Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
J Cell Sci ; 127(Pt 6): 1346-56, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24424027

RESUMEN

The target of rapamycin (TOR) kinase regulates cell growth and division. Rapamycin only inhibits a subset of TOR activities. Here we show that in contrast to the mild impact of rapamycin on cell division, blocking the catalytic site of TOR with the Torin1 inhibitor completely arrests growth without cell death in Schizosaccharomyces pombe. A mutation of the Tor2 glycine residue (G2040D) that lies adjacent to the key Torin-interacting tryptophan provides Torin1 resistance, confirming the specificity of Torin1 for TOR. Using this mutation, we show that Torin1 advanced mitotic onset before inducing growth arrest. In contrast to TOR inhibition with rapamycin, regulation by either Wee1 or Cdc25 was sufficient for this Torin1-induced advanced mitosis. Torin1 promoted a Polo and Cdr2 kinase-controlled drop in Wee1 levels. Experiments in human cell lines recapitulated these yeast observations: mammalian TOR (mTOR) was inhibited by Torin1, Wee1 levels declined and mitotic commitment was advanced in HeLa cells. Thus, the regulation of the mitotic inhibitor Wee1 by TOR signalling is a conserved mechanism that helps to couple cell cycle and growth controls.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Mitosis/efectos de los fármacos , Naftiridinas/farmacología , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/crecimiento & desarrollo , Secuencia de Aminoácidos , Dominio Catalítico , Muerte Celular , Resistencia a Medicamentos , Puntos de Control de la Fase G1 del Ciclo Celular , Células HeLa , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Diana Mecanicista del Complejo 2 de la Rapamicina , Datos de Secuencia Molecular , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Mutagénesis Sitio-Dirigida , Fosfatidilinositol 3-Quinasas/metabolismo , Transporte de Proteínas , Schizosaccharomyces/efectos de los fármacos , Schizosaccharomyces/enzimología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
2.
Mol Cancer ; 13: 154, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24941944

RESUMEN

BACKGROUND: The importance of the genetic background of cancer cells for the individual susceptibility to cancer treatments is increasingly apparent. In melanoma, the existence of a BRAF mutation is a main predictor for successful BRAF-targeted therapy. However, despite initial successes with these therapies, patients relapse within a year and have to move on to other therapies. Moreover, patients harbouring a wild type BRAF gene (including 25% with NRAS mutations) still require alternative treatment such as chemotherapy. Multiple genetic parameters have been associated with response to chemotherapy, but despite their high frequency in melanoma nothing is known about the impact of BRAF or NRAS mutations on the response to chemotherapeutic agents. METHODS: Using cell proliferation and DNA methylation assays, FACS analysis and quantitative-RT-PCR we have characterised the response of a panel of NRAS and BRAF mutant melanoma cell lines to various chemotherapy drugs, amongst them dacarbazine (DTIC) and temozolomide (TMZ) and DNA synthesis inhibitors. RESULTS: Although both, DTIC and TMZ act as alkylating agents through the same intermediate, NRAS and BRAF mutant cells responded differentially only to DTIC. Further analysis revealed that the growth-inhibitory effects mediated by DTIC were rather due to interference with nucleotide salvaging, and that NRAS mutant melanoma cells exhibit higher activity of the nucleotide synthesis enzymes IMPDH and TK1. Importantly, the enhanced ability of RAS mutant cells to use nucleotide salvaging resulted in resistance to DHFR inhibitors. CONCLUSION: In summary, our data suggest that the genetic background in melanoma cells influences the response to inhibitors blocking de novo DNA synthesis, and that defining the RAS mutation status could be used to stratify patients for the use of antifolate drugs.


