) to look for predictive biomarkers; secretomes represent a potential source for potential biomarkers that are expressed in intracellular signaling and therefore may reflect changes induced by targeted therapy. Using Gene Ontology, we classified by function the secretome proteins of 12 tumor cell lines of different histotypes. Representations and hierarchical relationships among the functional groups differed among the cell lines. Using bioinformatics tools, we identified proteins involved in intracellular signaling pathways. For example, we found that secretome proteins related to TGF-beta signaling in thyroid cancer cells, such as vasorin, CD109, and ßIG-H3 (TGFBI), were sensitive to RPI-1 and dasatinib treatments, which have been previously demonstrated to be effective in blocking cell proliferation. The secretome may be a valuable source of potential biomarkers for detecting cancer and measuring the effectiveness of cancer therapies.


Subject(s)
Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Neoplasms/chemistry , Neoplasms/metabolism , Proteome/analysis , Proteome/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cluster Analysis , Computational Biology , Dasatinib , Databases, Protein , Humans , Intracellular Space , Models, Biological , Neoplasm Proteins/analysis , Neoplasm Proteins/chemistry , Neoplasms/drug therapy , Phosphotyrosine/analysis , Phosphotyrosine/chemistry , Pyrimidines/pharmacology , Signal Transduction , Thiazoles/pharmacology , Transforming Growth Factor beta
5.
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
6.
PLoS One ; 6(2): e17141, 2011 Feb 22.
Article in English | MEDLINE | ID: mdl-21364949

ABSTRACT

Activated leukocyte cell adhesion molecule (ALCAM, CD166) is expressed in various tissues, cancers, and cancer-initiating cells. Alterations in expression of ALCAM have been reported in several human tumors, and cell adhesion functions have been proposed to explain its association with cancer. Here we documented high levels of ALCAM expression in human thyroid tumors and cell lines. Through proteomic characterization of ALCAM expression in the human papillary thyroid carcinoma cell line TPC-1, we identified the presence of a full-length membrane-associated isoform in cell lysate and of soluble ALCAM isoforms in conditioned medium. This finding is consistent with proteolytically shed ALCAM ectodomains. Nonspecific agents, such as phorbol myristate acetate (PMA) or ionomycin, provoked increased ectodomain shedding. Epidermal growth factor receptor stimulation also enhanced ALCAM secretion through an ADAM17/TACE-dependent pathway. ADAM17/TACE was expressed in the TPC-1 cell line, and ADAM17/TACE silencing by specific small interfering RNAs reduced ALCAM shedding. In addition, the CGS27023A inhibitor of ADAM17/TACE function reduced ALCAM release in a dose-dependent manner and inhibited cell migration in a wound-healing assay. We also provide evidence for the existence of novel O-glycosylated forms and of a novel 60-kDa soluble form of ALCAM, which is particularly abundant following cell stimulation by PMA. ALCAM expression in papillary and medullary thyroid cancer specimens and in the surrounding non-tumoral component was studied by western blot and immunohistochemistry, with results demonstrating that tumor cells overexpress ALCAM. These findings strongly suggest the possibility that ALCAM may have an important role in thyroid tumor biology.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule/genetics , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma , Carcinoma, Papillary , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ionomycin/pharmacology , Ionophores/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport/drug effects , Proteomics , RNA, Small Interfering/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Thyroid Cancer, Papillary , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
7.
Neoplasia ; 13(12): 1132-42, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22241959

ABSTRACT

PLX4032/vemurafenib is a first-in-class small-molecule BRAF(V600E) inhibitor with clinical activity in patients with BRAF mutant melanoma. Nevertheless, drug resistance develops in treated patients, and strategies to overcome primary and acquired resistance are required. To explore the molecular mechanisms involved in primary resistance to PLX4032, we investigated its effects on cell proliferation and signaling in a panel of 27 genetically characterized patient-derived melanoma cell lines. Cell sensitivity to PLX4032 was dependent on BRAF(V600E) and independent from other gene alterations that commonly occur in melanoma such as PTEN loss, BRAF, and MITF gene amplification. Two cell lines lacking sensitivity to PLX4032 and harboring a different set of genetic alterations were studied as models of primary resistance. Treatment with the MEK inhibitor UO126 but not with PLX4032 inhibited cell growth and ERK activation. Resistance to PLX4032 was maintained after CRAF down-regulation by siRNA indicating alternative activation of MEK-ERK signaling. Genetic characterization by multiplex ligation-dependent probe amplification and analysis of phosphotyrosine signaling by MALDI-TOF mass spectrometry analysis revealed the activation of MET and SRC signaling, associated with the amplification of MET and of CTNNB1 and CCND1 genes, respectively. The combination of PLX4032 with drugs or siRNA targeting MET was effective in inhibiting cell growth and reducing cell invasion and migration in melanoma cells with MET amplification; similar effects were observed after targeting SRC in the other cell line, indicating a role for MET and SRC signaling in primary resistance to PLX4032. Our results support the development of classification of melanoma in molecular subtypes for more effective therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Melanoma/enzymology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Sulfonamides/pharmacology , src-Family Kinases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Melanoma/genetics , Mutation , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Signal Transduction/drug effects , Vemurafenib , src-Family Kinases/antagonists & inhibitors
8.
Mol Cancer ; 9: 278, 2010 Oct 18.
Article in English | MEDLINE | ID: mdl-20955590

