Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Oncotarget ; 6(34): 36731-45, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26447612

ABSTRACT

Gastro-intestinal neuroendocrine tumors (GI-NETs) are rare neoplasms, frequently metastatic, raising difficult clinical and therapeutic challenges due to a poor knowledge of their biology. As neuroendocrine cells express both epithelial and neural cell markers, we studied the possible involvement in GI-NETs of axon guidance molecules, which have been shown to decrease tumor cell proliferation and metastatic dissemination in several tumor types. We focused on the role of Semaphorin 3F (SEMA3F) in ileal NETs, one of the most frequent subtypes of GI-NETs.SEMA3F expression was detected in normal neuroendocrine cells but was lost in most of human primary tumors and all their metastases. SEMA3F loss of expression was associated with promoter gene methylation. After increasing endogenous SEMA3F levels through stable transfection, enteroendocrine cell lines STC-1 and GluTag showed a reduced proliferation rate in vitro. In two different xenograft mouse models, SEMA3F-overexpressing cells exhibited a reduced ability to form tumors and a hampered liver dissemination potential in vivo. This resulted, at least in part, from the inhibition of mTOR and MAPK signaling pathways.This study demonstrates an anti-tumoral role of SEMA3F in ileal NETs. We thus suggest that SEMA3F and/or its cellular signaling pathway could represent a target for ileal NET therapy.


Subject(s)
Axon Guidance/physiology , Ileal Neoplasms/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neuroendocrine Tumors/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Disease Progression , Female , Heterografts , Humans , Ileal Neoplasms/genetics , Ileal Neoplasms/pathology , Membrane Proteins/genetics , Mice , Nerve Tissue Proteins/genetics , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Signal Transduction
2.
Neuroendocrinology ; 97(4): 331-40, 2013.
Article in English | MEDLINE | ID: mdl-23343749

ABSTRACT

BACKGROUND/AIMS: While the range of therapeutic options for well-differentiated gastroenteropancreatic neuroendocrine tumors has recently increased with the emergence of targeted therapies, such as mTOR inhibitors, there is no recent progress in the treatment of poorly differentiated neuroendocrine carcinomas (PDNECs). Since PDNECs have been shown to strongly express mTOR pathway components, the aim of the present study was to assess the antitumor effect of the mTOR inhibitor everolimus in preclinical models of PDNECs. METHODS: The expression of mTOR pathway components and their response to everolimus were assessed in two neuroendocrine cell lines: STC-1 and GluTag. A xenograft model of intrahepatic dissemination in the nude mouse, based on the intrasplenic injection of either STC-1 and GluTag tumor cells, was used. Animals were started on everolimus treatment 3 days after injection. The effects of treatment on tumor growth, proliferative capacities, apoptosis and in situ expression of mTOR pathway components were assessed. RESULTS: The expression of mTOR pathway components was comparable in STC-1 and GluTag cells and in human PDNECs and could be inhibited in vitro by everolimus. In vivo, the tumor volume of STC-1 and GluTag xenografts was significantly reduced in treated animals (6.05 ± 1.84% as compared to 21.76 ± 3.88% in controls). Everolimus treatment also induced a significant decrease in Ki67 index and in the phosphorylation levels of the two major effectors of mTOR, p70S6K and 4E-BP1. CONCLUSION: Our experimental data suggest that mTOR inhibition could be considered a therapeutic option for high-grade gastroenteropancreatic neuroendocrine tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Intestinal Neoplasms/drug therapy , Neuroendocrine Tumors/drug therapy , Pancreatic Neoplasms/drug therapy , Sirolimus/analogs & derivatives , Stomach Neoplasms/drug therapy , Adult , Aged , Animals , Cell Line, Tumor , Everolimus , Female , Humans , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Male , Mice , Mice, Nude , Middle Aged , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Signal Transduction/drug effects , Signal Transduction/physiology , Sirolimus/therapeutic use , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays
3.
Am J Pathol ; 178(1): 336-44, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21224070