Asunto(s)
GTP Fosfohidrolasas/genética , Melanoma/tratamiento farmacológico , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias Cutáneas/tratamiento farmacológico , Antineoplásicos/administración & dosificación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN/biosíntesis , ADN/genética , Metilación de ADN/genética , Dacarbazina/administración & dosificación , Dacarbazina/análogos & derivados , Inhibidores Enzimáticos/administración & dosificación , Humanos , Melanoma/genética , Melanoma/patología , Mutación , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Temozolomida
4.
Biochem Soc Trans ; 42(4): 727-32, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25109949

RESUMEN

The serine threonine kinases BRAF and MEK [MAPK (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) kinase] are major regulators of the ERK/MAPK pathway, which is deregulated in the majority of melanomas. Targeting BRAF is an effective therapy for advanced melanoma, but patients progress due to the development of resistance. This 'acquired resistance' is thought to be based on a minority of tumour cell populations that are resistant and will eventually re-establish tumour growth even in the presence of drug. In particular, mutations, amplifications or overexpression of genes encoding regulators of the MAPK pathway can confer this resistance, because it allows the melanoma cells to bypass inhibitor action by stimulating ERK activation through alternative routes. Furthermore, there are mechanisms that produce resistance by enhancing the tolerance of melanoma cells to the cytotoxic effects of the drug. These compensatory mechanisms can activate survival signals in the melanoma cells without reactivating ERK. Besides these cell-autonomous resistance mechanisms, stromal fibroblasts in the tumour microenvironment have been identified as a potential source of resistance, because these cells can produce growth factors that reactivate ERK through paracrine signalling. Understanding and further identifying mechanisms of resistance is crucial for the future treatment of advanced melanoma, because this can inform the design of improved therapies with more durable responses.


Asunto(s)
Melanoma/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Animales , Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/fisiología , Humanos , Melanoma/tratamiento farmacológico , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal/efectos de los fármacos
5.
Pharmaceutics ; 16(6)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38931850

RESUMEN

The standard of care for glioblastoma (GBM) involves surgery followed by adjuvant radio- and chemotherapy, but often within months, patients relapse, and this has been linked to glioma stem cells (GSCs), self-renewing cells with increased therapy resistance. The identification of the epidermal growth factor receptor (EGFR) and platelet-derived growth factor receptor (PDGFR) as key players in gliomagenesis inspired the development of inhibitors targeting these tyrosine kinases (TKIs). However, results from clinical trials testing TKIs have been disappointing, and while the role of GSCs in conventional therapy resistance has been extensively studied, less is known about resistance of GSCs to TKIs. In this study, we have used compartmentalised proteomics to analyse the adaptive response of GSCs to ponatinib, a TKI with activity against PDGFR. The analysis of differentially expressed proteins revealed that GSCs respond to ponatinib by broadly rewiring lipid metabolism, involving fatty acid beta-oxidation, cholesterol synthesis, and sphingolipid degradation. Inhibiting each of these metabolic pathways overcame ponatinib adaptation of GSCs, but interrogation of patient data revealed sphingolipid degradation as the most relevant pathway in GBM. Our data highlight that targeting lipid metabolism, and particularly sphingolipid degradation in combinatorial therapies, could improve the outcome of TKI therapies using ponatinib in GBM.

6.
Nature ; 445(7130): 851-7, 2007 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-17314971

RESUMEN

Melanoma is a cancer that arises from melanocytes, specialized pigmented cells that are found predominantly in the skin. The incidence of melanoma is rising steadily in western populations--the number of cases worldwide has doubled in the past 20 years. In its early stages malignant melanoma can be cured by surgical resection, but once it has progressed to the metastatic stage it is extremely difficult to treat and does not respond to current therapies. Recent discoveries in cell signalling have provided greater understanding of the biology that underlies melanoma, and these advances are being exploited to provide targeted drugs and new therapeutic approaches.


Asunto(s)
Melanoma/tratamiento farmacológico , Melanoma/fisiopatología , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/fisiopatología , Animales , Senescencia Celular , Humanos , Melanoma/genética , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/metabolismo , Transducción de Señal/efectos de los fármacos , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
7.
J Invest Dermatol ; 143(2): 305-316.e5, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36058299

RESUMEN

Circulating tumor cells are the key link between a primary tumor and distant metastases, but once in the bloodstream, loss of adhesion induces cell death. To identify the mechanisms relevant for melanoma circulating tumor cell survival, we performed RNA sequencing and discovered that detached melanoma cells and isolated melanoma circulating tumor cells rewire lipid metabolism by upregulating fatty acid (FA) transport and FA beta-oxidation‒related genes. In patients with melanoma, high expression of FA transporters and FA beta-oxidation enzymes significantly correlates with reduced progression-free and overall survival. Among the highest expressed regulators in melanoma circulating tumor cells were the carnitine transferases carnitine O-octanoyltransferase and carnitine acetyltransferase, which control the shuttle of peroxisome-derived medium-chain FAs toward mitochondria to fuel mitochondrial FA beta-oxidation. Knockdown of carnitine O-octanoyltransferase or carnitine acetyltransferase and short-term treatment with peroxisomal or mitochondrial FA beta-oxidation inhibitors thioridazine or ranolazine suppressed melanoma metastasis in mice. Carnitine O-octanoyltransferase and carnitine acetyltransferase depletion could be rescued by medium-chain FA supplementation, indicating that the peroxisomal supply of FAs is crucial for the survival of nonadherent melanoma cells. Our study identifies targeting the FA-based cross-talk between peroxisomes and mitochondria as a potential therapeutic opportunity to challenge melanoma progression. Moreover, the discovery of the antimetastatic activity of the Food and Drug Administration‒approved drug ranolazine carries translational potential.


Asunto(s)
Melanoma , Células Neoplásicas Circulantes , Ratones , Animales , Carnitina O-Acetiltransferasa/genética , Carnitina O-Acetiltransferasa/metabolismo , Carnitina Aciltransferasas/genética , Carnitina Aciltransferasas/metabolismo , Ranolazina , Oxidación-Reducción , Ácidos Grasos/metabolismo , Melanoma/tratamiento farmacológico , Carnitina/metabolismo
8.
Nat Metab ; 5(9): 1544-1562, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37563469

RESUMEN

Resistance of melanoma to targeted therapy and immunotherapy is linked to metabolic rewiring. Here, we show that increased fatty acid oxidation (FAO) during prolonged BRAF inhibitor (BRAFi) treatment contributes to acquired therapy resistance in mice. Targeting FAO using the US Food and Drug Administration-approved and European Medicines Agency-approved anti-anginal drug ranolazine (RANO) delays tumour recurrence with acquired BRAFi resistance. Single-cell RNA-sequencing analysis reveals that RANO diminishes the abundance of the therapy-resistant NGFRhi neural crest stem cell subpopulation. Moreover, by rewiring the methionine salvage pathway, RANO enhances melanoma immunogenicity through increased antigen presentation and interferon signalling. Combination of RANO with anti-PD-L1 antibodies strongly improves survival by increasing antitumour immune responses. Altogether, we show that RANO increases the efficacy of targeted melanoma therapy through its effects on FAO and the methionine salvage pathway. Importantly, our study suggests that RANO could sensitize BRAFi-resistant tumours to immunotherapy. Since RANO has very mild side-effects, it might constitute a therapeutic option to improve the two main strategies currently used to treat metastatic melanoma.


