Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cancers (Basel) ; 13(6)2021 Mar 23.
Article de Anglais | MEDLINE | ID: mdl-33807122

RÉSUMÉ

OBJECTIVES: This study assessed the antitumoral activity of the MEK inhibitor trametinib (TMT212) and the ERK1/2 inhibitor SCH772984, alone and in combination with the CDK4/6 inhibitor ribociclib (LEE011) in human neuroendocrine tumor (NET) cell lines in vitro. METHODS: Human NET cell lines BON1, QGP-1, and NCI-H727 were treated with trametinib or SCH772984, alone and in combination with ribociclib, to assess cell proliferation, cell cycle distribution, and protein signaling using cell proliferation, flow cytometry, and Western blot assays, respectively. RESULTS: Trametinib and SCH772984, alone and in combination with ribociclib, significantly reduced NET cell viability and arrested NET cells at the G1 phase of the cell cycle in all three cell lines tested. In addition, trametinib also caused subG1 events and apoptotic PARP cleavage in QGP1 and NCI-H727 cells. A western blot analysis demonstrated the use of trametinib alone and trametinib in combination with ribociclib to decrease the expression of pERK, cMyc, Chk1, pChk2, pCDK1, CyclinD1, and c-myc in a time-dependent manner in NCI-H727 and QGP-1 cells. CONCLUSIONS: MEK and ERK inhibition causes antiproliferative effects in human NET cell lines in vitro. The combination of the MEK inhibitor trametinib (TMT212) with the CDK4/6 inhibitor ribociclib (LEE011) causes additive antiproliferative effects. Future preclinical and clinical studies of MEK inhibition in NETs should be performed.

2.
Endocrinology ; 160(11): 2600-2617, 2019 11 01.
Article de Anglais | MEDLINE | ID: mdl-31322702

RÉSUMÉ

There are no officially approved therapies for metastatic pheochromocytomas apart from ultratrace 131I-metaiodbenzylguanidine therapy, which is approved only in the United States. We have, therefore, investigated the antitumor potential of molecular-targeted approaches in murine pheochromocytoma cell lines [monocyte chemoattractant protein (MPC)/monocyte chemoattractant protein/3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT)], immortalized mouse chromaffin Sdhb-/- cells, three-dimensional pheochromocytoma tumor models (MPC/MTT spheroids), and human pheochromocytoma primary cultures. We identified the specific phosphatidylinositol-3-kinase α inhibitor BYL719 and the mammalian target of rapamycin inhibitor everolimus as the most effective combination in all models. Single treatment with clinically relevant doses of BYL719 and everolimus significantly decreased MPC/MTT and Sdhb-/- cell viability. A targeted combination of both inhibitors synergistically reduced MPC and Sdhb-/- cell viability and showed an additive effect on MTT cells. In MPC/MTT spheroids, treatment with clinically relevant doses of BYL719 alone or in combination with everolimus was highly effective, leading to a significant shrinkage or even a complete collapse of the spheroids. We confirmed the synergism of clinically relevant doses of BYL719 plus everolimus in human pheochromocytoma primary cultures of individual patient tumors with BYL719 attenuating everolimus-induced AKT activation. We have thus established a method to assess molecular-targeted therapies in human pheochromocytoma cultures and identified a highly effective combination therapy. Our data pave the way to customized combination therapy to target individual patient tumors.


Sujet(s)
Tumeurs de la surrénale/traitement médicamenteux , Antinéoplasiques/usage thérapeutique , Évérolimus/usage thérapeutique , Phéochromocytome/traitement médicamenteux , Thiazoles/usage thérapeutique , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Antinéoplasiques/pharmacologie , Protocoles de polychimiothérapie antinéoplasique , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Tests de criblage d'agents antitumoraux , Synergie des médicaments , Évérolimus/pharmacologie , Femelle , Humains , Mâle , Souris , Adulte d'âge moyen , Culture de cellules primaires , Transduction du signal/effets des médicaments et des substances chimiques , Thiazoles/pharmacologie
3.
J Nucl Med ; 60(9): 1240-1246, 2019 09.
Article de Anglais | MEDLINE | ID: mdl-30796167

RÉSUMÉ

Peptide receptor radionuclide therapy in advanced neuroendocrine tumors (NETs) demonstrates a limited objective response rate. The therapeutic efficacy might be further increased by peptide receptor chemoradionuclide therapy. In this preclinical study, we explored the effects of 5-fluorouracil plus the DNA methyltransferase inhibitor decitabine or the histone deacetylase inhibitor tacedinaline on NET cells in vitro. Methods: Human NET cell lines BON1 and QGP1 were treated with 5-fluorouracil alone or in combination with decitabine or tacedinaline, respectively. Radiosensitivity was tested in combination with γ-irradiation at doses of 0, 2, 4, or 6 Gy by colony formation assay. Somatostatin receptor type 2 (SSTR2) expression and 68Ga-DOTATOC uptake by human NET cell lines were investigated by Western blot analysis and by a radioligand binding assay. Results: Treatment with 5-fluorouracil alone or in combination with decitabine or tacedinaline reduced tumor cell viability and induced apoptosis, enhanced radiosensitivity in BON1 and QGP1 cells, induced SSTR2 expression, and resulted in increased radioligand binding of 68Ga-DOTATOC in NET cells. Conclusion: This preclinical study demonstrated that 5-fluorouracil alone or in combination with decitabine or tacedinaline caused radiosensitization of tumor cells, upregulation of SSTR2 expression in tumor cells, and increased radioligand binding of 68Ga-DOTATOC to these tumor cells. These preclinical in vitro findings indicate that 5-fluorouracil in combination with epigenetic modifiers might be a putative strategy to improve the treatment efficacy of peptide receptor chemoradionuclide therapy in NET.