ABSTRACT

BACKGROUND: TPC-1 is a papillary thyroid carcinoma (PTC)-derived cell line that spontaneously expresses the oncogene RET/PTC1. TPC-1 treated with the RET/PTC1 inhibitor RPI-1 displayed a cytostatic and reversible inhibition of cell proliferation and a strong activation of focal adhesion kinase (FAK). As dasatinib inhibition of Src results in reduction of FAK activation, we evaluated the effects of TPC-1 treatment with dasatinib in combination with RPI-1. RESULTS: Dasatinib (100 nM) strongly reduced TPC-1 proliferation and induced marked changes in TPC-1 morphology. Cells appeared smaller and more contracted, with decreased cell spreading, due to the inhibition of phosphorylation of important cytoskeletal proteins (p130CAS, Crk, and paxillin) by dasatinib. The combination of RPI-1 with dasatinib demonstrated enhanced effects on cell proliferation (more than 80% reduction) and on the phosphotyrosine protein profile. In particular, RPI-1 reduced the phosphorylation of RET, MET, DCDB2, CTND1, and PLCγ, while dasatinib acted on the phosphorylation of EGFR, EPHA2, and DOK1. Moreover, dasatinib completely abrogated the phosphorylation of FAK at all tyrosine sites (Y576, Y577, Y861, Y925) with the exception of the autoactivation site (Y397). Notably, the pharmacological treatments induced an overexpression of integrin ß1 (ITB1) that was correlated with a mild enhancement in phosphorylation of ERK1/2 and STAT3, known for their roles in prevention of apoptosis and in increase of proliferation and survival. A reduction in Akt, p38 and JNK1/2 activation was observed. CONCLUSIONS: All data demonstrate that the combination of the two drugs effectively reduced cell proliferation (by more than 80%), significantly decreased Tyr phosphorylation of almost all phosphorylable proteins, and altered the morphology of the cells, supporting high cytostatic effects. Following the combined treatment, cell survival pathways appeared to be mediated by STAT3 and ERK activities resulting from integrin clustering and FAK autophosphorylation. EphA2 may also contribute, at least in part, to integrin and FAK activation. In conclusion, these data implicate ITB1 and EphA2 as promising therapeutic targets in PTC.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Indoles/antagonists & inhibitors , Indoles/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-ret/metabolism , Pyrimidines/pharmacology , Thiazoles/pharmacology , Thyroid Neoplasms/metabolism , Cell Line, Tumor , Dasatinib , Humans , Integrin beta1/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor, EphA2/metabolism , STAT3 Transcription Factor/metabolism , Thyroid Neoplasms/drug therapy , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Cancer ; 116(5): 1326-35, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20087959

ABSTRACT

BACKGROUND: The authors investigated whether early stage lung cancer could be identified by proteomic analyses of plasma. METHODS: For the first case-control study, plasma samples from 52 patients with lung cancer and from a group of 51 controls were analyzed by surface-enhanced laser desorption/ionization time-of-flight mass spectrometry. In a second case-control study, a classifier of 4 markers (mass-to-charge ratio, 11,681, 6843, 5607, and 8762) also was tested for validation on plasma from 16 consecutive patients with screen-detected cancer versus 406 healthy individuals. The most relevant marker was identified, and an enzyme-linked immunosorbent assay-based analysis revealed that signal intensity was correlated with concentration. RESULTS: The classifier had a sensitivity of 94.23% and a specificity of 76.47% in the first study but lost predictive value in the second study. Nevertheless, the 11,681 cluster, which was identified as serum amyloid protein A (SAA), resulted in a multiple logistic regression model that indicated a strong association with lung cancer. When both studies were considered as a together, the odds ratio (OR) for an SAA intensity > or =0.5 was 10.27 (95% confidence interval [CI], 4.64-22.74), whereas an analysis restricted to stage I cancers (TNM classification) revealed an OR of 8.45 (95% CI, 2.76-25.83) for T1 lung cancer and 21.22 (95% CI, 5.62-80.14) for T2 lung cancer. CONCLUSIONS: SAA levels were predictive of an elevated risk of lung cancer, supporting the general view that inflammation is implicated in lung cancer development.


