Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 54
Filtrer
1.
Neuro Oncol ; 25(4): 674-686, 2023 04 06.
Article de Anglais | MEDLINE | ID: mdl-36054930

RÉSUMÉ

BACKGROUND: Melanoma, the deadliest of skin cancers, has a high propensity to form brain metastases that are associated with a markedly worsened prognosis. In spite of recent therapeutic advances, melanoma brain lesions remain a clinical challenge, biomarkers predicting brain dissemination are not clear and differences with other metastatic sites are poorly understood. METHODS: We examined a genetically diverse panel of human-derived melanoma brain metastasis (MBM) and extracranial cell lines using targeted sequencing, a Reverse Phase Protein Array, protein expression analyses, and functional studies in vitro and in vivo. RESULTS: Brain-specific genetic alterations were not detected; however, MBM cells in vitro displayed lower proliferation rates and MBM-specific protein expression patterns associated with proliferation, DNA damage, adhesion, and migration. MBM lines displayed higher levels of RAC1 expression, involving a distinct RAC1-PAK1-JNK1 signaling network. RAC1 knockdown or treatment with small molecule inhibitors contributed to a less aggressive MBM phenotype in vitro, while RAC1 knockdown in vivo led to reduced tumor volumes and delayed tumor appearance. Proliferation, adhesion, and migration were higher in MBM vs nonMBM lines in the presence of insulin or brain-derived factors and were affected by RAC1 levels. CONCLUSIONS: Our findings indicate that despite their genetic variability, MBM engage specific molecular processes such as RAC1 signaling to adapt to the brain microenvironment and this can be used for the molecular characterization and treatment of brain metastases.


Sujet(s)
Tumeurs du cerveau , Mélanome , Tumeurs cutanées , Humains , Pronostic , Mélanome/anatomopathologie , Tumeurs du cerveau/génétique , Marqueurs biologiques , Microenvironnement tumoral , Protéine G rac1/métabolisme
2.
EMBO Rep ; 23(11): e54746, 2022 11 07.
Article de Anglais | MEDLINE | ID: mdl-36156348

RÉSUMÉ

Melanoma is the deadliest of skin cancers and has a high tendency to metastasize to distant organs. Calcium and metabolic signals contribute to melanoma invasiveness; however, the underlying molecular details are elusive. The MCU complex is a major route for calcium into the mitochondrial matrix but whether MCU affects melanoma pathobiology was not understood. Here, we show that MCUA expression correlates with melanoma patient survival and is decreased in BRAF kinase inhibitor-resistant melanomas. Knockdown (KD) of MCUA suppresses melanoma cell growth and stimulates migration and invasion. In melanoma xenografts, MCUA_KD reduces tumor volumes but promotes lung metastases. Proteomic analyses and protein microarrays identify pathways that link MCUA and melanoma cell phenotype and suggest a major role for redox regulation. Antioxidants enhance melanoma cell migration, while prooxidants diminish the MCUA_KD -induced invasive phenotype. Furthermore, MCUA_KD increases melanoma cell resistance to immunotherapies and ferroptosis. Collectively, we demonstrate that MCUA controls melanoma aggressive behavior and therapeutic sensitivity. Manipulations of mitochondrial calcium and redox homeostasis, in combination with current therapies, should be considered in treating advanced melanoma.


Sujet(s)
Calcium , Mélanome , Humains , Calcium/métabolisme , Protéomique , Mélanome/génétique , Mélanome/métabolisme , Oxydoréduction , Phénotype , Lignée cellulaire tumorale
3.
J Invest Dermatol ; 142(7): 1882-1892.e5, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-34883044

RÉSUMÉ

The upregulation of the adaptor protein NUMB triggers melanocytic differentiation from multipotent skin stem cells, which share many properties with aggressive melanoma cells. Although NUMB acts as a tumor suppressor in various human cancer types, little is known about its role in melanoma. In this study, we investigated the role of NUMB in melanoma progression and its regulatory mechanism. Analysis of The Cancer Genome Atlas melanoma datasets revealed that high NUMB expression in melanoma tissues correlates with improved patient survival. Moreover, NUMB expression is downregulated in metastatic melanoma cells. NUMB knockdown significantly increased the invasion potential of melanoma cells in a three-dimensional collagen matrix in vitro and in the lungs of a mouse model in vivo; it also significantly upregulated the expression of the NOTCH target gene CCNE. Previous studies suggested that Wnt signaling increases NUMB expression. By mimicking Wnt stimulation through glycogen synthase kinase-3 inhibition, we increased NUMB expression in melanoma cells. Furthermore, a glycogen synthase kinase-3 inhibitor reduced the invasion of melanoma cells in a NUMB-dependent manner. Together, our results suggest that NUMB suppresses invasion and metastasis in melanoma, potentially through its regulation of the NOTCH‒CCNE axis and that the inhibitors that upregulate NUMB can exert therapeutic effects in melanoma.


Sujet(s)
Mélanome , Protéines membranaires , Protéines de tissu nerveux , Animaux , Lignée cellulaire tumorale , Glycogen Synthase Kinases/métabolisme , Humains , Mélanome/traitement médicamenteux , Mélanome/génétique , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris , Protéines de tissu nerveux/génétique , Protéines de tissu nerveux/métabolisme , Voie de signalisation Wnt
4.
Cancer Res ; 81(21): 5540-5554, 2021 11 01.
Article de Anglais | MEDLINE | ID: mdl-34518212

RÉSUMÉ

Despite impressive advances in melanoma-directed immunotherapies, resistance is common and many patients still succumb to metastatic disease. In this context, harnessing natural killer (NK) cells, which have thus far been sidelined in the development of melanoma immunotherapy, could provide therapeutic benefits for cancer treatment. To identify molecular determinants of NK cell-mediated melanoma killing (NKmK), we quantified NK-cell cytotoxicity against a panel of genetically diverse melanoma cell lines and observed highly heterogeneous susceptibility. Melanoma protein microarrays revealed a correlation between NKmK and the abundance and activity of a subset of proteins, including several metabolic factors. Oxidative phoshorylation, measured by oxygen consumption rate, negatively correlated with melanoma cell sensitivity toward NKmK, and proteins involved in mitochondrial metabolism and epithelial-mesenchymal transition were confirmed to regulate NKmK. Two- and three-dimensional killing assays and melanoma xenografts established that the PI3K/AKT/mTOR signaling axis controls NKmK via regulation of NK cell-relevant surface proteins. A "protein-killing-signature" based on the protein analysis predicted NKmK of additional melanoma cell lines and the response of patients with melanoma to anti-PD-1 checkpoint therapy. Collectively, these findings identify novel NK cell-related prognostic biomarkers and may contribute to improved and personalized melanoma-directed immunotherapies. SIGNIFICANCE: NK-cell cytotoxicity assays and protein microarrays reveal novel biomarkers of NK cell-mediated melanoma killing and enable development of signatures to predict melanoma patient responsiveness to immunotherapies.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Biologie informatique/méthodes , Régulation de l'expression des gènes tumoraux , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Immunothérapie/méthodes , Cellules tueuses naturelles/immunologie , Mélanome/anatomopathologie , Animaux , Apoptose , Marqueurs biologiques tumoraux/génétique , Prolifération cellulaire , Cytotoxicité immunologique , Humains , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Cellules tueuses naturelles/métabolisme , Cellules tueuses naturelles/anatomopathologie , Mélanome/traitement médicamenteux , Mélanome/immunologie , Mélanome/métabolisme , Souris , Souris de lignée NOD , Souris SCID , Analyse par réseau de protéines , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Cancer Res ; 81(20): 5230-5241, 2021 10 15.
Article de Anglais | MEDLINE | ID: mdl-34462276