Sujet(s)
Benzamides/administration et posologie , Épigenèse génétique , Fluorouracil/administration et posologie , Tumeurs neuroendocrines/imagerie diagnostique , Phénylènediamines/administration et posologie , Radiosensibilisants/administration et posologie , Récepteur somatostatine/métabolisme , Apoptose , Lignée cellulaire tumorale , Survie cellulaire , Décitabine/administration et posologie , Relation dose-effet des rayonnements , Cytométrie en flux , Histone/métabolisme , Humains , Microscopie de fluorescence , Octréotide/analogues et dérivés , Octréotide/métabolisme , Composés organométalliques/métabolisme , Protéine-1 liant le suppresseur de tumeur p53/métabolisme
4.
Endocr Relat Cancer ; 25(10): 893-908, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-29895527

RÉSUMÉ

Pancreatic neuroendocrine tumors (panNETs) are often inoperable at diagnosis. The mTORC1 inhibitor everolimus has been approved for the treatment of advanced NETs. However, the regular development of resistance to everolimus limits its clinical efficacy. We established two independent everolimus-resistant panNET (BON1) cell lines (BON1 RR1, BON1 RR2) to find potential mechanisms of resistance. After 24 weeks of permanent exposure to 10 nM everolimus, BON1 RR1 and BON1 RR2 showed stable resistance with cellular survival rates of 96.70% (IC50 = 5200 nM) and 92.30% (IC50 = 2500 nM), respectively. The control cell line showed sensitivity to 10 nM everolimus with cellular survival declining to 54.70% (IC50 = 34 nM). Both resistant cell lines did not regain sensitivity over time and showed persistent stable resistance after a drug holiday of 13 weeks. The mechanisms of resistance in our cell line model included morphological adaptations, G1 cell cycle arrest associated with reduced CDK1(cdc2) expression and decreased autophagy. Cellular migration potential was increased and indirectly linked to c-Met activation. GSK3 was over-activated in association with reduced baseline IRS-1 protein levels. Specific GSK3 inhibition strongly decreased BON1 RR1/RR2 cell survival. The combination of everolimus with the PI3Kα inhibitor BYL719 re-established everolimus sensitivity through GSK3 inhibition and restoration of autophagy. We suggest that GSK3 over-activation combined with decreased baseline IRS-1 protein levels and decreased autophagy may be a crucial feature of everolimus resistance, and hence, a possible therapeutic target.


Sujet(s)
Antinéoplasiques/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Évérolimus/pharmacologie , Glycogen Synthase Kinase 3/génétique , Inhibiteurs de protéines kinases/pharmacologie , Antinéoplasiques/usage thérapeutique , Autophagie/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Évérolimus/usage thérapeutique , Humains , Inhibiteurs de protéines kinases/usage thérapeutique
5.
Endocr Relat Cancer ; 25(5): 547-560, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29563190

RÉSUMÉ

Tropomyosin receptor kinase (Trk) inhibitors are investigated as a novel targeted therapy in various cancers. We investigated the in vitro effects of the pan-Trk inhibitor GNF-5837 in human neuroendocrine tumor (NET) cells. The human neuroendocrine pancreatic BON1, bronchopulmonary NCI-H727 and ileal GOT1 cell lines were treated with GNF-5837 alone and in combination with everolimus. Cell viability decreased in a time- and dose-dependent manner in GOT1 cells in response to GNF-5837 treatment, while treatment in BON1 and NCI-H727 cells showed no effect on cellular viability. Trk receptor expression determined GNF-5837 sensitivity. GNF-5837 caused downregulation of PI3K-Akt-mTOR signaling, Ras-Raf-MEK-ERK signaling, the cell cycle and increased apoptotic cell death. The combinational treatment of GNF-5837 with everolimus showed a significant enhancement in inhibition of cell viability vs single substance treatments, due to a cooperative PI3K-Akt-mTOR and Ras-Raf-MEK-ERK pathway downregulation, as well as an enhanced cell cycle component downregulation. Immunohistochemical staining for Trk receptors were performed using a tissue microarray containing 107 tumor samples of gastroenteropancreatic NETs. Immunohistochemical staining with TrkA receptor and pan-Trk receptor antibodies revealed a positive staining in pancreatic NETs in 24.2% (8/33) and 33.3% (11/33), respectively. We demonstrated that the pan-Trk inhibitor GNF-5837 has promising anti-tumoral properties in human NET cell lines expressing the TrkA receptor. Immunohistochemical or molecular screening for Trk expression particularly in pancreatic NETs might serve as predictive marker for molecular targeted therapy with Trk inhibitors.


Sujet(s)
Tumeurs neuroendocrines/traitement médicamenteux , Inhibiteurs de protéines kinases/usage thérapeutique , Récepteur trkA/antagonistes et inhibiteurs , Humains , Tumeurs neuroendocrines/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie
6.
Neuroendocrinology ; 106(1): 1-19, 2018.
Article de Anglais | MEDLINE | ID: mdl-27871087

RÉSUMÉ

BACKGROUND/AIMS: The tumor suppressor p53 is depleted in many tumor cells by the E3 ubiquitin ligase mouse double minute 2 homolog (MDM2) through MDM2/p53 interaction. A novel target for inhibiting p53 degradation and for causing reexpression of p53wild type is inhibition of MDM2. The small molecule NVP-CGM097 is a novel MDM2 inhibitor. We investigated MDM2 inhibition as a target in neuroendocrine tumor cells in vitro. METHODS: Human neuroendocrine tumor cell lines from the pancreas (BON1), lung (NCI-H727), and midgut (GOT1) were incubated with the MDM2 inhibitor NVP-CGM097 (Novartis) at concentrations from 4 to 2,500 nM. RESULTS: While p53wild type GOT1 cells were sensitive to NVP-CGM097, p53mutated BON1 and p53mutated NCI-H727 cells were resistant to NVP-CGM097. Incubation of GOT1 cells with NVP-CGM097 at 100, 500, and 2,500 nM for 96 h caused a significant decline in cell viability to 84.9 ± 9.2% (p < 0.05), 77.4 ± 6.6% (p < 0.01), and 47.7 ± 9.2% (p < 0.01). In a Western blot analysis of GOT1 cells, NVP-CGM097 caused a dose-dependent increase in the expression of p53 and p21 tumor suppressor proteins and a decrease in phospho-Rb and E2F1. Experiments of co-incubation of NVP-CGM097 with 5-fluorouracil, temozolomide, or everolimus each showed additive antiproliferative effects in GOT1 cells. NVP-CGM097 and 5-fluorouracil increased p53 and p21 expression in an additive manner. CONCLUSIONS: MDM2 inhibition seems a promising novel therapeutic target in neuroendocrine tumors harboring p53wild type. Further investigations should examine the potential role of MDM2 inhibitors in neuroendocrine tumor treatment.


Sujet(s)
Antinéoplasiques/pharmacologie , Fluorouracil/pharmacologie , Isoquinoléines/pharmacologie , Tumeurs neuroendocrines/traitement médicamenteux , Pipérazines/pharmacologie , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Relation dose-effet des médicaments , Résistance aux médicaments antinéoplasiques , Association de médicaments , Facteur de transcription E2F1/métabolisme , Humains , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/métabolisme , Protéines proto-oncogènes c-mdm2/métabolisme , Protéine du rétinoblastome/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
7.
Neuroendocrinology ; 106(4): 335-351, 2018.
Article de Anglais | MEDLINE | ID: mdl-28968593

RÉSUMÉ

INTRODUCTION: Glycogen synthase kinase 3α/ß (GSK3α/ß) is a serine/threonine kinase that plays a critical role in cancer. AIMS: In this study, we evaluated the effects of the specific GSK3α/ß inhibitor AR-A014418 in vitro to gain novel insights into GSK3α/ß signaling in neuroendocrine tumors (NETs). MATERIALS AND METHODS: Human NET cell lines (BON1, QGP1, H727, and GOT1) were treated with different concentrations of AR-A014418 alone and in combination with lovastatin, everolimus, 5-fluorouracil (5-FU), and γ-irradiation. RESULTS: AR-A014418 significantly dose- and time-dependently decreased cell viability in all 4 NET cell lines through inhibition of epithelial growth factor receptor and mTORC1/p70S6K signaling, as well as cyclin D3 downregulation and induction of pChk1. In all cell lines tested, FACS analysis showed an AR-A014418-induced increase in the sub-G1 phase, reflecting cell death. Apoptosis induction was observed in H727, GOT1 and QGP1 cells, but not in BON1 cells. Furthermore, significant antimigratory effects upon GSK3α/ß inhibition were found and were associated with ß-catenin downregulation in all cell lines tested. Compensatory upregulation of pAkt and pERK in response to GSK3α/ß inhibition was prevented by combining AR-A014418 with the ERK and Akt inhibitor lovastatin. Accordingly, the lovastatin/AR-A014418 combination was synergistic in BON1 and QGP1 cells. Moreover, AR-A014418 displayed promising chemosensitizing effects on 5-FU in QGP1 and slight radiosensitizing properties in BON1 and QGP1 cells. CONCLUSION: Our data provide new insights into the role of GSK3α/ß in NETs and suggest that GSK3α/ß inhibition could be a novel therapeutic option in NETs, especially in combination with lovastatin or 5-FU, depending on tumor entity.


Sujet(s)
Glycogen synthase kinase 3 beta/métabolisme , Tumeurs neuroendocrines/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Humains , Thiazoles/pharmacologie , Urée/analogues et dérivés , Urée/pharmacologie
8.
PLoS One ; 12(5): e0178375, 2017.
Article de Anglais | MEDLINE | ID: mdl-28542590

RÉSUMÉ

Modulation of the redox system in cancer cells has been considered a promising target for anti-cancer therapy. The novel MTH1 inhibitor TH588 proved tremendous potential in terms of cancer cell eradication, yet its specificity has been questioned by recent reports, indicating that TH588 may also induce cancer cell death by alternative mechanisms than MTH1 inhibition. Here we used a panel of heterogeneous neuroendocrine tumor cells in order to assess cellular mechanisms and molecular signaling pathways implicated in the effects of TH588 alone as well as dual-targeting approaches combining TH588 with everolimus, cytotoxic 5-fluorouracil or γ-irradiation. Our results reflect that TH588 alone efficiently decreased the survival of neuroendocrine cancer cells by PI3K-Akt-mTOR axis downregulation, increased apoptosis and oxidative stress. However, in the dual-targeting approaches cell survival was further decreased due to an even stronger downregulation of the PI3K-Akt-mTOR axis and augmentation of apoptosis but not oxidative stress. Furthermore, we could attribute TH588 chemo- and radio-sensitizing properties. Collectively our data not only provide insights into how TH588 exactly kills cancer cells but also depict novel perspectives for combinatorial treatment approaches encompassing TH588.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Enzymes de réparation de l'ADN/antagonistes et inhibiteurs , Évérolimus/pharmacologie , Fluorouracil/pharmacologie , Tumeurs neuroendocrines/traitement médicamenteux , Tumeurs neuroendocrines/radiothérapie , Phosphoric monoester hydrolases/antagonistes et inhibiteurs , Pyrimidines/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Apoptose/effets des radiations , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des radiations , Régulation négative/effets des médicaments et des substances chimiques , Régulation négative/effets des radiations , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des radiations , Rayons gamma/usage thérapeutique , Humains , Cellules neuroendocrines/effets des médicaments et des substances chimiques , Cellules neuroendocrines/effets des radiations , Tumeurs neuroendocrines/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques , Stress oxydatif/effets des radiations , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Radiothérapie adjuvante , Transduction du signal/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme
9.
Neuroendocrinology ; 103(3-4): 383-401, 2016.
Article de Anglais | MEDLINE | ID: mdl-26338447

RÉSUMÉ

BACKGROUND/AIMS: The hepatocyte growth factor/transmembrane tyrosine kinase receptor c-Met has been defined as a potential target in antitumoral treatment of various carcinomas. We aimed to investigate the direct effect of c-Met inhibition on neuroendocrine tumor cells in vitro. METHODS: The effects of the multi-tyrosine kinase inhibitors cabozantinib and tivantinib and of the highly specific c-Met inhibitor INC280 were investigated in human pancreatic neuroendocrine BON1, bronchopulmonary NCI-H727 and midgut GOT1 cells in vitro. RESULTS: INC280, cabozantinib and tivantinib inhibited c-Met phosphorylation, respectively. However, while equimolar concentrations (10 µM) of cabozantinib and tivantinib inhibited cell viability and cell migration, INC280 had no inhibitory effect. Knockdown experiments with c-Met siRNA also did not demonstrate effects on cell viability. Cabozantinib and tivantinib caused a G2 arrest in neuroendocrine tumor cells. CONCLUSIONS: Our in vitro data suggest that c-Met inhibition alone is not sufficient to exert direct antitumoral or antimigratory effects in neuroendocrine tumor cells. The multi-tyrosine kinase inhibitors cabozantinib and tivantinib show promising antitumoral and antimigratory effects in neuroendocrine tumor cells, which are most probably 'off-target' effects, not mediated by c-Met.


Sujet(s)
Anilides/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Pyrrolidones/pharmacologie , Quinoléines/pharmacologie , Benzamides , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Relation dose-effet des médicaments , Récepteurs ErbB/métabolisme , Cytométrie en flux , Humains , Imidazoles/pharmacologie , Mitogen-Activated Protein Kinase Kinases/métabolisme , Tumeurs neuroendocrines/anatomopathologie , Protéine oncogène v-akt/métabolisme , Phosphorylation/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs temps , Transfection , Triazines/pharmacologie
10.
PLoS One ; 10(12): e0143830, 2015.
Article de Anglais | MEDLINE | ID: mdl-26636335

RÉSUMÉ

BACKGROUND: The mTORC1-inhibitor everolimus shows limited efficacy in treating patients with gastro-entero-pancreatic or pulmonary neuroendocrine tumors (NETs), and poor outcome in patients with malignant pheochromocytoma or hepatic carcinoma. We speculated that any effect may be enhanced by antogonising other signaling pathways. METHODS: Therefore, we tested the effect of lovastatin--known to inhibit both ERK and AKT signaling--and everolimus, separately and in combination, on cell viability and signaling pathways in human midgut (GOT), pancreatic (BON1), and pulmonary (H727) NET, hepatocellular carcinoma (HepG2, Huh7), and mouse pheochromocytoma (MPC, MTT) cell lines. RESULTS: Lovastatin and everolimus separately significantly reduced cell viability in H727, HepG2, Huh7, MPC and MTT cells at clinically relevant doses (P ≤ 0.05). However, high doses of lovastatin were necessary to affect GOT or BON1 cell viability. Clinically relevant doses of both drugs showed additive anti-tumor effects in H727, HepG2, Huh7, MPC and MTT cells (P ≤ 0.05), but not in BON1 or GOT cells. In all cell lines investigated, lovastatin inhibited EGFR and AKT signaling. Subsequently, combination treatment more strongly inhibited EGFR and AKT signaling than everolimus alone, or at least attenuated everolimus-induced EGFR or AKT activation. Vice versa, everolimus constantly decreased pp70S6K and combination treatment more strongly decreased pp70S6K than lovastatin alone, or attenuated lovastatin-induced p70S6K activation: in BON1 cells lovastatin-induced EGFR inhibition was least pronounced, possibly explaining the low efficacy and consequent absent additive effect. CONCLUSION: In summary, clinically relevant doses of lovastatin and everolimus were effective separately and showed additive effects in 5 out of 7 cell lines. Our findings emphasize the importance of targeting several interacting signaling pathways simultaneously when attempting to attenuate tumor growth. However, the variable reactions of the different cell lines highlight the necessity to understand the unique molecular aberrations in any tumor. Nevertheless, this combination seems worthy of being tested in vivo.


Sujet(s)
Évérolimus , Lovastatine , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Tumeurs/traitement médicamenteux , Animaux , Synergie des médicaments , Récepteurs ErbB/génétique , Récepteurs ErbB/métabolisme , Évérolimus/agonistes , Évérolimus/pharmacologie , Cellules HepG2 , Humains , Lovastatine/agonistes , Lovastatine/pharmacologie , Souris , Souris knockout , Tumeurs/génétique , Tumeurs/métabolisme , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Ribosomal Protein S6 Kinases, 70-kDa/génétique , Ribosomal Protein S6 Kinases, 70-kDa/métabolisme
11.
World J Gastroenterol ; 20(29): 10038-49, 2014 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-25110431

RÉSUMÉ

AIM: To investigate the effect of aspirin on neuroendocrine tumor (NET) cell growth and signaling in vitro. METHODS: Human pancreatic BON1, bronchopulmonary NCI-H727 and midgut GOT1 neuroendocrine tumor cells were treated with different concentrations of aspirin (from 0.001 to 5 mmol/L), and the resulting effects on metabolic activity/cell proliferation were measured using cell proliferation assays and SYBR-DNA-labeling after 72, 144 and 216 h of incubation. The effects of aspirin on the expression and phosphorylation of several critical proteins that are involved in the most common intracellular growth factor signaling pathways (especially Akt protein kinase B) and mammalian target of rapamycin (mTOR) were determined by Western blot analyses. Propidium iodide staining and flow cytometry were used to evaluate changes in cell cycle distribution and apoptosis. Statistical analysis was performed using a 2-tailed Student's t-test to evaluate the proliferation assays and cell cycle analyses. The results are expressed as the mean ± SD of 3 or 4 independently performed experiments. Statistical significance was set at P < 0.05. RESULTS: Treatment with aspirin suppressed the viability/proliferation of BON1, NCI-H727 and GOT1 cells in a time- and dose-dependent manner. Significant effects were observed at starting doses of 0.5-1 mmol/L and peaked at 5 mmol/L. For instance, after treatment with 1 mmol/L aspirin for 144 h, the viability of pancreatic BON1 cells decreased to 66% ± 13% (P < 0.05), the viability of bronchopulmonary NCI-H727 cells decreased to 53% ± 8% (P < 0.01) and the viability of midgut GOT1 cells decreased to 89% ± 6% (P < 0.01). These effects were associated with a decreased entry into the S phase, the induction of the cyclin-dependent kinase inhibitor p21 and reduced expression of cyclin-dependent kinase 4 and cyclin D3. Aspirin suppressed mTOR downstream signaling, evidenced by the reduced phosphorylation of the mTOR substrates 4E binding protein 1, serine/threonine kinase P70S6K and S6 ribosomal protein and inhibited glycogen synthase kinase 3 activity. We observed the (compensatory) activation of tuberous sclerosis 2, the serine/threonine specific protein kinase AKT and extracellular signal-regulated kinases. CONCLUSION: Aspirin demonstrates promising anticancer properties for NETs in vitro. Further preclinical and clinical studies are needed.


Sujet(s)
Antinéoplasiques/pharmacologie , Acide acétylsalicylique/pharmacologie , Tumeurs du poumon/enzymologie , Tumeurs neuroendocrines/enzymologie , Tumeurs du pancréas/enzymologie , Inhibiteurs de protéines kinases/pharmacologie , Sérine-thréonine kinases TOR/antagonistes et inhibiteurs , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Humains , Tumeurs du poumon/anatomopathologie , Tumeurs neuroendocrines/anatomopathologie , Tumeurs du pancréas/anatomopathologie , Phosphorylation , Transduction du signal/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Facteurs temps
12.
Neuroendocrinology ; 98(2): 128-36, 2013.
Article de Anglais | MEDLINE | ID: mdl-23797089

RÉSUMÉ

The majority of neuroendocrine tumors (NETs) of the gastroenteropancreatic system coexpress somatostatin receptors (SSTRs) and dopamine type 2 receptors (D2R), thus providing a rationale for the use of novel SSTR2/D2R chimeric compounds in NET disease. Here we investigate the antitumor potential of the SSTR2/D2R chimeric compounds BIM-23A760 and BIM-23A758 in comparison to the selective SSTR2 agonist BIM-23023 and the selective D2R agonist BIM-53097 on human NET cell lines of heterogeneous origin. While having only minor effects on human pancreatic and bronchus carcinoid cells (BON1 and NCI-H727), BIM-23A758 induced significant antitumor effects in human midgut carcinoid cells (GOT1). These effects involved apoptosis induction as well as inhibition of mitogen-activated protein kinase and Akt signaling. Consistent with their antitumor response to BIM-23A758, GOT1 cells showed relatively high expression levels of SSTR2 and D2R mRNA. In particular, GOT1 cells highly express the short transcript variant of D2R. In contrast to BIM-23A758, the SSTR2/D2R chimeric compound BIM-23A760 as well as the individual SSTR2 and D2R agonistic compounds BIM-23023 and BIM-53097 induced no or only minor antitumor responses in the examined NET cell lines. Taken together, our findings suggest that the novel SSTR2/D2R chimeric compound BIM-23A758 might be a promising substance for the treatment of NETs highly expressing SSTR2 and D2R. In particular, a sufficient expression of the short transcript variant of DR2 might play a pivotal role for effective treatment.


Sujet(s)
Antinéoplasiques/pharmacologie , Tumeur carcinoïde/anatomopathologie , Dopamine/analogues et dérivés , Tumeurs de l'intestin/anatomopathologie , Protéines de fusion recombinantes/pharmacologie , Somatostatine/analogues et dérivés , Tumeur carcinoïde/génétique , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Dopamine/pharmacologie , Humains , Tumeurs de l'intestin/génétique , Récepteur D2 de la dopamine/agonistes , Récepteur D2 de la dopamine/génétique , Récepteur somatostatine/agonistes , Récepteur somatostatine/génétique , Somatostatine/pharmacologie , Transcriptome
13.
Endocr Relat Cancer ; 19(3): 423-34, 2012 Jun.
Article de Anglais | MEDLINE | ID: mdl-22499437

RÉSUMÉ

The majority of neuroendocrine tumors (NETs) of the gastroenteropancreatic system show aberrant Akt activity. Several inhibitors of the phosphoinositide 3-kinase (PI(3)K)-Akt-mTOR signaling pathway are currently being evaluated in clinical phase II and III studies for the treatment of NETs with promising results. However, the molecular mechanisms and particularly the role of different Akt isoforms in NET signaling are not fully understood. In this study, we examine the effect of Akt inhibition on NET cells of heterogeneous origin. We show that the Akt inhibitor perifosine effectively inhibits Akt phosphorylation and cell viability in human pancreatic (BON1), bronchus (NCI-H727), and midgut (GOT1) NET cells. Perifosine treatment suppressed the phosphorylation of Akt downstream targets such as GSK3α/ß, MDM2, and p70S6K and induced apoptosis. To further investigate the role of individual Akt isoforms for NET cell function, we specifically blocked Akt1, Akt2, and Akt3 via siRNA transfection. In contrast to Akt2 knockdown, knockdown of Akt isoforms 1 and 3 decreased phosphorylation levels of GSK3α/ß, MDM2, and p70S6K and suppressed NET cell viability and colony-forming capacity. The inhibitory effect of simultaneous downregulation of Akt1 and Akt3 on tumor cell viability was significantly stronger than that caused by downregulation of all Akt isoforms, suggesting a particular role for Akt1 and Akt3 in NET signaling. Akt3 siRNA-induced apoptosis while all three isoform-specific siRNAs impaired BON1 cell invasion. Together, our data demonstrate potent antitumor effects of the pan-Akt inhibitor perifosine on NET cells in vitro and suggest that selective targeting of Akt1 and/or Akt3 might improve the therapeutic potential of Akt inhibition in NET disease.


Sujet(s)
Tumeurs neuroendocrines/métabolisme , Phosphoryl-choline/analogues et dérivés , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Apoptose/effets des médicaments et des substances chimiques , Tumeurs des bronches/traitement médicamenteux , Tumeurs des bronches/métabolisme , Tumeurs des bronches/anatomopathologie , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Fragmentation de l'ADN , Humains , Tumeurs de l'intestin/traitement médicamenteux , Tumeurs de l'intestin/métabolisme , Tumeurs de l'intestin/anatomopathologie , Invasion tumorale , Tumeurs neuroendocrines/traitement médicamenteux , Tumeurs neuroendocrines/anatomopathologie , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Phosphoryl-choline/pharmacologie , Isoformes de protéines/antagonistes et inhibiteurs , Isoformes de protéines/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Petit ARN interférent/génétique
14.
Cancer Lett ; 295(1): 100-9, 2010 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-20356670

RÉSUMÉ

Several studies have established a link between aberrant PI(3)K-Akt-mTOR- and Ras-Raf-MEK-Erk1/2 signaling and neuroendocrine tumor disease. In this study, we comparatively investigate the antitumor potential of novel small-molecule inhibitors targeting mTOR (RAD001), mTOR/PI(3)K (NVP-BEZ235) and Raf (Raf265) on human NET cell lines of heterogeneous origin. All inhibitors induced potent antitumor effects which involved the induction of apoptosis and G0/G1 arrest. However, the dual mTOR/PI(3)K inhibitor NVP-BEZ235 was more efficient compared to the single mTOR inhibitor RAD001. Consistently, NVP-BEZ235 prevented the negative feedback activation of Akt as observed after treatment with RAD001. Raf265 inhibited Erk1/2 phosphorylation but strongly induced Akt phosphorylation and VEGF secretion, suggesting the existence of a compensatory feedback loop on PI3K-Akt signaling. Finally, combined treatment with RAD001 or NVP-BEZ235 and Raf265 was more efficient than single treatment with either kinase inhibitor. Together, our data provide a rationale for dual targeting of PI(3)K-Akt-mTOR- and Ras-Raf-MEK-Erk1/2 signaling in NET disease.


Sujet(s)
Antinéoplasiques/pharmacologie , Protéines et peptides de signalisation intracellulaire/antagonistes et inhibiteurs , Tumeurs neuroendocrines/traitement médicamenteux , Tumeurs neuroendocrines/métabolisme , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Sirolimus/analogues et dérivés , Kinases raf/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Évérolimus , Extracellular Signal-Regulated MAP Kinases/métabolisme , Rétrocontrôle physiologique/effets des médicaments et des substances chimiques , Phase G1/effets des médicaments et des substances chimiques , Humains , Imidazoles/pharmacologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinases/métabolisme , Inhibiteurs des phosphoinositide-3 kinases , Phosphorylation , Pyrimidines/pharmacologie , Pyrroles/pharmacologie , Quinoléines/pharmacologie , Sirolimus/pharmacologie , Sérine-thréonine kinases TOR , Facteur de croissance endothéliale vasculaire de type A/métabolisme , Kinases raf/métabolisme
15.
Eur J Endocrinol ; 159(3): 317-27, 2008 Sep.
Article de Anglais | MEDLINE | ID: mdl-18524796

RÉSUMÉ

BACKGROUND/AIMS: Alterations of the IGF system have been described in several different types of cancer. However, no information is available about the role of the IGF system in patients with non-seminomatous germ cell cancer. METHODS: Free IGF-I, IGF-II, acid-labile subunit, and IGF-binding proteins (IGFBPs) 1-4 were analyzed by specific RIAs in 32 patients with untreated non-seminomas and compared with IGFBP levels of 38 healthy controls. Serum IGFBPs were analyzed by western ligand blotting (WLB) and immunoblotting. In 16 patients, IGFBP profiles were measured before, during, and after treatment. RESULTS: In patients with testicular cancer, IGF-II levels were on average 1.44-fold higher than in the healthy control group (1027+/-48 ng/ml versus 711+/-30 ng/ml, P<0.0001). IGFBP-2 levels were on average 2.6-fold higher (586+/-58 ng/ml versus 226+/-17 ng/ml, P<0.001). During follow-up, a decrease in IGFBP-2 levels was observed in all successfully treated patients, which correlated closely with a decrease in the tumor markers alpha-fetoprotein and human chorionic gonadotropin. Additionally, in all patients with recurrent disease, a significant further increase in IGFBP-2 levels (from 358+/-97 to 976+/-260 ng/ml) was detected. IGFBP-3 levels, as measured by RIA, were not different in patients with testicular cancer compared with controls. However, WLB analysis demonstrated markedly decreased intact IGFBP-3 bands in untreated patients and a significant increase after successful therapy. CONCLUSION: Our results demonstrate that markedly elevated IGF-II and IGFBP-2 serum levels in patients with non-seminomatous germ cell cancer, showing a significant decrease after successful therapy and an increase in recurrent disease. Additionally, indirect evidence points to an increased proteolytic activity for IGFBP-3 in untreated testicular cancer patients.


Sujet(s)
Gonadotrophine chorionique/sang , Protéine-2 de liaison aux IGF/sang , Tumeurs embryonnaires et germinales/sang , Tumeurs du testicule/sang , Alphafoetoprotéines/analyse , Adulte , Marqueurs biologiques tumoraux/sang , Études cas-témoins , Humains , Protéine-3 de liaison aux IGF , Protéines de liaison aux IGF/sang , Protéines de liaison aux IGF/métabolisme , Facteur de croissance IGF-II/analyse , Mâle , Adulte d'âge moyen , Tumeurs embryonnaires et germinales/métabolisme , Tumeurs embryonnaires et germinales/anatomopathologie , Tumeurs embryonnaires et germinales/thérapie , Maturation post-traductionnelle des protéines , Récidive , Tumeurs du testicule/métabolisme , Tumeurs du testicule/anatomopathologie , Tumeurs du testicule/thérapie , Régulation positive
16.
Cancer Res ; 67(10): 5025-32, 2007 May 15.
Article de Anglais | MEDLINE | ID: mdl-17510435

RÉSUMÉ

IFN-alpha is commonly used for biotherapy of neuroendocrine carcinomas. However, its antitumor efficacy is often limited due to IFN resistance. In this study, we evaluate the role of suppressor of cytokine signaling protein 1 (SOCS1) in modulating the effects of type I IFNs (IFN-alpha and IFN-beta) in human neuroendocrine BON1 and CM tumor cells. In both cell lines, type I IFNs activated signal transducers and activators of transcription (STAT) and significantly decreased cell viability. However, the effects of IFN-beta were significantly more pronounced than those of IFN-alpha and involved the induction of the intrinsic apoptotic pathway as shown by cleavage of caspase-8, Bid, and caspase-9. Stable overexpression of SOCS1 completely abolished the apoptotic effects of both type I IFNs. In contrast, small interfering RNA (siRNA)-mediated silencing of SOCS1 resulted in strongly enhanced type I IFN signaling as shown by increased and prolonged STAT phosphorylation and stronger induction of apoptosis. Silencing of SOCS1 was associated with down-regulation of basal Bcl-2 and Bcl-xL and up-regulation of basal Bak and Bax, suggesting that reduced SOCS1 expression might lower the threshold of susceptibility to type I IFN-mediated apoptosis by decreasing the ratio of antiapoptotic to proapoptotic molecules. In summary, our results indicate an important role of SOCS1 in IFN resistance of neuroendocrine tumor cells, mediated through negative regulation of type I IFN-induced Jak/STAT signaling. Knocking down SOCS1 by siRNA is a promising new approach to enhance the therapeutic potency of type I IFNs in neuroendocrine tumors.


Sujet(s)
Interféron de type I/pharmacologie , Tumeurs neuroendocrines/thérapie , Tumeurs du pancréas/thérapie , Protéines SOCS/antagonistes et inhibiteurs , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Apoptose/immunologie , Cycle cellulaire/génétique , Cycle cellulaire/immunologie , Lignée cellulaire tumorale , Association thérapeutique , Régulation négative , Humains , Interféron de type I/immunologie , Tumeurs neuroendocrines/génétique , Tumeurs neuroendocrines/immunologie , Tumeurs neuroendocrines/anatomopathologie , Tumeurs du pancréas/génétique , Tumeurs du pancréas/immunologie , Tumeurs du pancréas/anatomopathologie , Phosphorylation , Petit ARN interférent/génétique , Facteurs de transcription STAT/métabolisme , Transduction du signal , Protéine-1 suppressive de la signalisation des cytokines , Protéines SOCS/biosynthèse , Protéines SOCS/génétique , Transfection
17.
Neuroendocrinology ; 85(1): 54-60, 2007.
Article de Anglais | MEDLINE | ID: mdl-17310129

RÉSUMÉ

BACKGROUND/AIM: Tumors exhibiting constitutively activated PI(3)K/Akt/mTOR signaling are hypersensitive to mTOR inhibitors such as RAD001 (everolimus) which is presently being investigated in clinical phase II trials in various tumor entities, including neuroendocrine tumors (NETs). However, no preclinical data about the effects of RAD001 on NET cells have been published. In this study, we aimed to evaluate the effects of RAD001 on BON cells, a human pancreatic NET cell line that exhibits constitutively activated PI(3)K/Akt/mTOR signaling. METHODS: BON cells were treated with different concentrations of RAD001 to analyze its effect on cell growth using proliferation assays. Apoptosis was examined by Western blot analysis of caspase-3/PARP cleavage and by FACS analysis of DNA fragmentation. RESULTS: RAD001 potently inhibited BON cell growth in a dose-dependent manner which was dependent on the serum concentration in the medium. RAD001-induced growth inhibition involved G0/G1-phase arrest as well as induction of apoptosis. CONCLUSION: In summary, our data demonstrate antiproliferative and apoptotic effects of RAD001 in NET cells in vitro supporting its clinical use in current phase II trials in NET patients.


Sujet(s)
Prolifération cellulaire/effets des médicaments et des substances chimiques , Immunosuppresseurs/pharmacologie , Tumeurs neuroectodermiques/physiopathologie , Tumeurs du pancréas/physiopathologie , Sirolimus/analogues et dérivés , Apoptose/effets des médicaments et des substances chimiques , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Relation dose-effet des médicaments , Évérolimus , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Tumeurs neuroectodermiques/traitement médicamenteux , Tumeurs du pancréas/traitement médicamenteux , Sirolimus/pharmacologie
18.
Biochem Biophys Res Commun ; 344(4): 1334-41, 2006 Jun 16.
Article de Anglais | MEDLINE | ID: mdl-16650825

RÉSUMÉ

Interferon-alpha (IFN-alpha) is used for biotherapy of neuroendocrine carcinomas. The interferon-lambdas (IL-28A/B and IL-29) are a novel group of interferons. In this study, we investigated the effects of the IFN-lambdas IL-28A and IL-29 on human neuroendocrine BON1 tumor cells. Similar to IFN-alpha, incubation of BON1 cells with IL-28A (10 ng/ml) and IL-29 (10 ng/ml) induced phosphorylation of STAT1, STAT2, and STAT3, significantly decreased cell numbers in a proliferation assay, and induced apoptosis as demonstrated by poly(ADP-ribose) polymerase (PARP)-cleavage, caspase-3-cleavage, and DNA-fragmentation. Stable overexpression of suppressor of cytokine signaling proteins (SOCS1 and SOCS3) completely abolished the aforementioned effects indicating that SOCS proteins act as negative regulators of IFN-lambda signaling in BON1 cells. In conclusion, the novel IFN-lambdas IL-28A and IL-29 potently induce STAT signaling and antiproliferative effects in neuroendocrine BON1 tumor cells. Thus, IFN-lambdas may hint a promising new approach in the antiproliferative therapy of neuroendocrine tumors.


Sujet(s)
Cytokines/usage thérapeutique , Interférons/usage thérapeutique , Interleukines/usage thérapeutique , Tumeurs neuroendocrines/traitement médicamenteux , Apoptose , Caspase-3 , Caspases/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytokines/pharmacologie , Fragmentation de l'ADN , Humains , Interférons/pharmacologie , Interleukines/pharmacologie , Protéines et peptides de signalisation intracellulaire/métabolisme , Tumeurs neuroendocrines/métabolisme , Phosphorylation , Poly(ADP-ribose) polymerases , Récepteurs aux cytokines/métabolisme , Récepteurs aux interleukines/métabolisme , Récepteurs à l'interleukine-10 , Protéines de répression/métabolisme , Facteurs de transcription STAT/métabolisme , Protéine-1 suppressive de la signalisation des cytokines , Protéine-3 suppressive de la signalisation des cytokine , Protéines SOCS/métabolisme
19.
Neuroimmunomodulation ; 11(4): 224-32, 2004.
Article de Anglais | MEDLINE | ID: mdl-15249728

RÉSUMÉ

OBJECTIVE: This comparative in vitro study examined the effects of all known gp130 cytokines on murine corticotroph AtT-20 cell function. METHODS: Cytokines were tested at equimolar concentrations from 0.078 to 10 nM. Tyrosine phosphorylation of the signal transducer and activator of transcription (STAT)3 and STAT1, the STAT-dependent suppressor of cytokine signaling (SOCS)-3 promoter activity, SOCS-3 gene expression, STAT-dependent POMC promoter activity and adrenocorticotropic hormone (ACTH) secretion were determined. RESULTS: Leukemia inhibitory factor (LIF), human oncostatin M (OSM) and cardiotrophin (CT)-1 (LIFR/gp130 ligands), as well as ciliary neurotrophic factor (CNTF) and novel neurotrophin-1/B-cell stimulating factor-3 (CNTFR alpha/LIFR/gp130 ligands) are potent stimuli of corticotroph cells in vitro. In comparison, interleukin (IL)-6 (IL-6R/gp130 ligand) and IL-11 (IL-11R/gp130 ligand) exhibited only modest direct effects on corticotrophs, while murine OSM (OSMR/gp130 ligand) showed no effect. CONCLUSION: (i) CNTFR complex ligands are potent stimuli of corticotroph function, comparable to LIFR complex ligands; (ii) IL-6 and IL-11 are relatively weak direct stimuli of corticotroph function; (iii) differential effects of human and murine OSM suggest that LIFR/gp130 (OSMR type I) but not OSMR/gp130 (OSMR type II) are involved in corticotroph signaling. (iv) CT-1 has the hitherto unknown ability to stimulate corticotroph function, and (v) despite redundant immuno-neuroendocrine effects of different gp130 cytokines, corticotroph cells are preferably activated through the LIFR and CNTFR complexes.


Sujet(s)
Antigènes CD/métabolisme , Cytokines/pharmacologie , Axe hypothalamohypophysaire/immunologie , Glycoprotéines membranaires/métabolisme , Neuro-immunomodulation/immunologie , Adénohypophyse/immunologie , Hormone corticotrope/métabolisme , Animaux , Antigènes CD/effets des médicaments et des substances chimiques , Antigènes CD/immunologie , Lignée cellulaire , Récepteur gp130 de cytokines , Cytokines/immunologie , Cytokines/métabolisme , Protéines de liaison à l'ADN/effets des médicaments et des substances chimiques , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Relation dose-effet des médicaments , Expression des gènes/effets des médicaments et des substances chimiques , Expression des gènes/immunologie , Axe hypothalamohypophysaire/effets des médicaments et des substances chimiques , Sous-unité alpha du récepteur au facteur d'inhibition de la leucémie , Ligands , Glycoprotéines membranaires/effets des médicaments et des substances chimiques , Glycoprotéines membranaires/immunologie , Souris , Phosphorylation/effets des médicaments et des substances chimiques , Adénohypophyse/cytologie , Adénohypophyse/effets des médicaments et des substances chimiques , Pro-opiomélanocortine/génétique , Régions promotrices (génétique)/effets des médicaments et des substances chimiques , Régions promotrices (génétique)/immunologie , ARN messager/effets des médicaments et des substances chimiques , ARN messager/métabolisme , Récepteur facteur neurotrophique ciliaire/effets des médicaments et des substances chimiques , Récepteur facteur neurotrophique ciliaire/immunologie , Récepteur facteur neurotrophique ciliaire/métabolisme , Récepteurs aux cytokines/effets des médicaments et des substances chimiques , Récepteurs aux cytokines/immunologie , Récepteurs aux cytokines/métabolisme , Récepteurs OSM-LIF , Protéines de répression/génétique , Facteur de transcription STAT-1 , Facteur de transcription STAT-3 , Protéine-3 suppressive de la signalisation des cytokine , Protéines SOCS , Transactivateurs/effets des médicaments et des substances chimiques , Transactivateurs/génétique , Transactivateurs/métabolisme , Facteurs de transcription/génétique , Tyrosine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...