Asunto(s)
Melanoma , Estados Unidos , Animales , Ratones , Ranolazina/farmacología , Ranolazina/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Inmunoterapia , Inhibidores de Proteínas Quinasas/farmacología , Metionina
9.
Front Immunol ; 13: 861545, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35669782

RESUMEN

Cutaneous melanoma is one of the most aggressive human malignancies and shows increasing incidence. Mast cells (MCs), long-lived tissue-resident cells that are particularly abundant in human skin where they regulate both innate and adaptive immunity, are associated with melanoma stroma (MAMCs). Thus, MAMCs could impact melanoma development, progression, and metastasis by secreting proteases, pro-angiogenic factors, and both pro-inflammatory and immuno-inhibitory mediators. To interrogate the as-yet poorly characterized role of human MAMCs, we have purified MCs from melanoma skin biopsies and performed RNA-seq analysis. Here, we demonstrate that MAMCs display a unique transcriptome signature defined by the downregulation of the FcεRI signaling pathway, a distinct expression pattern of proteases and pro-angiogenic factors, and a profound upregulation of complement component C3. Furthermore, in melanoma tissue, we observe a significantly increased number of C3+ MCs in stage IV melanoma. Moreover, in patients, C3 expression significantly correlates with the MC-specific marker TPSAB1, and the high expression of both markers is linked with poorer melanoma survival. In vitro, we show that melanoma cell supernatants and tumor microenvironment (TME) mediators such as TGF-ß, IL-33, and IL-1ß induce some of the changes found in MAMCs and significantly modulate C3 expression and activity in MCs. Taken together, these data suggest that melanoma-secreted cytokines such as TGF-ß and IL-1ß contribute to the melanoma microenvironment by upregulating C3 expression in MAMCs, thus inducing an MC phenotype switch that negatively impacts melanoma prognosis.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Complemento C3/metabolismo , Humanos , Mastocitos , Melanoma/patología , Péptido Hidrolasas/metabolismo , Neoplasias Cutáneas/patología , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral/genética , Regulación hacia Arriba , Melanoma Cutáneo Maligno
10.
Cell Rep ; 39(12): 110995, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35732120

RESUMEN

Dysregulated cellular metabolism is a cancer hallmark for which few druggable oncoprotein targets have been identified. Increased fatty acid (FA) acquisition allows cancer cells to meet their heightened membrane biogenesis, bioenergy, and signaling needs. Excess FAs are toxic to non-transformed cells but surprisingly not to cancer cells. Molecules underlying this cancer adaptation may provide alternative drug targets. Here, we demonstrate that diacylglycerol O-acyltransferase 1 (DGAT1), an enzyme integral to triacylglyceride synthesis and lipid droplet formation, is frequently up-regulated in melanoma, allowing melanoma cells to tolerate excess FA. DGAT1 over-expression alone transforms p53-mutant zebrafish melanocytes and co-operates with oncogenic BRAF or NRAS for more rapid melanoma formation. Antagonism of DGAT1 induces oxidative stress in melanoma cells, which adapt by up-regulating cellular reactive oxygen species defenses. We show that inhibiting both DGAT1 and superoxide dismutase 1 profoundly suppress tumor growth through eliciting intolerable oxidative stress.


Asunto(s)
Diacilglicerol O-Acetiltransferasa , Melanoma , Animales , Diacilglicerol O-Acetiltransferasa/genética , Diacilglicerol O-Acetiltransferasa/metabolismo , Proteínas Oncogénicas/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno , Triglicéridos , Pez Cebra/metabolismo
11.
Cancers (Basel) ; 13(2)2021 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-33478100

RESUMEN

(1) Background: Despite the indisputable effectiveness of dexamethasone (DEXA) to reduce inflammation in glioblastoma (GBM) patients, its influence on tumour progression and radiotherapy response remains controversial. (2) Methods: We analysed patient data and used expression and cell biological analyses to assess effects of DEXA on GBM cells. We tested the efficacy of tyrosine kinase inhibitors in vitro and in vivo. (3) Results: We confirm in our patient cohort that administration of DEXA correlates with worse overall survival and shorter time to relapse. In GBM cells and glioma stem-like cells (GSCs) DEXA down-regulates genes controlling G2/M and mitotic-spindle checkpoints, and it enables cells to override the spindle assembly checkpoint (SAC). Concurrently, DEXA up-regulates Platelet Derived Growth Factor Receptor (PDGFR) signalling, which stimulates expression of anti-apoptotic regulators BCL2L1 and MCL1, required for survival during extended mitosis. Importantly, the protective potential of DEXA is dependent on intact tyrosine kinase signalling and ponatinib, sunitinib and dasatinib, all effectively overcome the radio-protective and pro-proliferative activity of DEXA. Moreover, we discovered that DEXA-induced signalling creates a therapeutic vulnerability for sunitinib in GSCs and GBM cells in vitro and in vivo. (4) Conclusions: Our results reveal a novel DEXA-induced mechanism in GBM cells and provide a rationale for revisiting the use of tyrosine kinase inhibitors for the treatment of GBM.

12.
Immunology ; 129(4): 610-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20074207

RESUMEN

As alpha-melanocyte-stimulating hormone (alpha-MSH) is released by immunocompetent cells and has potent immunosuppressive properties, it was determined whether human dendritic cells (DCs) express the receptor for this hormone. Reverse transcription-polymerase chain reaction detected messenger RNA specific for all of the known melanocortin receptors in DCs. Mixed lymphocyte reactions also revealed that treatment with [Nle(4), DPhe(7)]-alpha-MSH (NDP-MSH), a potent alpha-MSH analogue, significantly reduced the ability of DCs to stimulate allogeneic T cells. The expression of various cell surface adhesion, maturation and costimulatory molecules on DCs was also investigated. Although treatment with NDP-MSH did not alter the expression of CD83 and major histocompatibility complex class I and II, the surface expression of CD86 (B7.2), intercellular adhesion molecule (ICAM-1/CD54) and CD1a was reduced. In summary, our data indicate that NDP-MSH inhibits the functional activity of DCs, possibly by down-regulating antigen-presenting and adhesion molecules and that these events may be mediated via the extracellular signal-regulated kinase 1 and 2 pathway.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Receptores de Melanocortina/agonistas , alfa-MSH/farmacología , Antígenos CD1/inmunología , Antígenos CD1/metabolismo , Antígeno B7-2/inmunología , Antígeno B7-2/metabolismo , Células Cultivadas , Células Dendríticas/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Humanos , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Fosforilación/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Melanocortina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Trasplante Homólogo
13.
J Cell Biol ; 170(5): 703-8, 2005 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-16129781

RESUMEN

The protein kinase B-RAF is a human oncogene that is mutated in approximately 70% of human melanomas and transforms mouse melanocytes. Microphthalmia-associated transcription factor (MITF) is an important melanocyte differentiation and survival factor, but its role in melanoma is unclear. In this study, we show that MITF expression is suppressed by oncogenic B-RAF in immortalized mouse and primary human melanocytes. However, low levels of MITF persist in human melanoma cells harboring oncogenic B-RAF, suggesting that additional mechanisms regulate its expression. MITF reexpression in B-RAF-transformed melanocytes inhibits their proliferation. Furthermore, differentiation-inducing factors that elevate MITF expression in melanoma cells inhibit their proliferation, but when MITF up-regulation is prevented by RNA interference, proliferation is not inhibited. These data suggest that MITF is an anti-proliferation factor that is down-regulated by B-RAF signaling and that this is a crucial event for the progression of melanomas that harbor oncogenic B-RAF.


Asunto(s)
Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Melanocitos/fisiología , Melanoma/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Línea Celular Transformada , AMP Cíclico/metabolismo , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Melanocitos/citología , Melanoma/patología , Ratones , Factor de Transcripción Asociado a Microftalmía , Proteínas Proto-Oncogénicas B-raf/genética , Interferencia de ARN , Transducción de Señal/fisiología , Factores de Transcripción/genética
14.
Pigment Cell Melanoma Res ; 33(1): 74-85, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31323160

RESUMEN

Melanoma is the deadliest form of skin cancer; a primary driver of this high level of morbidity is the propensity of melanoma cells to metastasize. When malignant tumours develop distant metastatic lesions the new local tissue niche is known to impact on the biology of the cancer cells. However, little is known about how different metastatic tissue sites impact on frontline targeted therapies. Intriguingly, melanoma bone lesions have significantly lower response to BRAF or MEK inhibitor therapies. Here, we have investigated how the cellular niche of the bone can support melanoma cells by stimulating growth and survival via paracrine signalling between osteoblasts and cancer cells. Melanoma cells can enhance the differentiation of osteoblasts leading to increased production of secreted ligands, including RANKL. Differentiated osteoblasts in turn can support melanoma cell proliferation and survival via the secretion of RANKL that elevates the levels of the transcription factor MITF, even in the presence of BRAF inhibitor. By blocking RANKL signalling, either via neutralizing antibodies, genetic alterations or the RANKL receptor inhibitor SPD304, the survival advantage provided by osteoblasts could be overcome.


Asunto(s)
Melanoma/patología , Osteoblastos/patología , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/metabolismo , Ligando RANK/metabolismo , Transducción de Señal
15.
Pigment Cell Melanoma Res ; 33(5): 695-708, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32145051

RESUMEN

A major challenge for managing melanoma is its tumour heterogeneity based on individual co-existing melanoma cell phenotypes. These phenotypes display variable responses to standard therapies, and they drive individual steps of melanoma progression; hence, understanding their behaviour is imperative. Melanoma phenotypes are defined by distinct transcriptional states, which relate to different melanocyte lineage development phases, ranging from a mesenchymal, neural crest-like to a proliferative, melanocytic phenotype. It is thought that adaptive phenotype plasticity based on transcriptional reprogramming drives melanoma progression, but at which stage individual phenotypes dominate and moreover, how they interact is poorly understood. We monitored melanocytic and mesenchymal phenotypes throughout melanoma progression and detected transcriptional reprogramming at different stages, with a gain in mesenchymal traits in circulating melanoma cells (CTCs) and proliferative features in metastatic tumours. Intriguingly, we found that distinct phenotype populations interact in a cooperative manner, which generates tumours of greater "fitness," supports CTCs and expands organotropic cues in metastases. Fibronectin, expressed in mesenchymal cells, acts as key player in cooperativity and promotes survival of melanocytic cells. Our data reveal an important role for inter-phenotype communications at various stages of disease progression, suggesting these communications could act as therapeutic target.


Asunto(s)
Adaptación Fisiológica , Comunicación Celular , Progresión de la Enfermedad , Melanoma/patología , Animales , Línea Celular Tumoral , Proliferación Celular , Fibronectinas/metabolismo , Humanos , Melanocitos/patología , Mesodermo/patología , Ratones , Metástasis de la Neoplasia , Células Neoplásicas Circulantes/patología , Fenotipo
16.
Nat Rev Cancer ; 19(7): 377-391, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31209265

RESUMEN

Malignant melanoma is notorious for its inter- and intratumour heterogeneity, based on transcriptionally distinct melanoma cell phenotypes. It is thought that these distinct phenotypes are plastic in nature and that their transcriptional reprogramming enables heterogeneous tumours both to undergo different stages of melanoma progression and to adjust to drug exposure during treatment. Recent advances in genomic technologies and the rapidly expanding availability of large gene expression datasets have allowed for a refined definition of the gene signatures that characterize these phenotypes and have revealed that phenotype plasticity plays a major role in the resistance to both targeted therapy and immunotherapy. In this Review we discuss the definition of melanoma phenotypes through particular transcriptional states and reveal the prognostic relevance of the related gene expression signatures. We review how the establishment of phenotypes is controlled and which roles phenotype plasticity plays in melanoma development and therapy. Because phenotype plasticity in melanoma bears a great resemblance to epithelial-mesenchymal transition, the lessons learned from melanoma will also benefit our understanding of other cancer types.


Asunto(s)
Adaptación Fisiológica/fisiología , Melanoma/genética , Melanoma/patología , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Humanos , Inmunoterapia , Melanoma/terapia , Fenotipo
17.
Pigment Cell Melanoma Res ; 32(2): 280-291, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30277012

RESUMEN

The BRAF kinase and the MAPK pathway are targets of current melanoma therapies. However, MAPK pathway inhibition results in dynamic changes of downstream targets that can counteract inhibitor-action not only in during treatment, but also in acquired resistant tumours. One such dynamic change involves the expression of the transcription factor MITF, a crucial regulator of cell survival and proliferation in untreated as well as drug-addicted acquired resistant melanoma. Tight control over MITF expression levels is required for optimal melanoma growth, and while it is well established that the MAPK pathway regulates MITF expression, the actual mechanism is insufficiently understood. We reveal here, how BRAF through action on the transcription factors BRN2 and PAX3 executes control over the regulation of MITF expression in a manner that allows for considerable plasticity. This plasticity provides robustness to the BRAF mediated MITF regulation and explains the dynamics in MITF expression that are observed in patients in response to MAPK inhibitor therapy.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción PAX3/metabolismo , Factores del Dominio POU/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Secuencia de Bases , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/patología , Factor de Transcripción Asociado a Microftalmía/metabolismo , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Tirosina Quinasa del Receptor Axl
18.
Curr Biol ; 15(18): 1629-39, 2005 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-16169484

RESUMEN

BACKGROUND: Malignant melanoma is a highly aggressive neoplastic disease whose incidence is increasing rapidly. In recent years, the use of interferon alpha (IFNalpha) has become the most established adjuvant immunotherapy for melanoma of advanced stage. IFNalpha is a potent inhibitor of melanoma cell proliferation, and the signal transducer and activator of transcription STAT1 is crucial for its antiproliferative action. Although advanced melanomas clinically resistant to IFNalpha are frequently characterized by inefficient STAT1 signaling, the mechanisms underlying advanced-stage interferon resistance are poorly understood. RESULTS: Here, we demonstrate that IFNalpha activates STAT5 in melanoma cells and that in IFNalpha-resistant cells STAT5 is overexpressed. Significantly, the knockdown of STAT5 in interferon-resistant melanoma cells restored the growth-inhibitory response to IFNalpha. When STAT5 was overexpressed in IFNalpha-sensitive cells, it counteracted interferon-induced growth inhibition. The overexpressed STAT5 diminished IFNalpha-triggered STAT1 activation, most evidently through upregulation of the inhibitor of cytokine-signaling CIS. CONCLUSIONS: Our data demonstrate that overexpression and activation of STAT5 enable melanoma cells to overcome cytokine-mediated antiproliferative signaling. Thus, overexpression of STAT5 can counteract IFNalpha signaling in melanoma cells, and this finally can result in cytokine-resistant and progressively growing tumor cells. These findings have significant implications for the clinical failure of IFNalpha therapy of advanced melanoma because they demonstrate that IFNalpha induces the activation of STAT5 in melanoma cells, and in STAT5-overexpressing cells, this contributes to IFNalpha resistance.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/fisiología , Inmunoterapia/efectos adversos , Interferón-alfa/farmacología , Melanoma/metabolismo , Factor de Transcripción STAT1/metabolismo , Factor de Transcripción STAT5/metabolismo , Western Blotting , Línea Celular Tumoral , Citocinas/antagonistas & inhibidores , Cartilla de ADN , Resistencia a Antineoplásicos/efectos de los fármacos , Técnica del Anticuerpo Fluorescente , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Inmunohistoquímica , Inmunoprecipitación , Interferón-alfa/metabolismo , Melanoma/terapia , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
19.
Cancer Res ; 66(6): 3145-52, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16540665

RESUMEN

One of the most prominent features of malignant melanoma is the fast generation of metastasizing cells, resulting in the poor prognosis of patients with this tumor type. For this process, cells must gain the ability to migrate. The oncogenic receptor Xmrk (Xiphophorus melanoma receptor kinase) from the Xiphophorus melanoma system is a mutationally activated version of the epidermal growth factor receptor that induces the malignant transformation of pigment cells. Here, we show that the activation of Xmrk leads to a clear increase of pigment cell motility in a fyn-dependent manner. Stimulation of Xmrk induces its interaction with the focal adhesion kinase (FAK) and the interaction of active, receptor-bound fyn with FAK. This results in changes in FAK activity and induces the modulation of stress fibers and focal adhesions. Overexpression of dominant-negative FAK shows that the activity of innate FAK and a receptor-induced focal adhesion turnover are a prerequisite for pigment cell migration. Our findings show that in our system, Xmrk is sufficient for the induction of pigment cell motility and underlines a role of the src family protein tyrosine kinase fyn in melanoma development and progression.


Asunto(s)
Movimiento Celular/fisiología , Proteínas de Peces/fisiología , Adhesiones Focales/fisiología , Melanocitos/citología , Melanoma/patología , Proteínas Tirosina Quinasas Receptoras/fisiología , Animales , Ciprinodontiformes , Proteínas de Peces/antagonistas & inhibidores , Proteínas de Peces/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Humanos , Melanocitos/enzimología , Melanoma/enzimología , Ratones , Proteínas Proto-Oncogénicas c-fyn/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/metabolismo
20.
Oncogene ; 37(23): 3166-3182, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29545604

RESUMEN

Despite the general focus on an invasive and de-differentiated phenotype as main driver of cancer metastasis, in melanoma patients many metastatic lesions display a high degree of pigmentation, indicative for a differentiated phenotype. Indeed, studies in mice and fish show that melanoma cells switch to a differentiated phenotype at secondary sites, possibly because in melanoma differentiation is closely linked to proliferation through the lineage-specific transcriptional master regulator MITF. Importantly, while a lot of effort has gone into identifying factors that induce the de-differentiated/invasive phenotype, it is not well understood how the switch to the differentiated/proliferative phenotype is controlled. We identify collagen as a contributor to this switch. We demonstrate that collagen stiffness induces melanoma differentiation through a YAP/PAX3/MITF axis and show that in melanoma patients increased collagen abundance correlates with nuclear YAP localization. However, the interrogation of large patient datasets revealed that in the context of the tumour microenvironment, YAP function is more complex. In the absence of fibroblasts, YAP/PAX3-mediated transcription prevails, but in the presence of fibroblasts tumour growth factor-ß suppresses YAP/PAX3-mediated MITF expression and induces YAP/TEAD/SMAD-driven transcription and a de-differentiated phenotype. Intriguingly, while high collagen expression is correlated with poorer patient survival, the worst prognosis is seen in patients with high collagen expression, who also express MITF target genes such as the differentiation markers TRPM1, TYR and TYRP1, as well as CDK4. In summary, we reveal a distinct lineage-specific route of YAP signalling that contributes to the regulation of melanoma pigmentation and uncovers a set of potential biomarkers predictive for poor survival.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Colágeno/metabolismo , Melanoma/patología , Fosfoproteínas/metabolismo , Factores de Transcripción/metabolismo , Aciltransferasas , Proteínas Adaptadoras Transductoras de Señales/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Adhesión Celular , Diferenciación Celular , Línea Celular Tumoral , Linaje de la Célula , Núcleo Celular/metabolismo , Proliferación Celular , Matriz Extracelular/metabolismo , Matriz Extracelular/patología , Fibroblastos/metabolismo , Fibroblastos/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Melanoma/genética , Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Fosfoproteínas/genética , Factores de Transcripción/genética , Microambiente Tumoral , Proteínas Señalizadoras YAP
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA