Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Endocr Relat Cancer ; 28(1): 39-51, 2021 01.
Article in English | MEDLINE | ID: mdl-33112824

ABSTRACT

Although there is evidence of a significant rise of neuroendocrine neoplasms (NENs) incidence, current treatments are largely insufficient due to somewhat poor knowledge of these tumours. Despite showing differentiated features, NENs exhibit therapeutic resistance to most common treatments, similar to other cancers in many instances. Molecular mechanisms responsible for this resistance phenomenon are badly understood. We aimed at identifying signalling partners responsible of acquired resistance to treatments in order to develop novel therapeutic strategies. We engineered QGP-1 cells resistant to current leading treatments, the chemotherapeutic agent oxaliplatin and the mTor inhibitor everolimus. Cells were chronically exposed to the drugs and assessed for acquired resistance by viability assay. We used microarray-based kinomics to obtain highthroughput kinase activity profiles from drug sensitive vs resistant cells and identified 'hit' kinases hyperactivated in drug-resistant cells, including kinases from FGFR family, cyclin-dependant kinases and PKCs in oxaliplatin-resistant (R-Ox) QGP-1 cells. We then validated these 'hit' kinases and observed that ERK signalling is specifically enhanced in QGP-1 R-Ox cells. Finally, we assessed drug-resistant cells sensitivity to pharmacological inhibition of 'hit' kinases or their signalling partners. We found that FGFR inhibition markedly decreased ERK signalling and cell viability in QGP-1 R-Ox cells. These results suggest that the FGFR/ERK axis is hyperactivated in response to oxaliplatin-based chemotherapeutic strategy. Thus, this sensitive approach, based on the study of kinome activity, allows identifying potential candidates involved in drug resistance in NENs and may be used to broadly investigate markers of NENs therapeutic response.


Subject(s)
Antineoplastic Agents/therapeutic use , Neuroendocrine Tumors/drug therapy , Proteomics/methods , Aged , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Female , Humans , Male , Middle Aged
2.
Oncotarget ; 8(25): 41044-41063, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28454119

ABSTRACT

Therapeutic management of gastroenteropancreatic neuroendocrine tumors (GEP-NETs) is challenging. The mammalian target of rapamycin (mTOR) inhibitor everolimus recently obtained approval from the Food and Drug Administration for the treatment of patients with advanced pancreatic neuroendocrine tumors (pNETs). Despite its promising antitumor efficacy observed in cell lines, clinical benefit for patients is unsatisfactory. The limited therapeutic potential of everolimus in cancer cells has been attributed to Akt activation due to feedback loops relief following mTOR inhibition. Combined inhibition of Akt might then improve everolimus antitumoral effect. In this regard, the somatostatin analog (SSA) octreotide has been shown to repress the PI3K/Akt pathway in some tumor cell lines. Moreover, SSAs are well tolerated and routinely used to reduce symptoms caused by peptide release in patients carrying functional GEP-NETs. We have recently established and characterized primary cultures of human pNETs and demonstrated the anti-proliferative effects of both octreotide and pasireotide. In this study, we aim at determining the antitumor efficacy of everolimus alone or in combination with the SSAs octreotide and pasireotide in primary cultures of pNETs. Everolimus reduced both Chromogranin A secretion and cell viability and upregulated Akt activity in single treatment. Its anti-proliferative and anti-secretory efficacy was not improved combined with the SSAs. Both SSAs did not overcome everolimus-induced Akt upregulation. Furthermore, caspase-dependent apoptosis induced by SSAs was lost in combined treatments. These molecular events provide the first evidence supporting the lack of marked benefit in patients co-treated with everolimus and SSA.


Subject(s)
Chromogranin A/metabolism , Everolimus/pharmacology , Octreotide/pharmacology , Somatostatin/analogs & derivatives , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Drug Synergism , Humans , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Somatostatin/pharmacology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
3.
Soins Gerontol ; 22(124): 17-22, 2017.
Article in French | MEDLINE | ID: mdl-28413011

ABSTRACT

The Marseille gerontology network consists of a geriatric team providing support in patients' homes across the Marseille urban area. It forms an integral part of the geriatric care sector ensuring that elderly people living at home benefit from a global medical-psychological-social assessment. The aim is to enable elderly people to continue living and receiving care in their home.