RÉSUMÉ

Metastatic melanoma is challenging to clinically address. Although standard-of-care targeted therapy has high response rates in patients with BRAF-mutant melanoma, therapy relapse occurs in most cases. Intrinsically resistant melanoma cells drive therapy resistance and display molecular and biologic properties akin to neural crest-like stem cells (NCLSC) including high invasiveness, plasticity, and self-renewal capacity. The shared transcriptional programs and vulnerabilities between NCLSCs and cancer cells remains poorly understood. Here, we identify a developmental LPAR1-axis critical for NCLSC viability and melanoma cell survival. LPAR1 activity increased during progression and following acquisition of therapeutic resistance. Notably, genetic inhibition of LPAR1 potentiated BRAFi ± MEKi efficacy and ablated melanoma migration and invasion. Our data define LPAR1 as a new therapeutic target in melanoma and highlights the promise of dissecting stem cell-like pathways hijacked by tumor cells. SIGNIFICANCE: This study identifies an LPAR1-axis critical for melanoma invasion and intrinsic/acquired therapy resistance.


Sujet(s)
Marqueurs biologiques tumoraux/métabolisme , Résistance aux médicaments antinéoplasiques , Régulation de l'expression des gènes tumoraux , Mélanome/anatomopathologie , Crête neurale/anatomopathologie , Cellules souches neurales/anatomopathologie , Récepteurs à l'acide phosphatidique/métabolisme , Animaux , Antinéoplasiques/pharmacologie , Apoptose , Marqueurs biologiques tumoraux/génétique , Prolifération cellulaire , Humains , Mélanome/traitement médicamenteux , Mélanome/génétique , Mélanome/métabolisme , Souris , Souris de lignée NOD , Souris SCID , Crête neurale/effets des médicaments et des substances chimiques , Crête neurale/métabolisme , Cellules souches neurales/effets des médicaments et des substances chimiques , Cellules souches neurales/métabolisme , Pronostic , Récepteurs à l'acide phosphatidique/génétique , Transcriptome , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Cancer Res ; 80(20): 4314-4323, 2020 10 15.
Article de Anglais | MEDLINE | ID: mdl-32641416

RÉSUMÉ

Spread of cancer to the brain remains an unmet clinical need in spite of the increasing number of cases among patients with lung, breast cancer, and melanoma most notably. Although research on brain metastasis was considered a minor aspect in the past due to its untreatable nature and invariable lethality, nowadays, limited but encouraging examples have questioned this statement, making it more attractive for basic and clinical researchers. Evidences of its own biological identity (i.e., specific microenvironment) and particular therapeutic requirements (i.e., presence of blood-brain barrier, blood-tumor barrier, molecular differences with the primary tumor) are thought to be critical aspects that must be functionally exploited using preclinical models. We present the coordinated effort of 19 laboratories to compile comprehensive information related to brain metastasis experimental models. Each laboratory has provided details on the cancer cell lines they have generated or characterized as being capable of forming metastatic colonies in the brain, as well as principle methodologies of brain metastasis research. The Brain Metastasis Cell Lines Panel (BrMPanel) represents the first of its class and includes information about the cell line, how tropism to the brain was established, and the behavior of each model in vivo. These and other aspects described are intended to assist investigators in choosing the most suitable cell line for research on brain metastasis. The main goal of this effort is to facilitate research on this unmet clinical need, to improve models through a collaborative environment, and to promote the exchange of information on these valuable resources.


Sujet(s)
Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/secondaire , Tumeurs expérimentales/anatomopathologie , Animaux , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Techniques de culture cellulaire/méthodes , Lignée cellulaire tumorale , Humains , Souris , Rats , Tropisme , Microenvironnement tumoral , Tests d'activité antitumorale sur modèle de xénogreffe
7.
EMBO J ; 38(15): e100871, 2019 08 01.
Article de Anglais | MEDLINE | ID: mdl-31304984

