Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Curr Pharm Des ; 24(18): 1966-1971, 2018.
Article in English | MEDLINE | ID: mdl-29766783

ABSTRACT

The wound healing and the barrier restoration of the gastrointestinal (GI) mucosa must be continuously ensured to allow homeostasis of the gastrointestinal tract and of all the surrounding tissues. Several lines of the evidence report a key role of innate immunity, and in particular of Pattern Recognition Receptors (PRRs), in controlling the homeostasis of GI tract by sensing commensal and pathogen bacteria, activating the immune response and regulating epithelial repair, thus guaranteeing the morphological and functional recovery of the injured tissue. We will discuss the role of a particular class of PRRs - the Formyl Peptide Receptors - in the homeostasis of GI mucosa. We here report the results of studies that strongly suggest the possibility that the activation of FPRs is crucial in the maintenance of homeostasis of the GI tract and provide indications of the potential clinical relevance of new treatment regimens involving FPR modulation for several GI disorders.


Subject(s)
Gastrointestinal Tract/metabolism , Receptors, Formyl Peptide/metabolism , Animals , Anti-Bacterial Agents/pharmacology , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/immunology , Humans , Receptors, Formyl Peptide/drug effects , Receptors, Formyl Peptide/immunology , Wound Healing/drug effects , Wound Healing/immunology
2.
Oncogene ; 34(40): 5175-86, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25619830

ABSTRACT

There is increasing evidence that mast cells (MCs) and their mediators are involved in the remodeling of the tumor microenvironment and promote tumor growth, angiogenesis and metastasis. We have found that an increased density of MCs in thyroid cancer (TC) correlates with enhanced invasiveness. However, the MC-derived factors responsible for this activity and the mechanisms by which they enhance TC invasiveness remain unidentified. Here, we report that MCs, when activated by TC cells, produce soluble factors that induce epithelial-to-mesenchymal transition (EMT) and stemness features of TC cells. We identified CXCL8/interleukin (IL)-8 as the main mediator contained in activated MC conditioned media (CM) capable of inducing both EMT and stemness of TC cells. Mechanistically, MC CM or exogenous IL-8 stimulated Akt phosphorylation and Slug expression in TC cells. The inhibition of the Akt pathway or depletion of the Slug transcription factor by RNA interference, reverted EMT and stemness responses. TC cells stably transfected with exogenous IL-8 underwent EMT, displayed increased stemness and enhanced tumorigenicity with respect to control cells. The analysis of TC surgical specimens by immunohistochemical analysis demonstrated a positive correlation between MC density (Tryptase(+) cells) and stemness features (OCT4 staining). Taken together, our data identify an MC-dependent IL-8-Akt-Slug pathway that sustains EMT/stemness of TC cells. The blockade of this circuit might be exploited for the therapy of advanced TC.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Mast Cells/metabolism , Signal Transduction/physiology , Thyroid Neoplasms/pathology , Animals , Cell Line , Female , Heterografts , Humans , Immunoblotting , Immunohistochemistry , Interleukin-8/metabolism , Mice , Mice, Nude , Neoplastic Stem Cells/cytology , Proto-Oncogene Proteins c-akt/metabolism , Real-Time Polymerase Chain Reaction , Snail Family Transcription Factors , Tissue Array Analysis , Transcription Factors/metabolism , Transfection
3.
Oncogene ; 34(29): 3826-38, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25263443

ABSTRACT

N-formyl peptide receptors (FPR1, FPR2 and FPR3) are involved in innate immunity, inflammation and cancer. FPR expression, initially described in immune cells, was later observed in non-hematopoietic cell populations and tissues. Several studies suggested a role for FPRs in the progression of various tumor histotypes, including gastric cancer (GC), for which a positive association with a specific FPR1 polymorphism has recently been described. We previously showed that FPRs are expressed on gastric epithelium and are required for wound repair and restitution of barrier integrity. Here we assess the role of FPRs in GC. We characterized the functions of FPRs in GC epithelial cells (MKN28, AGS and MKN45) cultured in vitro by assessing migration, proliferation, resistance to apoptosis and activation of the epithelial-to-mesenchymal transition. Activation of each FPR induced the epithelial-to-mesenchymal transition, proliferation, resistance to apoptosis and migration of GC cells in culture. Blocking compounds or RNA interference of each FPR reverted these effects. We also defined the in vivo tumorigenic potential of GC epithelial cells silenced for FPRs by xenograft experiments in immunocompromised mice. Interestingly, FPR1 silencing in GC cells (shFPR1) significantly enhanced xenograft growth with respect to shCTR, shFPR2 and shFPR3 xenografts, because of augmented vessel density and cell proliferation. Accordingly, HIF-1α and VEGF mRNA levels were higher in shFPR1 xenografts than in controls. Moreover, the in vitro production of proangiogenic factors in response to FPR2/3 agonists (WKYMVm, LL-37, uPA, uPAR84-95, AnxA1) or to other proinflammatory mediators (IL-1α) was higher in shFPR1 GC cells than in shCTR, shFPR2 and shFPR3 cells, suggesting that FPR1 functions as an inhibitor of CG angiogenesis. Thus, we propose that FPR1 stimulation may represent a novel therapeutic approach to counteract tumor angiogenesis.


Subject(s)
Neovascularization, Pathologic/genetics , Receptors, Formyl Peptide/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/genetics , Cells, Cultured , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Mice, Inbred C57BL , Mice, Nude , Mice, SCID , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neovascularization, Pathologic/metabolism , Oligopeptides/pharmacology , RNA Interference , Receptors, Formyl Peptide/agonists , Receptors, Formyl Peptide/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/blood supply , Stomach Neoplasms/metabolism , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays/methods
4.
Oncogene ; 29(47): 6203-15, 2010 Nov 25.
Article in English | MEDLINE | ID: mdl-20729915

ABSTRACT

In different human carcinoma types, mast cell infiltrate increases with respect to normal tissue and mast cell density correlates with a bad prognosis. To assess the role of mast cells in human thyroid cancer, we compared the density of tryptase-positive mast cells in 96 papillary thyroid carcinomas (PTCs) versus normal thyroid tissue from 14 healthy individuals. Mast cell density was higher in 95% of PTCs (n=91) than in control tissue. Mast cell infiltrate correlated with extrathyroidal extension (P=0.0005) of PTCs. We show that thyroid cancer cell-line-derived soluble factors induce mast cell activation and chemoattraction in vitro. Different mast cell lines (HMC-1 and LAD2) and primary human lung mast cells induced thyroid cancer cell invasive ability, survival and DNA synthesis in vitro. The latter effect was mainly mediated by three mast-cell-derived mediators: histamine, and chemokines CXCL1/GROα and CXCL10/IP10. We show that xenografts of thyroid carcinoma cells (8505-C) could recruit mast cells injected into the tail vein of mice. Co-injection of human mast cells accelerated the growth of thyroid cancer cell (8505-C) xenografts in athymic mice. This effect was mediated by increased tumor vascularization and proliferation, and was reverted by treating mice with sodium cromoglycate (Cromolyn), a specific mast cell inhibitor. In conclusion, our study data suggest that mast cells are recruited into thyroid carcinomas and promote proliferation, survival and invasive ability of cancer cells, thereby contributing to thyroid carcinoma growth and invasiveness.


Subject(s)
Mast Cells/physiology , Thyroid Neoplasms/pathology , Animals , Cell Count , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic , Humans , Male , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Transplantation , Prognosis , Thyroid Neoplasms/blood supply , Thyroid Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism
5.
Oncogene ; 28(7): 937-49, 2009 Feb 19.
Article in English | MEDLINE | ID: mdl-19060924

ABSTRACT

XB130 is a recently cloned 130 kDa-adaptor protein and Src kinase substrate, structurally similar to actin-filament-associated protein. Here we show that XB130 is predominantly expressed in the thyroid. Given that XB130 is a thyroid-specific tyrosine kinase substrate, we asked whether it is targeted by RET/PTC, a genetically rearranged, constitutively active, thyroid-specific tyrosine kinase that plays a pathogenic role in papillary thyroid cancer. RET/PTC induced robust tyrosine phosphorylation of XB130, which promoted its subsequent association with the p85alpha subunit of phosphatidylinositol 3-kinase (PI 3-kinase). We identified tyrosine 54 of XB130 as the major target of RET/PTC-mediated phosphorylation and a critical binding site for the SH2 domains of p85alpha. Importantly, downregulation of XB130 in TPC1 papillary thyroid cancer cells, harboring the RET/PTC1 kinase, strongly reduced Akt activity without altering ERK1/2 phosphorylation, and concomitantly inhibited cell-cycle progression and survival in suspension. In conclusion, XB130 is a novel substrate of the RET/PTC kinase that links RET/PTC signaling to PI 3-kinase activation, and thereby plays an important role in sustaining proliferation and survival of thyroid tumor cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Oncogene Proteins, Fusion/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Thyroid Neoplasms/metabolism , Adaptor Proteins, Signal Transducing/genetics , Apoptosis , Blotting, Northern , Bromodeoxyuridine , Carcinoma, Papillary/metabolism , Carcinoma, Papillary/pathology , Cell Proliferation , Flow Cytometry , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oncogene Proteins, Fusion/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/pharmacology , Thyroid Neoplasms/pathology , Tumor Cells, Cultured , Tyrosine/metabolism
6.
Br J Cancer ; 97(11): 1545-51, 2007 Dec 03.
Article in English | MEDLINE | ID: mdl-17987038

ABSTRACT

Laryngeal dysplasia is a common clinical concern. Despite major advancements, a significant number of patients with this condition progress to invasive squamous cell carcinoma. Osteopontin (OPN) is a secreted glycoprotein, whose expression is markedly elevated in several types of cancers. We explored OPN as a candidate biomarker for laryngeal dysplasia. To this aim, we examined OPN expression in 82 cases of dysplasia and in hyperplastic and normal tissue samples. OPN expression was elevated in all severe dysplasia samples, but not hyperplastic samples, with respect to matched normal mucosa. OPN expression levels correlated positively with degree of dysplasia (P=0.0094) and negatively with disease-free survival (P<0.0001). OPN expression was paralleled by cell surface reactivity for CD44v6, an OPN functional receptor. CD44v6 expression correlated negatively with disease-free survival, as well (P=0.0007). Taken as a whole, our finding identify OPN and CD44v6 as predictive markers of recurrence or aggressiveness in laryngeal intraepithelial neoplasia, and overall, point out an important signalling complex in the evolution of laryngeal dysplasia.


Subject(s)
Glycoproteins/analysis , Hyaluronan Receptors/analysis , Laryngeal Diseases/pathology , Osteopontin/analysis , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Disease-Free Survival , Female , Humans , Immunohistochemistry , Laryngeal Diseases/metabolism , Male , Middle Aged , Pharyngeal Neoplasms/metabolism , Pharyngeal Neoplasms/pathology , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Prognosis
7.
Oncogene ; 25(39): 5467-74, 2006 Aug 31.
Article in English | MEDLINE | ID: mdl-16785999

ABSTRACT

Ras oncogenes are frequently mutated in thyroid carcinomas. To verify the role played by N-ras in thyroid carcinogenesis, we generated transgenic mice in which a human N-ras(Gln61Lys) oncogene (Tg-N-ras) was expressed in the thyroid follicular cells. Tg-N-ras mice developed thyroid follicular neoplasms; 11% developed follicular adenomas and approximately 40% developed invasive follicular carcinomas, in some cases with a mixed papillary/follicular morphology. About 25% of the Tg-N-ras carcinomas displayed large, poorly differentiated areas, featuring vascular invasion and forming lung, bone or liver distant metastases. N-ras(Gln61Lys) expression in cultured PC Cl 3 thyrocytes induced thyroid-stimulating hormone-independent proliferation and genomic instability with micronuclei formation and centrosome amplification. These findings support the notion that mutated ras oncogenes could be able to drive the formation of thyroid tumors that can progress to poorly differentiated, metastatic carcinomas.


Subject(s)
Genes, ras , Thyroid Gland/pathology , Thyroid Neoplasms/genetics , Adenocarcinoma, Follicular/genetics , Adenocarcinoma, Follicular/pathology , Adenoma/genetics , Adenoma/pathology , Amino Acid Substitution , Animals , Cell Differentiation , Humans , Mice , Mice, Transgenic , Neoplasm Invasiveness , Thyroid Neoplasms/pathology
8.
Oncogene ; 25(30): 4235-40, 2006 Jul 13.
Article in English | MEDLINE | ID: mdl-16501605

ABSTRACT

Activating mutations of the BRAF gene are the most common genetic alterations in papillary thyroid carcinomas (PTCs) and the T1799A transversion, resulting in BRAFV600E, appeared virtually unique in this cancer type. Here, we report on the identification in a classic PTC of a novel BRAF mutation, namely a 1795GTT insertion, resulting in BRAFV599Ins, and describe its biochemical and molecular characterization. Kinase assays carried out on BRAFV599Ins and BRAFV600E revealed a three- to five-fold increase in the enzymatic activity of both mutants with respect to BRAFWT. Similarly, evaluation of BRAF-induced phosphorylation of MEK, MAPK and RSK revealed a significant MAPK cascade activation in cells expressing BRAFV599Ins or BRAFV600E, but not in cells expressing BRAFWT. Molecular dynamic simulations showed a destabilization of the inactive conformation of the enzyme in both BRAFV599Ins and BRAFV600E mutants, but not in BRAFWT. The analysis of the interaction energies inside the catalytic site allowed to demonstrate the presence of repulsive electrostatic forces acting on the activation loop and moving from inward to outward of the mutant enzymes. Finally, focus assays in NIH-3T3 cells confirmed a high transformation rate in the cells transfected either with BRAFV599Ins or BRAFV600E. In conclusion, this study demonstrated that BRAFV599Ins, as BRAFV600E, is a 'gain of function' mutation, characterized by a constitutive catalytic activation, which accounts for its causative role in the studied PTC.


Subject(s)
Carcinoma, Papillary/genetics , Mutagenesis, Insertional , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/genetics , Thyroid Neoplasms/genetics , Aged , Animals , Carcinoma, Papillary/enzymology , Carcinoma, Papillary/pathology , Cell Line , Cell Transformation, Neoplastic/chemistry , Cell Transformation, Neoplastic/genetics , Computer Simulation , Crystallography, X-Ray , Female , Humans , Mice , NIH 3T3 Cells , Thermodynamics , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/pathology , Transfection
9.
Cell Mol Life Sci ; 61(23): 2954-64, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15583857

ABSTRACT

RET is the receptor for glial-derived neurotrophic factor growth factors. It is a paradigm of a single gene that causes different types of human cancer when targeted by different genetic alterations. Like other receptor tyrosine kinases, once activated, RET recruits a variety of signaling molecules that mediate biological responses. Here we review data on the signaling pathways that lead to RET-mediated cell transformation and recent evidence that manipulation of RET holds promise for thyroid cancer treatment.


Subject(s)
Neoplasms/metabolism , Proto-Oncogene Proteins/physiology , Receptor Protein-Tyrosine Kinases/physiology , Carcinoma, Papillary/metabolism , Cell Transformation, Neoplastic , Humans , Ligands , Models, Biological , Models, Genetic , Phosphorylation , Protein Structure, Tertiary , Proto-Oncogene Proteins c-ret , Signal Transduction , Thyroid Neoplasms/metabolism
10.
J Clin Endocrinol Metab ; 89(10): 4810-6, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15472167

ABSTRACT

Gain-of-function RET mutations are responsible for multiple endocrine neoplasia syndromes (MEN) 2A and 2B and familial medullary thyroid carcinoma (FMTC), whereas loss-of-function mutations are found in Hirschsprung disease. We report a new RET point mutation [R694Q (CGG-->CAG)], serendipitously found in a 23-yr-old woman with hypothyroidism due to atrophic Hashimoto's thyroiditis and primary ovarian failure, without altered calcitonin secretion. Familial history and clinical and biochemical evaluation of first-degree relatives were negative for FMTC, MEN 2A and 2B, and Hirschsprung disease. Genetic analysis showed that the mutation was inherited from the mother, who was submitted 2 yr before to thyroidectomy for goitrous Hashimoto's thyroiditis. Histological revision and immunohistochemical studies documented normal C cell number and morphology. We cloned the mutation in an expression vector encoding a full-length RET protein. The construct was transiently expressed in 293T cells in parallel with a wild-type RET and a C634R MEN 2A-associated RET mutant. Proteins were harvested from transfected cells, and tyrosine phosphorylation levels were assayed. The mutation did not exert significant potentiating effects on RET kinase. A focus assay was also performed on NIH3T3 fibroblasts; the mutant did not exert significant transforming activity. In conclusion, a new RET mutation was found in two subjects without any evidence of MEN and FMTC. In keeping with clinical data, transfection studies confirmed lack of activating activity. This serendipitous discovery, apparently devoid of oncogenic potential, underscores the problems that may be encountered in genomic studies on RET.


Subject(s)
Germ-Line Mutation , Oncogene Proteins/genetics , Primary Ovarian Insufficiency/genetics , Receptor Protein-Tyrosine Kinases/genetics , Thyroiditis, Autoimmune/genetics , Adult , Family Health , Female , Humans , Male , Point Mutation , Primary Ovarian Insufficiency/complications , Primary Ovarian Insufficiency/pathology , Proto-Oncogene Proteins c-ret , Thyroid Gland/pathology , Thyroiditis, Autoimmune/complications , Thyroiditis, Autoimmune/pathology
12.
Oncogene ; 20(48): 6973-82, 2001 Oct 25.
Article in English | MEDLINE | ID: mdl-11704822

ABSTRACT

Thyroid papillary carcinomas are characterized by RET/PTC rearrangements that cause the tyrosine kinase domain of the RET receptor to fuse with N-terminal sequences encoded by heterologous genes. This results in the aberrant expression of a ligand-independent and constitutively active RET kinase. We analysed actin reorganization induced by the RET/PTC1 oncogene in PC Cl 3 rat thyroid epithelial cells. Differently from oncogenes Src, Ras and Raf, RET/PTC1 caused actin filaments to form prominent stress fibers. Moreover, stress fibers were identified in human thyroid papillary carcinoma cell lines harboring RET/PTC1 rearrangements but not in thyroid carcinoma cells negative for RET/PTC rearrangements. RET/MEN 2A, a constitutively active but unrearranged membrane-bound RET oncoprotein, did not induce stress fibers in PC Cl 3 cells. Induction of stress fibers by RET/PTC1 was restricted to thyroid cells; it did not occur in NIH3T3 fibroblasts or MCF7 mammary cells. RET/PTC1-mediated stress fiber formation depended on Rho but not Rac small GTPase activity. In addition, inhibition of Rho, but not of Rac, caused apoptosis of RET/PTC1-expressing thyroid cells. We conclude that Rho is implicated in the actin reorganization and cell survival mediated by the chimeric RET/PTC1 oncogene in thyroid epithelial cells, both phenotypes being cell type- and oncogene type-specific.


Subject(s)
Carcinoma, Papillary/pathology , Drosophila Proteins , Oncogene Proteins, Fusion/physiology , Signal Transduction/physiology , Stress Fibers/physiology , Thyroid Gland/cytology , Thyroid Neoplasms/pathology , rho GTP-Binding Proteins/physiology , 3T3 Cells , Actins/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis , Breast Neoplasms/pathology , Cell Line , Cell Line, Transformed , Cell Survival , DNA Replication , Dimerization , Female , Humans , MAP Kinase Signaling System , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2a/metabolism , Neoplasm Invasiveness , Neoplasm Proteins/metabolism , Organ Specificity , Phenotype , Protein-Tyrosine Kinases , Proto-Oncogene Proteins , Proto-Oncogene Proteins c-ret , Rats , Receptor Protein-Tyrosine Kinases , Recombinant Fusion Proteins/physiology , Transfection , Tumor Cells, Cultured
13.
Mol Cell Biol ; 21(13): 4177-87, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11390647

ABSTRACT

The receptor tyrosine kinase RET functions as the signal transducing receptor for the GDNF (for "glial cell-derived neurotrophic factors") family of ligands. Mutations in the RET gene were implicated in Hirschsprung disease (HSCR), multiple endocrine neoplasia type 2 (MEN 2), and thyroid carcinomas. In this report we demonstrate that the docking protein FRS2 is tyrosine phosphorylated by ligand-stimulated and by constitutively activated oncogenic forms of RET. Complex formation between RET and FRS2 is mediated by binding of the phosphotyrosine-binding domain of FRS2 to pY1062, a residue in RET that also functions as a binding site for Shc. However, overexpression of FRS2 but not Shc potentiates mitogen-activated protein (MAP) kinase activation by RET oncoproteins. We demonstrate that oncogenic RET-PTC proteins are associated with FRS2 constitutively, leading to tyrosine phosphorylation of FRS2, MAP kinase stimulation, and cell proliferation. However, loss-of-function HSCR-associated RET mutants exhibit impaired FRS2 binding and reduced MAP kinase activation. These experiments demonstrate that FRS2 couples both ligand-regulated and oncogenic forms of RET, with the MAP kinase signaling cascade as part of the response of RET under normal biological conditions and pathological conditions, such as MEN 2 and papillary thyroid carcinomas.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Drosophila Proteins , MAP Kinase Signaling System/physiology , Membrane Proteins/metabolism , Phosphoproteins/metabolism , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line , Genes, Reporter/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors , Hirschsprung Disease/genetics , Humans , Ligands , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mitogen-Activated Protein Kinases/metabolism , Multiple Endocrine Neoplasia Type 2a/genetics , Mutation , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphorylation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-ret , Receptor Protein-Tyrosine Kinases/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1
14.
Oncogene ; 20(2): 209-18, 2001 Jan 11.
Article in English | MEDLINE | ID: mdl-11313948

ABSTRACT

Tyrosine 1062 of Ret, which represents an intracytoplasmic docking site for multiple signaling molecules, is essential for Ret-mediated activation of phosphatidylinositol 3-Kinase (PI3-K). PI3-K, in turn, has been implicated in inducing cell survival and neoplastic transformation mediated by Ret. We have examined the mechanisms by which Ret stimulates PI3-K. Here we show that the Insulin Receptor Substrate-1 (IRS-1) is tyrosine phosphorylated and associated with the p85 regulatory subunit of PI3-K in response to Ret activation. IRS-1 coimmunoprecipitates with Ret and co-expression of IRS-1 results in the potentiation of Ret-mediated activation of Akt(PKB), a bona fide effector of PI3-K. The association with the PTB domain of IRS-1 depends on the phosphorylation of tyrosine 1062 of Ret. The deletion of asparagine 1059 (delN1059) and the substitution of leucine 1061 (L1061P), two Ret mutations identified in families affected by congenital megacolon (Hirschsprung's disease), impair the binding of IRS-1 to Ret as well as Ret-mediated Akt(PKB) stimulation. Finally, we show that Shc, which was previously identified as another ligand of Y1062 of Ret, competes with IRS-1 for the binding to Ret pY1062. All together, these findings suggest that IRS-1 is a component of the signaling pathway which leads to Ret-mediated PI3-K activation, a pathway which can be targeted by Hirschsprung-associated Ret mutations. The alternative binding of Shc and IRS-1 to Ret pY1062 can be a system to modulate the activation of different intracellular signaling pathways and to elicit different biological responses following Ret activation.


Subject(s)
Drosophila Proteins , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Amino Acid Motifs , Amino Acid Substitution , Animals , Binding Sites , Binding, Competitive , Insulin Receptor Substrate Proteins , Mice , Mutation , Phosphoproteins/genetics , Phosphorylation , Polypyrimidine Tract-Binding Protein , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-ret , RNA-Binding Proteins/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Ribonucleoproteins/metabolism
15.
Mol Cell Biol ; 21(7): 2485-95, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11259597

ABSTRACT

The high-mobility group I (HMGI) nonhistone chromosomal proteins HMGI(Y) and HMGI-C have been implicated in defining chromatin structure and in regulating the transcription of several genes. These proteins have been implicated in adipocyte homeostasis: a severe deficiency of fat tissue is found in mice with targeted disruption of the HMGI-C locus, and lipomagenesis in humans is frequently associated with somatic mutations of HMGI genes. The aim of this study was to examine the role of HMGI(Y) proteins in adipocytic cell growth and differentiation. First, we found that differentiation of the preadipocytic 3T3-L1 cell line caused early induction of HMGI(Y) gene expression. Suppression of HMGI(Y) expression by antisense technology dramatically increased the growth rate and impaired adipocytic differentiation in these cells. The process of adipogenic differentiation involves the interplay of several transcription factors, among which is the CCAAT/enhancer-binding protein (C/EBP) family of proteins. These factors are required for the transcriptional activation of adipocyte-specific genes. We also tested the hypothesis that HMGI(Y) might participate in transcriptional control of adipocyte-specific promoters. We found that HMGI(Y) proteins bind C/EBPbeta in vivo and in vitro. Furthermore, we show that HMGI(Y) strongly potentiates the capacity of C/EBPbeta to transactivate the leptin promoter, an adipose-specific promoter. Taken together, these results indicate that the HMGI(Y) proteins play a critical role in adipocytic cell growth and differentiation.


Subject(s)
Adipocytes/cytology , Adipocytes/physiology , High Mobility Group Proteins/physiology , Transcription Factors/physiology , 3T3 Cells , Animals , Cell Differentiation/physiology , Cell Division/physiology , HMGA1a Protein , Mice
16.
Cancer Res ; 61(4): 1426-31, 2001 Feb 15.
Article in English | MEDLINE | ID: mdl-11245446

ABSTRACT

Mutations of the Ret receptor tyrosine kinase are responsible for inheritance of multiple endocrine neoplasia (MEN2A and MEN2B) and familial medullary thyroid carcinoma syndromes. Although several familial medullary thyroid carcinoma and most MEN2A mutations involve substitutions of extracellular cysteine residues, in most MEN2B cases there is a methionine-to-threonine substitution at position 918 (M918T) of the Ret kinase domain. The mechanism by which the MEN2B mutation converts Ret into a potent oncogene is poorly understood. Both MEN2A and MEN2B oncoproteins exert constitutive activation of the kinase. However, the highly aggressive MEN2B phenotype is not supported by higher levels of Ret-MEN2B kinase activity compared with Ret-MEN2A. It has been proposed that Ret-MEN2B is more than just an activated Ret kinase and that the M918T mutation, by targeting the kinase domain of Ret, might alter Ret substrate specificity, thus affecting Ret autophosphorylation sites and the ability of Ret to phosphorylate intracellular substrates. We show that the Ret-MEN2B mutation causes specific potentiated phosphorylation of tyrosine 1062 (Y1062) compared with Ret-MEN2A. Phosphorylated Y1062 is part of a Ret multiple effector docking site that mediates recruitment of the Shc adapter and of phosphatidylinositol-3 kinase (PI3K). Accordingly, we show that Ret-MEN2B is more active than Ret-MEN2A in associating with She and in causing constitutive activation of the Ras/mitogen-activated protein kinase and PI3K/Akt cascades. We conclude that the MEN2B mutation specifically potentiates the ability of Ret to autophosphorylate Y1062 and consequently to couple to the Ras/mitogen-activated protein kinase and the PI3K/Akt pathways. The more efficient triggering of these pathways may account for the difference between MEN2A and MEN2B syndromes.


Subject(s)
Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport , Drosophila Proteins , Multiple Endocrine Neoplasia Type 2b/metabolism , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , 3T3 Cells , Animals , COS Cells , Enzyme Activation , MAP Kinase Signaling System/physiology , Mice , Mitogen-Activated Protein Kinases/metabolism , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2a/metabolism , Multiple Endocrine Neoplasia Type 2b/genetics , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proteins/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-ret , Rats , Receptor Protein-Tyrosine Kinases/genetics , Shc Signaling Adaptor Proteins , Src Homology 2 Domain-Containing, Transforming Protein 1 , Thyroid Neoplasms/genetics , Thyroid Neoplasms/metabolism , Tyrosine/metabolism , ras Proteins/metabolism
17.
J Clin Endocrinol Metab ; 85(10): 3898-907, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11061555

ABSTRACT

Point mutations of the RET receptor tyrosine kinase are responsible for the inheritance of multiple endocrine neoplasia (MEN) type 2 syndromes and are also present in a fraction of sporadic medullary thyroid carcinomas. Somatic rearrangements of the RET gene generating the chimeric RET/papillary thyroid carcinoma (PTC) oncogenes are the predominant molecular lesions associated with papillary carcinoma, the most frequent thyroid malignancy in humans. Oncogenic mutations cause constitutive activation of the kinase function of RET, which, in turn, results in the autophosphorylation of RET tyrosine residues critical for signaling. In vitro kinase assays previously revealed six putative RET autophosphorylation sites. The aim of the present study was to assess the phosphorylation of two such residues, tyrosines 1015 and 1062 (Y1015 and Y1062), in the in vivo signaling of RET and RET-derived oncogenes. Using phosphorylated RET-specific antibodies, we demonstrate that both Y1015 and Y1062 are rapidly phosphorylated upon ligand triggering of RET. Moreover, regardless of the nature of the underlying activating mutation, the concomitant phosphorylation of Y1015 and Y1062 is a common feature of the various oncogenic RET products (MEN2A, MEN2B, and PTC). This study shows that Ab-pY1062 is a useful tool with which to detect activated RET in human tumor cells and surgical samples. Finally, the microinjection of Ab-pY1062 antibodies into living cells demonstrates that Ret/PTC1 signaling is required to maintain the mitogenesis of a human carcinoma cell line expressing the Ret/PTC1 oncoprotein.


Subject(s)
Drosophila Proteins , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Antibodies, Blocking/pharmacology , COS Cells , DNA/biosynthesis , Fibroblasts , Humans , Immunohistochemistry , Microinjections , Molecular Sequence Data , Oncogene Proteins/genetics , Oncogenes/genetics , Phosphorylation , Point Mutation/physiology , 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/metabolism , Transfection , Tumor Cells, Cultured , Tyrosine/genetics
18.
Exp Cell Res ; 260(2): 257-67, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11035920

ABSTRACT

Rat thyroid differentiated cells (PC Cl 3) are an excellent model system with which to study the interaction between differentiation and cell transformation. We previously demonstrated that PC Cl 3 cells expressing the adenovirus E1A gene no longer depend on thyrotropin for growth and do not express thyroid differentiation markers. Here we show that an E1A mutant unable to bind the RB protein failed to transform the PC Cl 3 cells. Conversely, mutations in the E1A p300 interacting region did not affect its transforming ability. The pivotal role of RB family proteins in the thyroid cell transformation is supported by the thyrotropin independence induced by the E7 gene of human papilloma virus type 16, but not by a mutated form in the RB-binding region.


Subject(s)
Adenovirus E1A Proteins/genetics , Carrier Proteins , Cell Cycle Proteins , Cell Transformation, Neoplastic , DNA-Binding Proteins , Retinoblastoma Protein/metabolism , Animals , Cell Differentiation , Cell Division , Cell Line , E2F Transcription Factors , Flow Cytometry/methods , Humans , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomavirus E7 Proteins , Phenotype , Rats , Rats, Inbred F344 , Retinoblastoma-Binding Protein 1 , Thyroid Gland/cytology , Thyrotropin/metabolism , Transcription Factor DP1 , Transcription Factors/genetics , Transcription Factors/physiology , Transfection
19.
Cancer Res ; 60(14): 3727-31, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10919641

ABSTRACT

The RET tyrosine kinase is a functional receptor for neurotrophic ligands of the glial cell line-derived neurotrophic factor (GDNF) family. Loss of function of RET is associated with congenital megacolon or Hirschsprung's disease, whereas germ-line point mutations causing RET activation are responsible for multiple endocrine neoplasia type 2 (MEN2A, MEN2B, and familial medullary thyroid carcinoma) syndromes. Here we show that the expression of a constitutively active RET-MEN2A oncogene promotes survival of rat pheochromocytoma PC12 cells upon growth factor withdrawal. Moreover, we show that the RET-MEN2A-mediated survival depends on signals transduced by the phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) cascades. Thus, in PC12 cells, RET-MEN2A associates with the PI3K regulatory subunit p85 and promotes activation of Akt (also referred to as protein kinase B) in a PI3K-dependent fashion; in addition, RET-MEN2A promotes MAPK activation. PI3K recruitment and Akt activation as well as MAPK activation depend on RET-MEN2A tyrosine residue 1062. As a result, tyrosine 1062 of RET-MEN2A is essential for RET-MEN2A-mediated survival of PC12 cells cultured in growth factor-depleted media.


Subject(s)
Drosophila Proteins , MAP Kinase Signaling System , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Tyrosine/metabolism , Animals , Blotting, Western , Cell Survival , Chromones/pharmacology , Culture Media, Serum-Free , DNA Fragmentation , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Glial Cell Line-Derived Neurotrophic Factor Receptors , In Situ Nick-End Labeling , Ligands , Morpholines/pharmacology , Multiple Endocrine Neoplasia Type 2a/genetics , Multiple Endocrine Neoplasia Type 2a/metabolism , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Precipitin Tests , Protein Isoforms , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-ret , Rats , Receptor Protein-Tyrosine Kinases/chemistry , Signal Transduction , Transfection
20.
Int J Oncol ; 16(4): 689-94, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10717236

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) and neurturin (NTN) are related growth factors which exert trophic effects on several neuronal populations and developing kidney. GDNF-family ligands interact with membrane receptors designated GFRalphas which, in turn, mediate stimulation of the Ret receptor tyrosine kinase. Here we show that Ret, GFRalpha-1 (the GDNF receptor), and GFRalpha-2 (the NTN receptor) are expressed by testicular germ cells, while GDNF and NTN are expressed by Sertoli cells. Both GDNF and NTN stimulate DNA synthesis in spermatogonia. Furthermore, Ret, ligands and co-receptors are expressed in germ cell tumors. Thus, GDNF-family ligands may act as paracrine factors in spermatogenesis and this circuit may be active in germ cell tumors.


Subject(s)
DNA/biosynthesis , Drosophila Proteins , Nerve Growth Factors/physiology , Nerve Tissue Proteins/physiology , Proto-Oncogene Proteins/analysis , Receptor Protein-Tyrosine Kinases/analysis , Spermatogonia/metabolism , Animals , Germinoma/metabolism , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors , Humans , Immunohistochemistry , Male , Mice , Nerve Growth Factors/analysis , Nerve Tissue Proteins/analysis , Neurturin , Proto-Oncogene Proteins c-ret , Testicular Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...