Subject(s)
Health Services for the Aged/organization & administration , Aged , Geriatric Nursing , Geriatrics , Humans
4.
Endocr Relat Cancer ; 21(5): 691-704, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25012983

ABSTRACT

Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) raise difficult therapeutic problems despite the emergence of targeted therapies. Somatostatin analogs (SSA) remain pivotal therapeutic drugs. However, the tachyphylaxis and the limited antitumoral effects observed with the classical somatostatin 2 (sst2) agonists (octreotide and lanreotide) led to the development of new SSA, such as the pan sst receptor agonist pasireotide. Our aim was to compare the effects of pasireotide and octreotide on cell survival, chromogranin A (CgA) secretion, and sst2 phosphorylation/trafficking in pancreatic NET (pNET) primary cells from 15 tumors. We established and characterized the primary cultures of human pancreatic tumors (pNETs) as powerful preclinical models for understanding the biological effects of SSA. At clinically relevant concentrations (1-10 nM), pasireotide was at least as efficient as octreotide in inhibiting CgA secretion and cell viability through caspase-dependent apoptosis during short treatments, irrespective of the expression levels of the different sst receptors or the WHO grade of the parental tumor. Interestingly, unlike octreotide, which induces a rapid and persistent partial internalization of sst2 associated with its phosphorylation on Ser341/343, pasireotide did not phosphorylate sst2 and induced a rapid and transient internalization of the receptor followed by a persistent recycling at the cell surface. These results provide the first evidence, to our knowledge, of striking differences in the dynamics of sst2 trafficking in pNET cells treated with the two SSAs, but with similar efficiency in the control of CgA secretion and cell viability.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Intestinal Neoplasms/metabolism , Neuroendocrine Tumors/metabolism , Octreotide/pharmacology , Pancreatic Neoplasms/metabolism , Receptors, Somatostatin/metabolism , Somatostatin/analogs & derivatives , Stomach Neoplasms/metabolism , Adult , Aged , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Chromogranin A/metabolism , Cyclic AMP/metabolism , DNA Fragmentation , Female , Humans , Male , Middle Aged , Somatostatin/pharmacology , Tumor Cells, Cultured
5.
Cell Signal ; 24(12): 2237-48, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22940095

ABSTRACT

The Ras/Raf/MEK/ERK is a conserved signalling pathway involved in the control of fundamental cellular processes. Despite extensive research, how this pathway can process a myriad of diverse extracellular inputs into substrate specificity to determine biological outcomes is not fully understood. It has been established that the ERK1/2 pathway is an integrative point in the control of the pituitary function exerted by various extracellular signals. In addition we previously established that the GTPases Ras and Rap1 play a key role in the regulation of ERK1/2-dependent prolactin transcription by EGF or the cAMP-dependent neuropeptide VIP. In this report, using the FRET-based biosensor of ERK activity (EKAR) in the pituitary GH4C1 cell line, we show that both EGF and VIP tightly control the spatiotemporal dynamic of activated ERK with different magnitude and duration. Importantly, we provide the first evidence of a differential control of cytoplasmic and nuclear pools of activated ERK by the GTPases Ras and Rap1. Ras is required for nuclear magnitude and duration of EGF-dependent ERK activation, whereas it is required for both VIP-activated cytoplasmic and nuclear ERK pools. Rap1 is exclusively involved in VIP-activated ERK nuclear pool. Moreover, consistent with the control of the nuclear pool of activated ERK by the GTPases, we observe the same differential role of Ras and Rap1 on ERK nuclear translocation triggered by EGF or VIP. Together these findings identify Ras and Rap1 as determinant partners in shaping nuclear and cytoplasmic ERK kinetics in response to EGF and VIP, which in turn should control pituitary secretion.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , Epidermal Growth Factor/pharmacology , Fluorescence Resonance Energy Transfer , Pituitary Gland/cytology , Pituitary Gland/metabolism , Prolactin/metabolism , Rats , Signal Transduction/drug effects , Vasoactive Intestinal Peptide/pharmacology
6.
Ann N Y Acad Sci ; 1220: 60-70, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21388404

ABSTRACT

In pituitary somatolactotroph cells, G protein-coupled receptors and receptor tyrosine kinases binding their specific ligands trigger an enzymatic cascade that converges to MAP kinase activation in the subcellular compartment. Different signaling pathways, such as AC/cAMP/PKA and PI3K/Akt pathways, interact with MAP kinase to regulate key physiological functions, such as hormonal secretion and cell proliferation. Abnormalities affecting these signaling pathways have been identified as preponderant factors of pituitary tumorigenesis. In addition to trans-sphenoidal surgery, somatostatin analogs are used to control hormonal hypersecretion in GH-secreting adenomas. However, a subset of these tumors remains uncontrolled with these treatFments, calling for new therapeutic approaches. In these cases, novel multivalent somatostatin analogs or new somatostatin-dopamine chimeric molecules could be of interest. Another attractive therapeutic approach may be to use one or several inhibitors acting downstream in the signaling pathway, such as mammalian target of rapamycin inhibitor. Cotargeting therapy and gene therapy are promising tools for these problematic pituitary tumors.


Subject(s)
Pituitary Gland/physiopathology , Signal Transduction , Animals , Humans , Pituitary Gland/metabolism
8.
Int J Cancer ; 121(6): 1245-52, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17514647

ABSTRACT

Gs alpha, the alpha-subunit of the heterotrimeric GTP-binding protein, is coded from the GNAS gene, which is imprinted in a tissue-specific manner. Gs alpha is paternally silenced in normal pituitary, but Gs alpha imprinting relaxation is found in some tumoral tissue. In addition, Gs alpha mRNA levels are high in some somatotroph adenomas not bearing the active Gs alpha mutant, the gsp oncogene. In this study, the impact of loss of imprinting on Gs alpha expression level and on tumoral phenotype has been investigated. We compared the expression and imprinting of 4 transcripts of GNAS locus (NESP55, XL alpha s, exon 1A, Gs alpha) of 60 somatotroph adenomas with those of 23 lactotroph adenomas. The paternal and maternal transcripts were quantified using allele-specific real-time PCR and FokI polymorphism. Moreover, the methylation of exon 1A DMR was analyzed. As is the case for the gsp oncogene, high Gs alpha expression in gsp- tumors was associated with smaller tumor size and better octreotide sensitivity. A strong imprinting relaxation (percentage of paternal Gs alpha expression >or=7.5%) was found only in gsp- tumors. The loss of Gs alpha imprinting was associated with a decrease in exon 1A mRNA expression. Unexpectedly, the methylation status of exon 1A DMR was not modified in relaxed tumors. Maternal Gs alpha mRNA level decreased with exon 1A level, and consequently the loss of Gs alpha imprinting did not induce the expected Gs alpha overexpression. Finally, XL alpha s mRNA level correlated with that of paternal Gs alpha and of NESP55 showing the complexity of gene regulation in the GNAS locus.


Subject(s)
Adenoma/genetics , Drug Resistance, Neoplasm/genetics , GTP-Binding Protein alpha Subunits, Gs/genetics , Genomic Imprinting , Growth Hormone-Secreting Pituitary Adenoma/genetics , Adenoma/drug therapy , Adenoma/pathology , Antineoplastic Agents, Hormonal/therapeutic use , Blotting, Western , DNA Methylation , Growth Hormone-Secreting Pituitary Adenoma/drug therapy , Growth Hormone-Secreting Pituitary Adenoma/pathology , Humans , Octreotide/therapeutic use , Prolactinoma/drug therapy , Prolactinoma/genetics , Prolactinoma/pathology , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction
9.
Endocrinology ; 147(12): 6036-45, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16935846

ABSTRACT

In pituitary cells, prolactin (PRL) synthesis and release are controlled by multiple transduction pathways. In the GH4C1 somatolactotroph cell line, we previously reported that MAPK ERK-1/2 are a point of convergence between the pathways involved in the PRL gene regulation. In the present study, we focused on the involvement of the phosphoinositide 3-kinase (PI3K)/Akt pathway in the MAPK ERK-1/2 regulation and PRL secretion in pituitary cells. Either specific pharmacological PI3K and Akt inhibitors (LY294002, Akt I, and phosphoinositide analog-6) or Akt dominant-negative mutant (K179M) enhanced ERK-1/2 phosphorylation in unstimulated GH4C1 cells. Under the same conditions, PI3K and Akt inhibition also both increased Raf-1 kinase activity and the levels of GTP-bound (active form) monomeric G protein Rap1, which suggests that a down-regulation of the ERK-1/2 cascade is induced by the PI3K/Akt signaling pathway in unstimulated cells. On the contrary, ERK-1/2 phosphorylation, Raf-1 activity, and Rap1 activation were almost completely blocked in IGF-I-stimulated cells previously subjected to PI3K or Akt inhibition. Although the PRL promoter was not affected by either PI3K/Akt inhibition or activation, PRL release increased in response to the pharmacological PI3K/Akt inhibitors in unstimulated GH4C1 and rat pituitary primary cells. The IGF-I-stimulated PRL secretion was diminished, on the contrary, by the pharmacological PI3K/Akt inhibitors. Taken together, these findings indicate that the PI3K/Akt pathway exerts dual regulatory effects on both the Rap1/Raf-1/ERK-1/2 cascade and PRL release in pituitary cells, i.e. negative effects in unstimulated cells and positive ones in IGF-I-stimulated cells.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphatidylinositol 3-Kinases/physiology , Prolactin/metabolism , Proto-Oncogene Proteins c-akt/physiology , Proto-Oncogene Proteins c-raf/metabolism , Somatotrophs/metabolism , rap1 GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , Female , Pituitary Gland/cytology , Pituitary Gland/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Rats , Rats, Wistar , Receptor Cross-Talk , Signal Transduction , Transcriptional Activation , ras Proteins/metabolism
10.
J Biol Chem ; 278(51): 51386-94, 2003 Dec 19.
Article in English | MEDLINE | ID: mdl-14551200

ABSTRACT

In pituitary cells, transcriptional regulation of the prolactin (PRL) gene and prolactin secretion are controlled by multiple transduction pathways through the activation of G protein coupled receptors and receptor tyrosine kinases. In the somatolactotrope GH4C1 cell line, we have previously identified crosstalk between the MAPKinase cascade ERK1/2 and the cAMP/protein kinase A pathway after the activation of the VPAC2 receptor by vasoactive intestinal polypeptide (VIP) or pituitary adenylyl cyclase-activating polypeptide (PACAP38). In the present study, we focus on the involvement of the GTPases Ras and Rap1 as downstream components of signal transmission initiated by activation of the VPAC2 receptor. By using pull-down experiments, we show that VIP and PACAP38 preferentially activate Rap1, whereas thyrotropin releasing hormone (TRH) and epidermal growth factor (EGF) mainly activate Ras GTPase. Experiments involving the expression of the dominant-negative mutants of Ras and Rap1 signaling (RasN17 or Rap1N17) indicate that both GTPases Ras and Rap1 are recruited for the ERK activation by VIP and PACAP38, whereas Rap1 is poorly involved in TRH or EGF-induced ERK activation. The use of U0126, a selective inhibitor of MAPKinase kinase, provides evidence that MAPKinase contributes to the regulation of the PRL gene. Moreover, cotransfection of RasN17 or Rap1N17 with the PRL proximal promoter luciferase reporter construct indicates that Rap1 may be responsible for VIP/PACAP-induced activation of the PRL promoter. Interestingly, Ras would be involved as a negative regulator of VIP/PACAP-induced PRL gene activation, in contrast to its stimulatory role in the regulation of the PRL promoter by TRH and EGF.


Subject(s)
Neuropeptides/metabolism , Prolactin/genetics , Vasoactive Intestinal Peptide/metabolism , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Animals , Cell Line, Tumor , Epidermal Growth Factor/metabolism , Gene Expression Regulation , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide , Pituitary Gland/cytology , Prolactin/biosynthesis , Promoter Regions, Genetic , Rats , Receptors, Vasoactive Intestinal Peptide/metabolism , Receptors, Vasoactive Intestinal Peptide, Type II , Signal Transduction , Thyrotropin-Releasing Hormone/metabolism , Transcription, Genetic , Transcriptional Activation , rap1 GTP-Binding Proteins/physiology , ras Proteins/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...