ABSTRACT

The mammalian target of rapamycin (mTOR) inhibitors, such as rapalogues, are a promising new tool for the treatment of metastatic gastroenteropancreatic endocrine tumors. However, their mechanisms of action remain to be established. We used two murine intestinal endocrine tumoral cell lines, STC-1 and GLUTag, to evaluate the antitumor effects of rapamycin in vitro and in vivo in a preclinical model of liver endocrine metastases. In vitro, rapamycin inhibited the proliferation of cells in the basal state and after stimulation by insulin-like growth factor-1. Simultaneously, p70S6 kinase and 4EBP1 phosphorylation was inhibited. In vivo, rapamycin substantially inhibited the intrahepatic growth of STC-1 cells, irrespectively of the timing of its administration and even when the treatment was administered after cell intrahepatic engraftment. In addition, treated animals had significantly prolonged survival (mean survival time: 47.7 days in treated animals versus 31.8 days in controls) and better clinical status. Rapamycin treatment was associated with a significant decrease in mitotic index and in intratumoral vascular density within STC-1 tumors. Furthermore, the antitumoral effect obtained after treatment with a combination of rapamycin and phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 was more significant than with rapamycin alone in both cell lines. Our results suggest that the antitumor efficacy of rapamycin in neuroendocrine tumors results from a combination of antiproliferative and antiangiogenic effects. Interestingly, a more potent antitumor efficiency could be obtained by simultaneously targeting several levels of the PI3K/mTOR pathway.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Chromones/therapeutic use , Intestinal Neoplasms/drug therapy , Morpholines/therapeutic use , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/therapeutic use , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Intestinal Neoplasms/enzymology , Intestinal Neoplasms/pathology , Mice , Phosphorylation/drug effects
4.
Neuroendocrinology ; 91(3): 268-78, 2010.
Article in English | MEDLINE | ID: mdl-20389030

ABSTRACT

Gastroenteropancreatic (GEP) endocrine tumors are hypervascular tumors able to synthesize and secrete high amounts of VEGF. We aimed to study the regulation of VEGF production in GEP endocrine tumors and to test whether some of the drugs currently used in their treatment, such as somatostatin analogues and mTOR inhibitors, may interfere with VEGF secretion. We therefore analyzed the effects of the somatostatin analogue octreotide, the mTOR inhibitor rapamycin, the PI3K inhibitor LY294002, the MEK1 inhibitor PD98059 and the p38 inhibitor SB203850 on VEGF secretion, assessed by ELISA and Western blotting, in three murine endocrine cell lines, STC-1, INS-r3 and INS-r9. Octreotide and rapamycin induced a significant decrease in VEGF production by all three cell lines; LY294002 significantly inhibited VEGF production by STC-1 and INS-r3 only. We detected no effect of PD98059 whereas SB203850 significantly inhibited VEGF secretion in INS-r3 and INS-r9 cells only. By Western blotting analysis, we observed decreased intracellular levels of VEGF and HIF-1alpha under octreotide, rapamycin and LY294002. For rapamycin and LY294002, this effect was likely mediated by the inhibition of the mTOR/HIF-1/VEGF pathway. In addition to its well-known anti-secretory effects, octreotide may also act through the inhibition of the PI3K/Akt pathway, as suggested by the decrease in Akt phosphorylation detected in all three cell lines. In conclusion, our study points out to the complex regulation of VEGF synthesis and secretion in neoplastic GEP endocrine cells and suggests that the inhibition of VEGF production by octreotide and rapamycin may contribute to their therapeutic effects.


Subject(s)
Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Neuroendocrine Tumors/enzymology , Neuroendocrine Tumors/metabolism , Octreotide/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Chromones/pharmacology , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Imidazoles/pharmacology , Insulin/metabolism , Insulin Secretion , MAP Kinase Kinase 1/antagonists & inhibitors , Mice , Morpholines/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyridines/pharmacology , Rats , Sirolimus/pharmacology , Somatostatin/drug effects , Somatostatin/metabolism , TOR Serine-Threonine Kinases , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
5.
Gastroenterology ; 135(5): 1698-1709.e2, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18775714

ABSTRACT

BACKGROUND & AIMS: Missense mutations account for 30% of mutations identified in patients with the multiple endocrine neoplasia type 1 (MEN1) syndrome. They raise several issues: the distinction between pathogenic mutations and polymorphisms is sometimes difficult and the functional effects of missense mutations are unclear. We aimed to evaluate the functional consequences of missense MEN1 mutations in an appropriate endocrine cellular context. METHODS: From the INS-1 insulinoma cell line, we established clones conditionally over expressing wild-type (WT) menin or its A160T, H317Y, and A541T variants. We compared the consequences of WT or variant menin over expression on apoptotic response after gamma-irradiation and analyzed the interactions of these proteins with p53. RESULTS: WT menin over expression sensitized INS-r3 cells to apoptosis through amplification of caspase-3 activation, increased p53 acetylation, and accelerated p21 activation; moreover, over expressed WT menin could be recovered in p53-containing complexes. For all 3 missense mutations tested, the functional effects observed with WT were impaired significantly and only low amounts of variant menin proteins were recovered in p53-containing complexes. CONCLUSIONS: Taking advantage of a new endocrine cellular model, we show a loss of function for 2 missense disease-related menin mutants and for a controversial variant as well. Furthermore, our results suggest the existence of functional interactions between p53 and menin for the control of apoptosis, which may cast new light on the mechanisms of endocrine tumorigenesis.


Subject(s)
Apoptosis/drug effects , DNA, Neoplasm/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Mutation, Missense , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Cell Count , Cell Proliferation , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic , Genes, p53/genetics , Humans , Immunoblotting , Multiple Endocrine Neoplasia Type 1/metabolism , Multiple Endocrine Neoplasia Type 1/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Polymerase Chain Reaction , Proto-Oncogene Proteins/metabolism , Tumor Cells, Cultured
6.
Mol Cell Endocrinol ; 291(1-2): 109-15, 2008 Sep 10.
Article in English | MEDLINE | ID: mdl-18590796

ABSTRACT

In a previous study, we demonstrated that the Men1 gene is mainly expressed in the proliferative crypt compartment of the small intestine and that a reduction of menin expression in the crypt-like IEC-17 cell line induces an increase in proliferation rate concomitant with an increase in cyclin D1 expression. The aim of the present study was to test the hypothesis that the NF-kappaB pathway may be involved in cyclin D1 overexpression. Transcriptional activity of the cyclin D1 gene promoter was increased upon reduction of menin expression. Blockade of the NF-kappaB pathway restored proliferation, cell cycle, cyclin D1 gene transcription and cyclin D1 expression levels to those observed in the presence of menin. These data support a correlation between cyclin D1 expression, NF-kappaB activity and menin expression in this epithelial cell line and are relevant to the physiological function of menin in regulating proliferation in the intestinal epithelium.


Subject(s)
Epithelial Cells/physiology , Intestinal Mucosa/cytology , Proto-Oncogene Proteins/metabolism , Transcription Factor RelA/metabolism , Transcription Factors/metabolism , Animals , Cell Cycle/physiology , Cell Line , Cyclin D1/genetics , Cyclin D1/metabolism , Duodenum/cytology , Duodenum/physiology , Epithelial Cells/cytology , Gene Expression Regulation , Humans , Promoter Regions, Genetic , Proto-Oncogene Proteins/genetics , Rats , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/genetics , Transcription Factors/genetics , Transcription, Genetic
7.
J Biol Chem ; 279(23): 24477-84, 2004 Jun 04.
Article in English | MEDLINE | ID: mdl-15054094

ABSTRACT

Menin, the product of the tumor suppressor gene MEN1, is widely expressed in mammalian endocrine and non-endocrine tissues, including intestine. Its known abundant expression in several types of cells with high proliferative capacity led us to investigate the physiological function of the protein menin in intestinal epithelium, one of the most rapidly growing epithelia. Here we showed that the Men1 gene is mainly expressed in the crypt compartment of the proximal small intestine and that its expression was increased during fasting in vivo, both suggesting a role of menin in the control of cell growth. Indeed, specific reduction of menin expression by transfected antisense cDNA in the rat duodenal crypt-like cell line, IEC-17, increased cell proliferation. The latter is correlated to a loss of cell-cycle arrest in G(1) phase by resting cells and an overexpression of cyclin D1 and cyclin-dependent kinase (Cdk)-4. Furthermore, these cells lost the inhibition of proliferation induced by transforming growth factor-beta1, associated with a decrease of transforming growth factor-beta type II receptor expression. As a result of deregulated proliferation, antisense menin transfected IEC-17 cells became tumorigenic as shown in vitro as well as in vivo in immunosuppressed animals. These results indicate that menin contributes to proliferation control in intestinal epithelial cells. The present study reveals an unknown physiological function for menin in intestine that may be important in the regulation of epithelial homeostasis.


Subject(s)
Epithelial Cells/metabolism , Intestines/cytology , Proto-Oncogene Proteins/biosynthesis , Agar/metabolism , Animals , Blotting, Western , Cell Cycle , Cell Division , Cell Line , Cell Separation , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , Cytoskeletal Proteins/metabolism , DNA, Complementary/metabolism , Down-Regulation , Fasting , Flow Cytometry , G1 Phase , Heterozygote , Immunohistochemistry , Immunosuppression Therapy , In Situ Hybridization , Intestine, Small/metabolism , Luciferases/metabolism , Mice , Mice, Inbred BALB C , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/pharmacology , Plasmids/metabolism , Protein Serine-Threonine Kinases , RNA, Messenger/metabolism , Rats , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Time Factors , Trans-Activators/metabolism , Transfection , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...