RÉSUMÉ

Reactive oxygen species (ROS) are emerging as important regulators of cancer growth and metastatic spread. However, how cells integrate redox signals to affect cancer progression is not fully understood. Mitochondria are cellular redox hubs, which are highly regulated by interactions with neighboring organelles. Here, we investigated how ROS at the endoplasmic reticulum (ER)-mitochondria interface are generated and translated to affect melanoma outcome. We show that TMX1 and TMX3 oxidoreductases, which promote ER-mitochondria communication, are upregulated in melanoma cells and patient samples. TMX knockdown altered mitochondrial organization, enhanced bioenergetics, and elevated mitochondrial- and NOX4-derived ROS. The TMX-knockdown-induced oxidative stress suppressed melanoma proliferation, migration, and xenograft tumor growth by inhibiting NFAT1. Furthermore, we identified NFAT1-positive and NFAT1-negative melanoma subgroups, wherein NFAT1 expression correlates with melanoma stage and metastatic potential. Integrative bioinformatics revealed that genes coding for mitochondrial- and redox-related proteins are under NFAT1 control and indicated that TMX1, TMX3, and NFAT1 are associated with poor disease outcome. Our study unravels a novel redox-controlled ER-mitochondria-NFAT1 signaling loop that regulates melanoma pathobiology and provides biomarkers indicative of aggressive disease.


Sujet(s)
Mélanome/anatomopathologie , Protéines membranaires/métabolisme , Facteurs de transcription NFATC/métabolisme , Oxydoréduction , Protein Disulfide-Isomerases/métabolisme , Thiorédoxines/métabolisme , Animaux , Lignée cellulaire tumorale , Noyau de la cellule/métabolisme , Évolution de la maladie , Réticulum endoplasmique/métabolisme , Régulation de l'expression des gènes tumoraux , Humains , Mâle , Mélanome/métabolisme , Protéines membranaires/génétique , Souris , Mitochondries/métabolisme , NADPH Oxidase 4/métabolisme , Transplantation tumorale , Transport des protéines , Espèces réactives de l'oxygène/métabolisme , Transduction du signal , Analyse de survie , Thiorédoxines/génétique , Régulation positive
8.
Methods Mol Biol ; 1925: 183-196, 2019.
Article de Anglais | MEDLINE | ID: mdl-30674028

RÉSUMÉ

Oxidative modifications of cellular building blocks such as proteins, lipids, and DNA have a major impact on cell behavior, fate, and clinical outcome. Reactive oxygen species (ROS) are important factors that influence these redox processes. Calcium ion (Ca2+) dynamics and signals are also essential regulators of key cellular processes. Therefore, the combined and precise monitoring of ROS and Ca2+ in single cells, with a high spatial and temporal resolution and in physiological environments, is essential to better understand their functional impact. Here, we describe protocols to detect one of the most prominent ROS (hydrogen peroxide, H2O2) using genetically encoded protein sensors and fluorescent dyes. We also provide guidelines on how to simultaneously detect Ca2+ and H2O2 and how to examine the influence of Ca2+ signals on cellular ROS production and vice versa.


Sujet(s)
Techniques de biocapteur/méthodes , Calcium/analyse , Colorants fluorescents/analyse , Peroxyde d'hydrogène/analyse , Protéines luminescentes/analyse , Microscopie de fluorescence/méthodes , Techniques de culture cellulaire/méthodes , Cellules HEK293 , Humains , Protéines luminescentes/génétique , Espèces réactives de l'oxygène/analyse
9.
Pflugers Arch ; 470(8): 1149-1163, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-29926229

RÉSUMÉ

The important role of mitochondria in cancer biology is gaining momentum. With their regulation of cell survival, metabolism, basic cell building blocks, and immunity, among other functions, mitochondria affect not only cancer progression but also the response and resistance to current treatments. Calcium ions are constantly shuttled in and out of mitochondria; thus, playing an important role in the regulation of various cellular processes. The mitochondrial calcium uniporter (MCU) channel and its associated regulators transport calcium across the inner mitochondrial membrane to the mitochondrial matrix. Due to this central role and the capacity to affect cell behavior and fate, the MCU complex is being investigated in different cancers and cancer-related conditions. Here, we review current knowledge on the role of the MCU complex in multiple cancer types and models; we also provide a perspective for future research and clinical considerations.


Sujet(s)
Canaux calciques/métabolisme , Calcium/métabolisme , Mitochondries/métabolisme , Tumeurs/métabolisme , Animaux , Humains , Membranes mitochondriales/métabolisme
10.
Bio Protoc ; 8(2): e2705, 2018 Jan 20.
Article de Anglais | MEDLINE | ID: mdl-34179249

RÉSUMÉ

Reactive oxygen species (ROS) are not only known for their toxic effects on cells, but they also play an important role as second messengers. As such, they control a variety of cellular functions such as proliferation, metabolism, differentiation and apoptosis. Thus, ROS are involved in the regulation of multiple physiological and pathophysiological processes. It is now apparent that there are transient and local changes in ROS in the cell; in so-called 'microdomains' or in specific cellular compartments, which affect signaling events. These ROS hotspots need to be studied in more depth to understand their function and regulation. Therefore, it is necessary to identify and quantify redox signals in single cells with high spatial and temporal resolution. Genetically encoded fluorescence-based protein sensors provide such necessary tools to examine redox-signaling processes. A big advantage of these sensors is the possibility to target them specifically. Mitochondria are essential for energy metabolism and are one of the major sources of ROS in mammalian cells. Therefore, the evaluation of redox potential and ROS production in these organelles is of great interest. Herein, we provide a protocol for the real-time visualization of mitochondrial hydrogen peroxide (H2O2) using the H2O2-specific ratiometric sensor mitoHyPer in adherent mammalian cells.

11.
Cell Rep ; 21(7): 1953-1967, 2017 Nov 14.
Article de Anglais | MEDLINE | ID: mdl-29141225

RÉSUMÉ

Therapy of advanced melanoma is changing dramatically. Following mutational and biological subclassification of this heterogeneous cancer, several targeted and immune therapies were approved and increased survival significantly. To facilitate further advancements through pre-clinical in vivo modeling, we have established 459 patient-derived xenografts (PDX) and live tissue samples from 384 patients representing the full spectrum of clinical, therapeutic, mutational, and biological heterogeneity of melanoma. PDX have been characterized using targeted sequencing and protein arrays and are clinically annotated. This exhaustive live tissue resource includes PDX from 57 samples resistant to targeted therapy, 61 samples from responders and non-responders to immune checkpoint blockade, and 31 samples from brain metastasis. Uveal, mucosal, and acral subtypes are represented as well. We show examples of pre-clinical trials that highlight how the PDX collection can be used to develop and optimize precision therapies, biomarkers of response, and the targeting of rare genetic subgroups.


Sujet(s)
Hétérogreffes/anatomopathologie , Mélanome/anatomopathologie , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes , Animaux , Cellules cultivées , Hétérogreffes/métabolisme , Humains , Mélanome/classification , Mélanome/génétique , Souris
12.
Oncotarget ; 8(70): 115140-115152, 2017 Dec 29.
Article de Anglais | MEDLINE | ID: mdl-29383148

RÉSUMÉ

Angiogenesis is a critical step during tumor progression. Anti-angiogenic therapy has only provided modest benefits in delaying tumor progression despite its early promise in cancer treatment. It has been postulated that anti-angiogenic therapy may promote the emergence of a more aggressive cancer cell phenotype by generating increased tumor hypoxia-a well-recognized promoter of tumor progression. TH-302 is a 2-nitroimidazole triggered hypoxia-activated prodrug (HAP) which has been shown to selectively target the hypoxic tumor compartment and reduce tumor volume. Here, we show that melanoma cells grown under hypoxic conditions exhibit increased resistance to a wide variety of therapeutic agents in vitro and generate larger and more aggressive tumors in vivo than melanoma cells grown under normoxic conditions. However, hypoxic melanoma cells exhibit a pronounced sensitivity to TH-302 which is further enhanced by the addition of sunitinib. Short term sunitinib treatment fails to prolong the survival of melanoma bearing genetically engineered mice (Tyr::CreER; BRafCA;Ptenlox/lox ) but increases tumor hypoxia. Long term TH-302 alone modestly prolongs the overall survival of melanoma bearing mice. Combination therapy of TH-302 with sunitinib further increases the survival of treated mice. These studies provide a translational rationale for combining hypoxic tumor cell targeted therapies with anti-angiogenics for treatment of melanoma.

13.
Oncotarget ; 7(44): 71211-71222, 2016 11 01.
Article de Anglais | MEDLINE | ID: mdl-27655717

RÉSUMÉ

The discovery of activating BRAF mutations in approximately 50% of melanomas has led to the development of MAPK pathway inhibitors, which have transformed melanoma therapy. However, not all BRAF-V600E melanomas respond to MAPK inhibition. Therefore, it is important to understand why tumors with the same oncogenic driver have variable responses to MAPK inhibitors. Here, we show that concurrent loss of PTEN and activation of the Notch pathway is associated with poor response to the ERK inhibitor SCH772984, and that co-inhibition of Notch and ERK decreased viability in BRAF-V600E melanomas. Additionally, patients with low PTEN and Notch activation had significantly shorter progression free survival when treated with BRAF inhibitors. Our studies provide a rationale to further develop combination strategies with Notch antagonists to maximize the efficacy of MAPK inhibition in melanoma. Our findings should prompt the evaluation of combinations co-targeting MAPK/ERK and Notch as a strategy to improve current therapies and warrant further evaluation of co-occurrence of aberrant PTEN and Notch activation as predictive markers of response to therapy.


Sujet(s)
Extracellular Signal-Regulated MAP Kinases/antagonistes et inhibiteurs , Indazoles/usage thérapeutique , Mélanome/traitement médicamenteux , Pipérazines/usage thérapeutique , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes B-raf/génétique , Récepteurs Notch/antagonistes et inhibiteurs , Amyloid precursor protein secretases/antagonistes et inhibiteurs , Animaux , Lignée cellulaire tumorale , Humains , Mélanome/génétique , Mélanome/anatomopathologie , Souris , Phosphohydrolase PTEN/physiologie , Phosphatidylinositol 3-kinases/physiologie , Récepteurs Notch/physiologie
14.
Oncotarget ; 7(34): 54897-54912, 2016 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-27448973

RÉSUMÉ

Therapeutic strategies for the treatment of metastatic melanoma show encouraging results in the clinic; however, not all patients respond equally and tumor resistance still poses a challenge. To identify novel therapeutic targets for melanoma, we screened a panel of structurally diverse organometallic inhibitors against human-derived normal and melanoma cells. We observed that a compound that targets PIM kinases (a family of Ser/Thr kinases) preferentially inhibited melanoma cell proliferation, invasion, and viability in adherent and three-dimensional (3D) melanoma models. Assessment of tumor tissue from melanoma patients showed that PIM kinases are expressed in pre- and post-treatment tumors, suggesting PIM kinases as promising targets in the clinic. Using knockdown studies, we showed that PIM1 contributes to melanoma cell proliferation and tumor growth in vivo; however, the presence of PIM2 and PIM3 could also influence the outcome. The inhibition of all PIM isoforms using SGI-1776 (a clinically-available PIM inhibitor) reduced melanoma proliferation and survival in preclinical models of melanoma. This was potentiated in the presence of the BRAF inhibitor PLX4720 and in the presence of PI3K inhibitors. Our findings suggest that PIM inhibitors provide promising additions to the targeted therapies available to melanoma patients.