Subject(s)
Biomarkers, Tumor/blood , Lung Neoplasms/blood , Serum Amyloid A Protein/analysis , Aged , C-Reactive Protein/analysis , Case-Control Studies , Female , Humans , Inflammation/complications , Male , Middle Aged , Risk , Sensitivity and Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
10.
Cancer Res ; 66(20): 10179-87, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17047083

ABSTRACT

Constitutive activation of the RET receptor tyrosine kinase underlies the genesis and progression of multiple endocrine neoplasia type 2 (MEN 2), a dominantly inherited cancer predisposition. Importantly, although kinase activation represents a common theme in neoplasias, not all activating mutations are functionally equivalent. Consistent with this, we ascertained a patient with classical features of MEN 2B, but lacking either of the classical mutations in RET (M918T or A883F). Instead, the patient harbors a novel pair of germ line missense mutations in cis at codons 804 and 805. We evaluated the potential physiochemical effects of these substitutions in silico, predicting both to be moderately deleterious in isolation, but severely deleterious in combination. Consistent with this postulate, we show that the identified tandem mutations (V804M/E805K) are biologically active, transforming cells in culture and that their transforming capacity in combination is distinctly synergistic. Furthermore, the V804M/E805K tandem lesion confers resistance to the small molecule receptor tyrosine kinase inhibitor, PP1, suggesting a mode of action distinct from that known for classical MEN 2B mutations. To address this question, we used homology molecular modeling in silico to model the active site of RET. We predict that RET804 constitutes a critical gatekeeper residue that, when mutated in combination with RET805, induces a conformational change in the hinge region that locks the active site in a position permissive for ATP hydrolysis. Our findings have implications both in the clinic and in the successful development of novel kinase-targeted anticancer drugs.


Subject(s)
Multiple Endocrine Neoplasia Type 2b/enzymology , Multiple Endocrine Neoplasia Type 2b/genetics , Mutation , Proto-Oncogene Proteins c-ret/genetics , Amino Acid Sequence , Base Sequence , Binding Sites , Cell Transformation, Neoplastic/genetics , Enzyme Activation , Female , Humans , Middle Aged , Models, Molecular , Molecular Sequence Data , Pedigree , Protein Conformation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-ret/chemistry , Proto-Oncogene Proteins c-ret/metabolism , Sequence Alignment , Sequence Homology, Amino Acid
11.
Am J Pathol ; 168(4): 1262-75, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16565500

ABSTRACT

In rare families RET tyrosine kinase receptor substitutions located in exon 10 (especially at positions 609, 618, and 620) can concomitantly cause the MEN 2A (multiple endocrine neoplasia type 2A) or FMTC (familial medullary thyroid carcinoma) cancer syndromes, and Hirschsprung's disease (HSCR). No animal model mimicking the co-existence of the MEN 2 pathology and HSCR is available, and the association of these activating mutations with a developmental defect still represents an unresolved problem. The aim of this work was to investigate the significance of the RET(C620R) substitution in the pathogenesis of both gain- and loss-of-function RET-associated diseases. We report the generation of a line of mice carrying the C620R mutation in the Ret gene. Although Ret(C620R) homozygotes display severe defects in kidney organogenesis and enteric nervous system development leading to perinatal lethality. Ret(C620R) heterozygotes recapitulate features characteristic of HSCR including hypoganglionosis of the gastrointestinal tract. Surprisingly, heterozygotes do not show any defects in the thyroid that might be attributable to a gain-of-function mutation. The Ret(C620R) allele is responsible for HSCR and affects the development of kidneys and the enteric nervous system (ENS). These mice represent an interesting model for studying new therapeutic approaches for the treatment of HSCR disease.


Subject(s)
Gastrointestinal Tract/embryology , Hirschsprung Disease/pathology , Kidney/embryology , Proto-Oncogene Proteins c-ret/genetics , Amino Acid Substitution , Animals , Cells, Cultured , Disease Models, Animal , Enteric Nervous System/abnormalities , Enteric Nervous System/embryology , Enteric Nervous System/metabolism , Female , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/metabolism , Hirschsprung Disease/embryology , Hirschsprung Disease/genetics , Kidney/pathology , Male , Mice , Mice, Mutant Strains , Proto-Oncogene Proteins c-ret/metabolism , Thyroid Gland/abnormalities , Thyroid Gland/embryology , Thyroid Gland/metabolism
12.
Mol Endocrinol ; 20(7): 1633-43, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16469774

ABSTRACT

We report the finding of a novel missense mutation at codon 833 in the tyrosine kinase of the RET proto-oncogene in a patient with a carcinoma of the thyroid. In vitro experiments demonstrate that the R833C mutation induces transformed foci only when present in the long 3' splice isoform and, in keeping with a model in which the receptor has to dimerize to be completely activated, glial cell line-derived neurotrophic factor stimulation leads the RET(R833C) receptor to a higher level of activation. Tyrosine kinase assays show that the RET(R833C) long isoform has weak intrinsic kinase activity and phosphorylation of an exogenous substrate is not elevated even in the presence of glial cell line-derived neurotrophic factor. Furthermore, the R833C mutation is capable of sustaining the transformed phenotype in vivo but does not confer upon the transformed cells the ability to degrade the basement membrane in a manner analogous to metastasis. Our functional characterization of the R833C substitution suggests that, like the V804M and S891A mutations, this tyrosine kinase mutation confers a weak activating potential upon RET. This is the first report demonstrating that the introduction of an intracellular cysteine can activate RET. However, this does not occur via dimerization in a manner analogous to the extracellular cysteine mutants.


Subject(s)
Cell Transformation, Neoplastic/genetics , Mutation, Missense , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-ret/genetics , Animals , Cysteine/metabolism , Dimerization , Female , Gene Expression Regulation , Humans , Mice , Middle Aged , NIH 3T3 Cells , Neoplasm Invasiveness/genetics , Protein Structure, Tertiary/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret/chemistry , Up-Regulation
13.
Oncogene ; 23(44): 7436-40, 2004 Sep 23.
Article in English | MEDLINE | ID: mdl-15273715

ABSTRACT

Papillary thyroid carcinoma (PTC) is associated with RET and NTRK1 rearrangements and BRAF mutations. A series of 60 PTCs collected in a single center from Italian patients were histologically re-examined and subclassified as well differentiated or tall cell variant. The sample collection was analysed for the presence of all the reported PTC-associated genetic alterations through DNA or cDNA amplification, followed by automated sequencing. The analysis of exons 11 and 15 of BRAF gene revealed the T1796A (V599E) mutation in 32% of cases, and this alteration is significantly associated with PTC tall cell variant. Oncogenic rearrangements of RET and NTRK1 receptors were found in 33 and 5% of cases, respectively. No Ras mutations were detected. Overall, genetic alterations were detected in two-thirds of samples, and in no single case more than one mutational event was found simultaneously. Gene expression profiling of a subset of 31 tumors performed using cDNA microarray chips showed no strong differences in global gene expression among the different cases. However, a supervised analysis of the obtained data identified a subset of genes differentially expressed in tumors carrying BRAF mutation or RTK rearrangement.


Subject(s)
Gene Expression Profiling/methods , Mutation/genetics , Oncogene Proteins/genetics , Proto-Oncogene Proteins c-raf/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor, trkA/genetics , Thyroid Neoplasms/genetics , Adenocarcinoma, Papillary/genetics , Amino Acid Substitution , Gene Rearrangement , Genetic Variation , Humans , Italy , Mutation, Missense , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins c-ret
14.
Cancer Res ; 63(9): 2234-43, 2003 May 01.
Article in English | MEDLINE | ID: mdl-12727845

ABSTRACT

RET tyrosine kinase oncoproteins are potential targets for anticancer therapy. We show here that along with the inhibition of RET tyrosine phosphorylation, the pyrazolo-pyrimidine inhibitor PP1 induces RETMEN2A and RETMEN2B oncoprotein destruction. In fact, as a consequence of PP1 treatment, RET oncoproteins translocate from the outer limiting membrane to inner cellular compartments and are rapidly addressed to the degradative pathway. The cleavage of RET oncoproteins is associated with an impairment of RET mitogenic signaling pathways that causes a reversion of the oncogenic transformation and establishes a long-term cytostatic effect. By using specific inhibitors of both the proteosome and the lysosome, we assessed that PP1 targets RET oncoproteins to proteosomal, rather than lysosomal, degradation. In this context of studies, we interestingly demonstrated that RETMEN2A and RETMEN2B receptors are constitutively ubiquitinated and interact with the ubiquitin ligase c-Cbl. Moreover, PP1 does not modify these interactions, although it indeed causes RET dephosphorylation. Therefore, even if the degradative pathway stimulated by the inhibitor appears to be mediated by the proteosome, PP1 does not seem to enhance nor promote receptor ubiquitination. These observations lead us to favor two models for PP1-induced RET oncoprotein degradation: either PP1-mediated RET dephosphorylation per se targets the oncoproteins for destruction or alternatively, PP1 insertion in the RET ATP-binding pocket promotes a mechanism for fast stress-induced degradation. The use of PP1, which therefore acts as a degradation-inducing factor, may represent a promising new strategy to selectively target RET oncogenic products for destruction and holds promise for future medullary thyroid cancer therapy.


Subject(s)
Enzyme Inhibitors/pharmacology , Proto-Oncogene Proteins/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Animals , Humans , Mice , Multiple Endocrine Neoplasia Type 2a/metabolism , Multiple Endocrine Neoplasia Type 2b/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/metabolism , Ubiquitin/metabolism
15.
Endocrinology ; 144(3): 922-8, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12586769

ABSTRACT

The thyroid TRK oncogenes are generated by chromosomal rearrangements juxtaposing the neurotrophic tyrosine receptor kinase type 1 (NTRK1) tyrosine kinase domain to foreign activating sequences. TRK oncoproteins display a constitutive tyrosine kinase activity resulting in the capability to transform NIH3T3 cells. The TRK oncoproteins' signal transduction has been in part elucidated, and it involves several signal transducers activated by the NGF-stimulated NTRK1 receptor. In this paper, we investigate the role of FRS2 and FRS3, two related adapter proteins activated by fibroblast growth factor and NTRK1 receptors, in the signaling of the thyroid TRK-T1 and TRK-T3 oncogenes. By a combination of in vitro and in vivo assays, we demonstrate that both fibroblast growth factor receptor substrate (FRS)2 and FRS3 are recruited and activated by TRK-T1 and TRK-T3. Interaction studies using different TRK-T3 mutants indicate that FRS3 is recruited by the same tyrosine residue interacting with Shc and FRS2. Expression studies show different expression patterns of the FRS adapters in normal and tumor thyroid samples: FRS3 is expressed in both normal and thyroid tumor samples, whereas FRS2 is not expressed in normal thyroid but is differentially expressed in some tumors. Altogether, our data indicate that the FRS2 and FRS3 adapters may have a role in thyroid carcinogenesis triggered by TRK oncogenes.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , Lipoproteins/metabolism , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Receptor, trkA/physiology , Thyroid Gland/chemistry , 3T3 Cells , Animals , Blotting, Western , Carrier Proteins/genetics , Fibroblast Growth Factors/pharmacology , Gene Expression , Immunosorbent Techniques , Lipoproteins/genetics , Membrane Proteins/genetics , Mice , Mutagenesis, Site-Directed , Phosphoproteins/genetics , RNA, Messenger/analysis , Receptor, trkA/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Thyroid Neoplasms/genetics , Transfection
16.
Tumori ; 89(5): 550-2, 2003.
Article in English | MEDLINE | ID: mdl-14870784

ABSTRACT

Medullary thyroid carcinoma (MTC) responds very poorly to chemotherapy. Mutations in the RET gene are critical for MTC pathogenesis. RET therefore represents a rational target for the development of novel MTC therapies. The accumulation of evidence from laboratory studies strongly suggests that PP1 inhibitor is a cytostatic agent for cells expressing RET oncoproteins. PP1 functions as a potent and selective inhibitor of RET oncoprotein phosphorylation, promoting its proteasomal degradation.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Medullary/drug therapy , Multiple Endocrine Neoplasia Type 2a/metabolism , Multiple Endocrine Neoplasia Type 2b/metabolism , Oncogene Proteins/drug effects , Oncogene Proteins/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/drug effects , Receptor Protein-Tyrosine Kinases/metabolism , Thyroid Neoplasms/drug therapy , Carcinoma, Medullary/metabolism , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-ret , Thyroid Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...