Sujet(s)
Imidazoles/pharmacologie , Mélanome/traitement médicamenteux , Protéines proto-oncogènes c-pim-1/antagonistes et inhibiteurs , Pyridazines/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe , Animaux , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/génétique , Analyse de profil d'expression de gènes/méthodes , Composés hétérocycliques 3 noyaux/administration et posologie , Composés hétérocycliques 3 noyaux/pharmacologie , Humains , Imidazoles/administration et posologie , Indoles/administration et posologie , Indoles/pharmacologie , Mélanome/génétique , Mélanome/métabolisme , Souris de lignée NOD , Souris knockout , Souris SCID , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes/antagonistes et inhibiteurs , Protéines proto-oncogènes/génétique , Protéines proto-oncogènes/métabolisme , Protéines proto-oncogènes c-pim-1/génétique , Protéines proto-oncogènes c-pim-1/métabolisme , Pyridazines/administration et posologie , Pyrimidinones/administration et posologie , Pyrimidinones/pharmacologie , Interférence par ARN , Sulfonamides/administration et posologie , Sulfonamides/pharmacologie , Charge tumorale/effets des médicaments et des substances chimiques , Charge tumorale/génétique , ortho-Aminobenzoates/administration et posologie , ortho-Aminobenzoates/pharmacologie
16.
Pigment Cell Melanoma Res ; 29(3): 317-28, 2016 May.
Article de Anglais | MEDLINE | ID: mdl-26850518

RÉSUMÉ

Targeted therapies for mutant BRAF metastatic melanoma are effective but not curative due to acquisition of resistance. PI3K signaling is a common mediator of therapy resistance in melanoma; thus, the need for effective PI3K inhibitors is critical. However, testing PI3K inhibitors in adherent cultures is not always reflective of their potential in vivo. To emphasize this, we compared PI3K inhibitors of different specificity in two- and three-dimensional (2D, 3D) melanoma models and show that drug response predictions gain from evaluation using 3D models. Our results in 3D demonstrate the anti-invasive potential of PI3K inhibitors and that drugs such as PX-866 have beneficial activity in physiological models alone and when combined with BRAF inhibition. These assays finally help highlight pathway effectors that could be involved in drug response in different environments (e.g. p4E-BP1). Our findings show the advantages of 3D melanoma models to enhance our understanding of PI3K inhibitors.


Sujet(s)
Mélanome/anatomopathologie , Modèles biologiques , Inhibiteurs des phosphoinositide-3 kinases , Inhibiteurs de protéines kinases/pharmacologie , Animaux , Adhérence cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Collagène/pharmacologie , Stérane/pharmacologie , Indoles/pharmacologie , Mélanome/enzymologie , Souris de lignée NOD , Mutation/génétique , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Protéines proto-oncogènes B-raf/métabolisme , Protéines proto-oncogènes c-akt/antagonistes et inhibiteurs , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/anatomopathologie , Sulfonamides/pharmacologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques
17.
J Physiol ; 594(11): 2825-35, 2016 06 01.
Article de Anglais | MEDLINE | ID: mdl-26864956

RÉSUMÉ

Calcium signalling within normal and cancer cells regulates many important cellular functions such as migration, proliferation, differentiation and cytokine secretion. Store operated Ca(2+) entry (SOCE) via the Ca(2+) release activated Ca(2+) (CRAC) channels, which are composed of the plasma membrane based Orai channels and the endoplasmic reticulum stromal interaction molecules (STIMs), is a major Ca(2+) entry route in many cell types. Orai and STIM have been implicated in the growth and metastasis of multiple cancers; however, while their involvement in cancer is presently indisputable, how Orai-STIM-controlled Ca(2+) signals affect malignant transformation, tumour growth and invasion is not fully understood. Here, we review recent studies linking Orai-STIM Ca(2+) channels with cancer, with a particular focus on melanoma. We highlight and examine key molecular players and the signalling pathways regulated by Orai and STIM in normal and malignant cells, we expose discrepancies, and we reflect on the potential of Orai-STIMs as anticancer drug targets. Finally, we discuss the functional implications of future discoveries in the field of Ca(2+) signalling.


Sujet(s)
Mélanocytes/métabolisme , Mélanome/métabolisme , Protéine ORAI1/physiologie , Molécule-1 d'interaction stromale/physiologie , Animaux , Humains , Mélanocytes/anatomopathologie , Mélanome/anatomopathologie
19.
Cancer Discov ; 3(12): 1378-93, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-24104062

RÉSUMÉ

UNLABELLED: An emerging concept in melanoma biology is that of dynamic, adaptive phenotype switching, where cells switch from a highly proliferative, poorly invasive phenotype to a highly invasive, less proliferative one. This switch may hold significant implications not just for metastasis, but also for therapy resistance. We demonstrate that phenotype switching and subsequent resistance can be guided by changes in expression of receptors involved in the noncanonical Wnt5A signaling pathway, ROR1 and ROR2. ROR1 and ROR2 are inversely expressed in melanomas and negatively regulate each other. Furthermore, hypoxia initiates a shift of ROR1-positive melanomas to a more invasive, ROR2-positive phenotype. Notably, this receptor switch induces a 10-fold decrease in sensitivity to BRAF inhibitors. In patients with melanoma treated with the BRAF inhibitor vemurafenib, Wnt5A expression correlates with clinical response and therapy resistance. These data highlight the fact that mechanisms that guide metastatic progression may be linked to those that mediate therapy resistance. SIGNIFICANCE: These data show for the fi rst time that a single signaling pathway, the Wnt signaling pathway, can effectively guide the phenotypic plasticity of tumor cells, when primed to do so by a hypoxic microenvironment. Importantly, this increased Wnt5A signaling can give rise to a subpopulation of highly invasive cells that are intrinsically less sensitive to novel therapies for melanoma, and targeting the Wnt5A/ROR2 axis could improve the efficacy and duration of response for patients with melanoma on vemurafenib.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Hypoxie cellulaire , Résistance aux médicaments antinéoplasiques , Indoles/usage thérapeutique , Mélanome/traitement médicamenteux , Mélanome/métabolisme , Récepteurs orphelins de type récepteur à tyrosine kinase/métabolisme , Sulfonamides/usage thérapeutique , Animaux , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Régulation de l'expression des gènes tumoraux , Techniques de knock-down de gènes , Humains , Mélanome/génétique , Mélanome/secondaire , Mélanome expérimental , Souris , Souris nude , Métastase tumorale , Phénotype , Récepteurs orphelins de type récepteur à tyrosine kinase/génétique , Vémurafénib , Voie de signalisation Wnt
20.
Cell Rep ; 4(6): 1090-9, 2013 Sep 26.
Article de Anglais | MEDLINE | ID: mdl-24055054

RÉSUMÉ

Although BRAF and MEK inhibitors have proven clinical benefits in melanoma, most patients develop resistance. We report a de novo MEK2-Q60P mutation and BRAF gain in a melanoma from a patient who progressed on the MEK inhibitor trametinib and did not respond to the BRAF inhibitor dabrafenib. We also identified the same MEK2-Q60P mutation along with BRAF amplification in a xenograft tumor derived from a second melanoma patient resistant to the combination of dabrafenib and trametinib. Melanoma cells chronically exposed to trametinib acquired concurrent MEK2-Q60P mutation and BRAF-V600E amplification, which conferred resistance to MEK and BRAF inhibitors. The resistant cells had sustained MAPK activation and persistent phosphorylation of S6K. A triple combination of dabrafenib, trametinib, and the PI3K/mTOR inhibitor GSK2126458 led to sustained tumor growth inhibition. Hence, concurrent genetic events that sustain MAPK signaling can underlie resistance to both BRAF and MEK inhibitors, requiring novel therapeutic strategies to overcome it.


Sujet(s)
MAP Kinase Kinase 2/génétique , Mélanome/traitement médicamenteux , Mélanome/génétique , Mutation , Inhibiteurs de protéines kinases/pharmacologie , Protéines proto-oncogènes B-raf/génétique , Lignée cellulaire tumorale , Résistance aux médicaments antinéoplasiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Amplification de gène , Humains , MAP Kinase Kinase 2/antagonistes et inhibiteurs , MAP Kinase Kinase 2/composition chimique , Mâle , Mélanome/enzymologie , Mélanome/anatomopathologie , Adulte d'âge moyen , Modèles moléculaires , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Ribosomal Protein S6 Kinases/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE