Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
PLoS Biol ; 20(2): e3001317, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35192608

RESUMEN

Cell invasion is an initiating event during tumor cell metastasis and an essential process during development. A screen of C. elegans orthologs of genes overexpressed in invasive human melanoma cells has identified several components of the conserved DNA pre-replication complex (pre-RC) as positive regulators of anchor cell (AC) invasion. The pre-RC genes function cell-autonomously in the G1-arrested AC to promote invasion, independently of their role in licensing DNA replication origins in proliferating cells. While the helicase activity of the pre-RC is necessary for AC invasion, the downstream acting DNA replication initiation factors are not required. The pre-RC promotes the invasive fate by regulating the expression of extracellular matrix genes and components of the PI3K signaling pathway. Increasing PI3K pathway activity partially suppressed the AC invasion defects caused by pre-RC depletion, suggesting that the PI3K pathway is one critical pre-RC target. We propose that the pre-RC, or a part of it, acts in the postmitotic AC as a transcriptional regulator that facilitates the switch to an invasive phenotype.


Asunto(s)
Caenorhabditis elegans/genética , Ciclo Celular/genética , Movimiento Celular/genética , Replicación del ADN/genética , Origen de Réplica/genética , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/citología , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Femenino , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Larva/citología , Larva/genética , Larva/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/genética , Vulva/citología , Vulva/metabolismo
2.
EMBO J ; 38(15): e95874, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31267558

RESUMEN

MAPK inhibitors (MAPKi) show outstanding clinical response rates in melanoma patients harbouring BRAF mutations, but resistance is common. The ability of melanoma cells to switch from melanocytic to mesenchymal phenotypes appears to be associated with therapeutic resistance. High-throughput, subcellular proteome analyses and RNAseq on two panels of primary melanoma cells that were either sensitive or resistant to MAPKi revealed that only 15 proteins were sufficient to distinguish between these phenotypes. The two proteins with the highest discriminatory power were PTRF and IGFBP7, which were both highly upregulated in the mesenchymal-resistant cells. Proteomic analysis of CRISPR/Cas-derived PTRF knockouts revealed targets involved in lysosomal activation, endocytosis, pH regulation, EMT, TGFß signalling and cell migration and adhesion, as well as a significantly reduced invasive index and ability to form spheres in 3D culture. Overexpression of PTRF led to MAPKi resistance, increased cell adhesion and sphere formation. In addition, immunohistochemistry of patient samples showed that PTRF expression levels were a significant biomarker of poor progression-free survival, and IGFBP7 levels in patient sera were shown to be higher after relapse.


Asunto(s)
Resistencia a Antineoplásicos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Melanoma/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteómica/métodos , Proteínas de Unión al ARN/metabolismo , Adulto , Anciano , Carbamatos/farmacología , Adhesión Celular , Línea Celular Tumoral , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Masculino , Melanoma/tratamiento farmacológico , Melanoma/genética , Persona de Mediana Edad , Mapas de Interacción de Proteínas , Análisis de Secuencia de ARN , Sulfonamidas/farmacología , Análisis de Supervivencia , Regulación hacia Arriba , Vemurafenib/farmacología
3.
Nature ; 547(7664): 453-457, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28678785

RESUMEN

Plasticity of the cell state has been proposed to drive resistance to multiple classes of cancer therapies, thereby limiting their effectiveness. A high-mesenchymal cell state observed in human tumours and cancer cell lines has been associated with resistance to multiple treatment modalities across diverse cancer lineages, but the mechanistic underpinning for this state has remained incompletely understood. Here we molecularly characterize this therapy-resistant high-mesenchymal cell state in human cancer cell lines and organoids and show that it depends on a druggable lipid-peroxidase pathway that protects against ferroptosis, a non-apoptotic form of cell death induced by the build-up of toxic lipid peroxides. We show that this cell state is characterized by activity of enzymes that promote the synthesis of polyunsaturated lipids. These lipids are the substrates for lipid peroxidation by lipoxygenase enzymes. This lipid metabolism creates a dependency on pathways converging on the phospholipid glutathione peroxidase (GPX4), a selenocysteine-containing enzyme that dissipates lipid peroxides and thereby prevents the iron-mediated reactions of peroxides that induce ferroptotic cell death. Dependency on GPX4 was found to exist across diverse therapy-resistant states characterized by high expression of ZEB1, including epithelial-mesenchymal transition in epithelial-derived carcinomas, TGFß-mediated therapy-resistance in melanoma, treatment-induced neuroendocrine transdifferentiation in prostate cancer, and sarcomas, which are fixed in a mesenchymal state owing to their cells of origin. We identify vulnerability to ferroptic cell death induced by inhibition of a lipid peroxidase pathway as a feature of therapy-resistant cancer cells across diverse mesenchymal cell-state contexts.


Asunto(s)
Glutatión Peroxidasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Cadherinas/metabolismo , Muerte Celular , Línea Celular Tumoral , Linaje de la Célula , Transdiferenciación Celular , Resistencia a Antineoplásicos/genética , Transición Epitelial-Mesenquimal , Humanos , Hierro/metabolismo , Peróxidos Lipídicos/metabolismo , Masculino , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Melanoma/metabolismo , Melanoma/patología , Mesodermo/efectos de los fármacos , Mesodermo/enzimología , Mesodermo/metabolismo , Mesodermo/patología , Neoplasias/genética , Neoplasias/patología , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/enzimología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteómica , Proteínas Proto-Oncogénicas B-raf/genética , Reproducibilidad de los Resultados , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética
4.
Clin Proteomics ; 15: 13, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29541007

RESUMEN

BACKGROUND: MAP kinase inhibitor (MAPKi) therapy for BRAF mutated melanoma is characterized by high response rates but development of drug resistance within a median progression-free survival (PFS) of 9-12 months. Understanding mechanisms of resistance and identifying effective therapeutic alternatives is one of the most important scientific challenges in melanoma. Using proteomics, we want to specifically gain insight into the pathophysiological process of cerebral metastases. METHODS: Cerebral metastases from melanoma patients were initially analyzed by a LC-MS shotgun approach performed on a QExactive HF hybrid quadrupole-orbitrap mass spectrometer. For further validation steps after bioinformatics analysis, a targeted LC-QQQ-MS approach, as well as Western blot, immunohistochemistry and immunocytochemistry was performed. RESULTS: In this pilot study, we were able to identify 5977 proteins by LC-MS analysis (data are available via ProteomeXchange with identifier PXD007592). Based on PFS, samples were classified into good responders (PFS ≥ 6 months) and poor responders (PFS [Formula: see text] 3 months). By evaluating these proteomic profiles according to gene ontology (GO) terms, KEGG pathways and gene set enrichment analysis (GSEA), we could characterize differences between the two distinct groups. We detected an EMT feature (up-regulation of N-cadherin) as classifier between the two groups, V-type proton ATPases, cell adhesion proteins and several transporter and exchanger proteins to be significantly up-regulated in poor responding patients, whereas good responders showed an immune activation, among other features. We identified class-discriminating proteins based on nearest shrunken centroids, validated and quantified this signature by a targeted approach and could correlate parts of this signature with resistance using the CPL/MUW proteome database and survival of patients by TCGA analysis. We further validated an EMT-like signature as a major discriminator between good and poor responders on primary melanoma cells derived from cerebral metastases. Higher immune activity is demonstrated in patients with good response to MAPKi by immunohistochemical staining of biopsy samples of cerebral melanoma metastases. CONCLUSIONS: Employing proteomic analysis, we confirmed known extra-cerebral resistance mechanisms in the cerebral metastases and further discovered possible brain specific mechanisms of drug efflux, which might serve as treatment targets or as predictive markers for these kinds of metastasis.

5.
Curr Opin Oncol ; 28(2): 172-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26742019

RESUMEN

PURPOSE OF REVIEW: BRAF inhibitors achieve outstanding clinical response rates in BRAF-mutated melanoma patients but therapeutic resistance is common. Although combinatorial targeted therapy has recently improved patient survival, resistance still occurs, which might be because of the plasticity and heterogeneity of melanoma. Proteomics complements the mostly genomics-based approaches used so far to gain additional insights into the pathophysiological mechanisms driving melanoma progression under treatment. RECENT FINDINGS: Few proteomics studies have investigated mitogen-activated protein kinase inhibitor (MAPKi) resistance. Three technologies have been described: shotgun analysis, pressure cycling technology-sequential window acquisition of all theoretical masses (which offers an optimized protein extraction by the pressure cycling technology), and selected reaction monitoring for selected candidate evaluation. Preliminary data demonstrate that BRAFi resistance might be associated with enhanced expression of the lysosomal compartment, cell adhesion, and epithelial-mesenchymal transformation. Melanoma cells change their phenotypes in response to targeted therapy with MAPKi from a proliferative to an invasive state gaining epithelial-mesenchymal transformation features, which are associated with drug resistance. SUMMARY: Performing proteomics may lead to an enhanced understanding of the underlying mechanisms of MAPKi resistance and might offer new insights for rational therapies. Selected reaction monitoring can be used to evaluate predictive or pharmacodynamic biomarkers for tracking therapeutic responses and identifying early features of resistance.


Asunto(s)
Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/fisiología , Melanoma/tratamiento farmacológico , Proteínas Quinasas Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Neoplasias Cutáneas/tratamiento farmacológico , Anticuerpos Monoclonales/uso terapéutico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Humanos , Indoles/uso terapéutico , Ipilimumab , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Espectrometría de Masas , Melanoma/patología , Proteómica/métodos , Neoplasias Cutáneas/patología , Sulfonamidas/uso terapéutico , Vemurafenib
6.
Clin Cancer Res ; 30(1): 159-175, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-37861398

RESUMEN

PURPOSE: Despite high clinical need, there are no biomarkers that accurately predict the response of patients with metastatic melanoma to anti-PD-1 therapy. EXPERIMENTAL DESIGN: In this multicenter study, we applied protein depletion and enrichment methods prior to various proteomic techniques to analyze a serum discovery cohort (n = 56) and three independent serum validation cohorts (n = 80, n = 12, n = 17). Further validation analyses by literature and survival analysis followed. RESULTS: We identified several significantly regulated proteins as well as biological processes such as neutrophil degranulation, cell-substrate adhesion, and extracellular matrix organization. Analysis of the three independent serum validation cohorts confirmed the significant differences between responders (R) and nonresponders (NR) observed in the initial discovery cohort. In addition, literature-based validation highlighted 30 markers overlapping with previously published signatures. Survival analysis using the TCGA database showed that overexpression of 17 of the markers we identified correlated with lower overall survival in patients with melanoma. CONCLUSIONS: Ultimately, this multilayered serum analysis led to a potential marker signature with 10 key markers significantly altered in at least two independent serum cohorts: CRP, LYVE1, SAA2, C1RL, CFHR3, LBP, LDHB, S100A8, S100A9, and SAA1, which will serve as the basis for further investigation. In addition to patient serum, we analyzed primary melanoma tumor cells from NR and found a potential marker signature with four key markers: LAMC1, PXDN, SERPINE1, and VCAN.


Asunto(s)
Melanoma , Humanos , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/metabolismo , Proteómica , Biomarcadores de Tumor/metabolismo , Análisis de Supervivencia
7.
Cancer Res ; 83(7): 1128-1146, 2023 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-36946761

RESUMEN

Clinical management of melanomas with NRAS mutations is challenging. Targeting MAPK signaling is only beneficial to a small subset of patients due to resistance that arises through genetic, transcriptional, and metabolic adaptation. Identification of targetable vulnerabilities in NRAS-mutated melanoma could help improve patient treatment. Here, we used multiomics analyses to reveal that NRAS-mutated melanoma cells adopt a mesenchymal phenotype with a quiescent metabolic program to resist cellular stress induced by MEK inhibition. The metabolic alterations elevated baseline reactive oxygen species (ROS) levels, leading these cells to become highly sensitive to ROS induction. In vivo xenograft experiments and single-cell RNA sequencing demonstrated that intratumor heterogeneity necessitates the combination of a ROS inducer and a MEK inhibitor to inhibit both tumor growth and metastasis. Ex vivo pharmacoscopy of 62 human metastatic melanomas confirmed that MEK inhibitor-resistant tumors significantly benefited from the combination therapy. Finally, oxidative stress response and translational suppression corresponded with ROS-inducer sensitivity in 486 cancer cell lines, independent of cancer type. These findings link transcriptional plasticity to a metabolic phenotype that can be inhibited by ROS inducers in melanoma and other cancers. SIGNIFICANCE: Metabolic reprogramming in drug-resistant NRAS-mutated melanoma cells confers sensitivity to ROS induction, which suppresses tumor growth and metastasis in combination with MAPK pathway inhibitors.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Humanos , Especies Reactivas de Oxígeno , Proteínas Proto-Oncogénicas B-raf/genética , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Neoplasias Cutáneas/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Línea Celular Tumoral , Mutación , Proteínas de la Membrana/genética , GTP Fosfohidrolasas/genética
8.
Cancer Res ; 81(23): 6044-6057, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34645608

RESUMEN

CD271 (NGFR) is a neurotrophin receptor that belongs to the tumor necrosis receptor (TNFR) family. Upon ligand binding, CD271 can mediate either survival or cell death. Although the role of CD271 as a marker of tumor-initiating cells is still a matter of debate, its role in melanoma progression has been well documented. Moreover, CD271 has been shown to be upregulated after exposure to both chemotherapy and targeted therapy. In this study, we demonstrate that activation of CD271 by a short ß-amyloid-derived peptide (Aß(25-35)) in combination with either chemotherapy or MAPK inhibitors induces apoptosis in 2D and 3D cultures of eight melanoma cell lines. This combinatorial treatment significantly reduced metastasis in a zebrafish xenograft model and led to significantly decreased tumor volume in mice. Administration of Aß(25-35) in ex vivo tumors from immunotherapy- and targeted therapy-resistant patients significantly reduced proliferation of melanoma cells, showing that activation of CD271 can overcome drug resistance. Aß(25-35) was specific to CD271-expressing cells and induced CD271 cleavage and phosphorylation of JNK (pJNK). The direct protein-protein interaction of pJNK with CD271 led to PARP1 cleavage, p53 and caspase activation, and pJNK-dependent cell death. Aß(25-35) also mediated mitochondrial reactive oxygen species (mROS) accumulation, which induced CD271 overexpression. Finally, CD271 upregulation inhibited mROS production, revealing the presence of a negative feedback loop in mROS regulation. These results indicate that targeting CD271 can activate cell death pathways to inhibit melanoma progression and potentially overcome resistance to targeted therapy. SIGNIFICANCE: The discovery of a means to specifically activate the CD271 death domain reveals unknown pathways mediated by the receptor and highlights new treatment possibilities for melanoma.


Asunto(s)
Péptidos beta-Amiloides/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Melanoma/tratamiento farmacológico , Terapia Molecular Dirigida , Proteínas del Tejido Nervioso/agonistas , Receptores de Factor de Crecimiento Nervioso/agonistas , Animales , Apoptosis , Proliferación Celular , Quimioterapia Combinada , Femenino , Humanos , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Desnudos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
9.
Elife ; 102021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33438577

RESUMEN

The microphthalmia-associated transcription factor (MITF) is a critical regulator of melanocyte development and differentiation. It also plays an important role in melanoma where it has been described as a molecular rheostat that, depending on activity levels, allows reversible switching between different cellular states. Here, we show that MITF directly represses the expression of genes associated with the extracellular matrix (ECM) and focal adhesion pathways in human melanoma cells as well as of regulators of epithelial-to-mesenchymal transition (EMT) such as CDH2, thus affecting cell morphology and cell-matrix interactions. Importantly, we show that these effects of MITF are reversible, as expected from the rheostat model. The number of focal adhesion points increased upon MITF knockdown, a feature observed in drug-resistant melanomas. Cells lacking MITF are similar to the cells of minimal residual disease observed in both human and zebrafish melanomas. Our results suggest that MITF plays a critical role as a repressor of gene expression and is actively involved in shaping the microenvironment of melanoma cells in a cell-autonomous manner.


Asunto(s)
Transición Epitelial-Mesenquimal , Matriz Extracelular/metabolismo , Adhesiones Focales/metabolismo , Factor de Transcripción Asociado a Microftalmía/genética , Línea Celular Tumoral , Humanos , Melanoma/metabolismo , Factor de Transcripción Asociado a Microftalmía/metabolismo
11.
Virol J ; 7: 175, 2010 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-20670430

RESUMEN

BACKGROUND: Human adenoviruses (Ads) have substantial potential for clinical applications in cancer patients. Conditionally replicating adenoviruses (CRAds) include oncolytic adenoviruses in which expression of the immediate early viral transactivator protein E1A is controlled by a cancer cell-selective promoter. To enhance efficacy, CRAds are further armed to contain therapeutic genes. Due to size constraints of the capsid geometry, the capacity for packaging transgenes into Ads is, however, limited. To overcome this limitation, the employment of E1A-deleted replication-deficient viruses carrying therapeutic genes in combination with replication-competent CRAd vectors expressing E1A in trans has been proposed. Most trans-complementing studies involved transgene expressions from strong ubiquitous promoters, and thereby relied entirely on the cancer cell specificity of the CRAd vector. RESULTS: Here we tested the trans-complementation of a CRAd and a replication-deficient transgene vector containing the same cancer cell-selective promoter. Hereto, we generated two new vectors expressing IL-2 and CD40L from a bicistronic expression cassette under the control of the melanoma/melanocyte-specific tyrosinase enhancer tyrosinase promoter (TETP), which we previously described for the melanoma-specific CRAd vector AdDeltaEP-TETP. These vectors gave rise to tightly controlled melanoma-specific transgene expression levels, which were only 5 to 40-fold lower than those from vectors controlled by the nonselective CMV promoter. Reporter analyses using Ad-CMV-eGFP in combination with AdDeltaEP-TETP revealed a high level of trans-complementation in melanoma cells (up to about 30-fold), but not in non-melanoma cells, unlike the AdCMV-eGFP/wtAd5 binary vector system, which was equally efficient in melanoma and non-melanoma cells. Similar findings were obtained when replacing the transgene vector AdCMV-eGFP with AdCMV-IL-2 or AdCMV-CD40L. However, the combination of the novel AdTETP-CD40L/IL-2 vector with AdDeltaEP-TETP or wtAd5 gave reproducible moderate 3-fold enhancements of IL-2 by trans-complementation only. CONCLUSIONS: The cancer cell-selective TETP tested here did not give the expected enforceable transgene expression typically achieved in the Ad trans-complementing system. Reasons for this could include virus-mediated down regulation of limiting transcription factors, and/or competition for such factors by different promoters. Whether this finding is unique to the particular promoter system tested here, or also occurs with other promoters warrants further investigations.


Asunto(s)
Adenovirus Humanos/genética , Técnicas de Transferencia de Gen/instrumentación , Vectores Genéticos/genética , Melanoma/genética , Monofenol Monooxigenasa/genética , Regiones Promotoras Genéticas , Activación Transcripcional , Adenovirus Humanos/fisiología , Animales , Ligando de CD40/genética , Ligando de CD40/metabolismo , Línea Celular Tumoral , Terapia Genética/instrumentación , Vectores Genéticos/fisiología , Humanos , Interleucina-2/genética , Interleucina-2/metabolismo , Melanoma/enzimología , Ratones , Replicación Viral
12.
Clin Cancer Res ; 25(22): 6852-6867, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31375515

RESUMEN

PURPOSE: Treatment of BRAFV600E -mutant melanomas with MAPK inhibitors (MAPKi) results in significant tumor regression, but acquired resistance is pervasive. To understand nonmutational mechanisms underlying the adaptation to MAPKi and to identify novel vulnerabilities of melanomas treated with MAPKi, we focused on the initial response phase during treatment with MAPKi. EXPERIMENTAL DESIGN: By screening proteins expressed on the cell surface of melanoma cells, we identified the fatty acid transporter CD36 as the most consistently upregulated protein upon short-term treatment with MAPKi. We further investigated the effects of MAPKi on fatty acid metabolism using in vitro and in vivo models and analyzing patients' pre- and on-treatment tumor specimens. RESULTS: Melanoma cells treated with MAPKi displayed increased levels of CD36 and of PPARα-mediated and carnitine palmitoyltransferase 1A (CPT1A)-dependent fatty acid oxidation (FAO). While CD36 is a useful marker of melanoma cells during adaptation and drug-tolerant phases, the upregulation of CD36 is not functionally involved in FAO changes that characterize MAPKi-treated cells. Increased FAO is required for BRAFV600E -mutant melanoma cells to survive under the MAPKi-induced metabolic stress prior to acquiring drug resistance. The upfront and concomitant inhibition of FAO, glycolysis, and MAPK synergistically inhibits tumor cell growth in vitro and in vivo. CONCLUSIONS: Thus, we identified a clinically relevant therapeutic approach that has the potential to improve initial responses and to delay acquired drug resistance of BRAFV600E -mutant melanoma.


Asunto(s)
Adaptación Biológica , Ácidos Grasos/metabolismo , Melanoma/genética , Melanoma/metabolismo , Mutación , Oxidación-Reducción , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/genética , Alelos , Animales , Biomarcadores , Antígenos CD36/genética , Antígenos CD36/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genotipo , Glucólisis , Humanos , Inmunofenotipificación , Melanoma/patología , Ratones , Modelos Biológicos , Estadificación de Neoplasias , PPAR alfa/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Oncotarget ; 7(47): 77163-77174, 2016 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-27791198

RESUMEN

Acquired chemotherapeutic resistance of cancer cells can result from a Darwinistic evolution process in which heterogeneity plays an important role. In order to understand the impact of genetic heterogeneity on acquired resistance and second line therapy selection in metastatic melanoma, we sequenced the exomes of 27 lesions which were collected from 3 metastatic melanoma patients treated with targeted or non-targeted inhibitors. Furthermore, we tested the impact of a second NRAS mutation in 7 BRAF inhibitor resistant early passage cell cultures on the selection of second line therapies.We observed a rapid monophyletic evolution of melanoma subpopulations in response to targeted therapy that was not observed in non-targeted therapy. We observed the acquisition of NRAS mutations in the BRAF mutated patient treated with a BRAF inhibitor in 1 of 5 of his post-resistant samples. In an additional cohort of 5 BRAF-inhibitor treated patients we detected 7 NRAS mutations in 18 post-resistant samples. No NRAS mutations were detected in pre-resistant samples. By sequencing 65 single cell clones we prove that NRAS mutations co-occur with BRAF mutations in single cells. The double mutated cells revealed a heterogeneous response to MEK, ERK, PI3K, AKT and multi RTK - inhibitors.We conclude that BRAF and NRAS co-mutations are not mutually exclusive. However, the sole finding of double mutated cells in a resistant tumor is not sufficient to determine follow-up therapy. In order to target the large pool of heterogeneous cells in a patient, we think combinational therapy targeting different pathways will be necessary.


Asunto(s)
Resistencia a Antineoplásicos , GTP Fosfohidrolasas/genética , Melanoma/genética , Proteínas de la Membrana/genética , Mutación , Proteínas Proto-Oncogénicas B-raf/genética , Línea Celular Tumoral , Evolución Molecular , Femenino , Humanos , Masculino , Melanoma/tratamiento farmacológico , Metástasis de la Neoplasia , Inhibidores de Proteínas Quinasas/uso terapéutico , Análisis de la Célula Individual , Secuenciación del Exoma
14.
Genome Biol ; 16: 42, 2015 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-25885555

RESUMEN

BACKGROUND: Melanoma is the most fatal skin cancer displaying a high degree of molecular heterogeneity. Phenotype switching is a mechanism that contributes to melanoma heterogeneity by altering transcription profiles for the transition between states of proliferation/differentiation and invasion/stemness. As phenotype switching is reversible, epigenetic mechanisms, like DNA methylation, could contribute to the changes in gene expression. RESULTS: Integrative analysis of methylation and gene expression datasets of five proliferative and five invasion melanoma cell cultures reveal two distinct clusters. SOX9 is methylated and lowly expressed in the highly proliferative group. SOX9 overexpression results in decreased proliferation but increased invasion in vitro. In a B16 mouse model, sox9 overexpression increases the number of lung metastases. Transcriptional analysis of SOX9-overexpressing melanoma cells reveals enrichment in epithelial to mesenchymal transition (EMT) pathways. Survival analysis of The Cancer Genome Atlas melanoma dataset shows that metastatic patients with high expression levels of SOX9 have significantly worse survival rates. Additional survival analysis on the targets of SOX9 reveals that most SOX9 downregulated genes have survival benefit for metastatic patients. CONCLUSIONS: Our genome-wide DNA methylation and gene expression study of 10 early passage melanoma cell cultures reveals two phenotypically distinct groups. One of the genes regulated by DNA methylation between the two groups is SOX9. SOX9 induces melanoma cell invasion and metastasis and decreases patient survival. A number of genes downregulated by SOX9 have a negative impact on patient survival. In conclusion, SOX9 is an important gene involved in melanoma invasion and negatively impacts melanoma patient survival.


Asunto(s)
Melanoma/genética , Invasividad Neoplásica/genética , Factor de Transcripción SOX9/biosíntesis , Neoplasias Cutáneas/genética , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Metilación de ADN/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Melanoma/patología , Ratones , Persona de Mediana Edad , Invasividad Neoplásica/patología , Estadificación de Neoplasias , Pronóstico , Factor de Transcripción SOX9/genética , Transducción de Señal , Neoplasias Cutáneas/patología , Análisis de Supervivencia
15.
J Invest Dermatol ; 133(2): 316-24, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22971848

RESUMEN

Recently, Biedermann et al. (2010) have demonstrated that human eccrine sweat gland cells can develop a multilayered epidermis. The question still remains whether these cells can fulfill exclusive and very specific functional properties of epidermal keratinocytes, such as the incorporation of melanin, a feature absent in sweat gland cells. We added human melanocytes to eccrine sweat gland cells to let them develop into an epidermal analog in vivo. The interaction between melanocytes and sweat gland-derived keratinocytes was investigated. The following results were gained: (1) macroscopically, a pigmentation of the substitutes was seen 2-3 weeks after transplantation; (2) we confirmed the development of a multilayered, stratified epidermis with melanocytes distributed evenly throughout the basal layer; (3) melanocytic dendrites projected to suprabasal layers; and (4) melanin was observed to be integrated into former eccrine sweat gland cells. These skin substitutes were similar or equal to skin substitutes cultured from human epidermal keratinocytes. The only differences observed were a delay in pigmentation and less melanin uptake. These data suggest that eccrine sweat gland cells can form a functional epidermal melanin unit, thereby providing striking evidence that they can assume one of the most characteristic keratinocyte properties.


Asunto(s)
Queratinocitos/citología , Melaninas/metabolismo , Melanocitos/citología , Trasplante de Piel/métodos , Piel Artificial , Glándulas Sudoríparas/citología , Adolescente , Animales , Células Cultivadas , Niño , Preescolar , Dermis/citología , Dermis/metabolismo , Células Epidérmicas , Epidermis/metabolismo , Femenino , Humanos , Lactante , Queratinocitos/metabolismo , Masculino , Melanocitos/metabolismo , Ratones , Ratones Desnudos , Técnicas de Cultivo de Órganos , Ratas , Receptor PAR-2/metabolismo , Pigmentación de la Piel/fisiología , Glándulas Sudoríparas/metabolismo , Trasplante Heterólogo
16.
J Invest Dermatol ; 133(10): 2436-2443, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23474946

RESUMEN

We have previously reported a model for melanoma progression in which oscillation between melanoma cell phenotypes characterized by invasion or proliferation is fundamental to tumor heterogeneity and disease progression. In this study we examine the possible role of hypoxia as one of the microenvironmental influences driving metastatic progression by promoting a switch from a proliferative to an invasive phenotype. Immunohistochemistry on primary human cutaneous melanoma biopsies showed intratumoral heterogeneity for cells expressing melanocytic markers, and a loss of these markers correlated with hypoxic regions. Furthermore, we show that the downregulation of melanocytic markers is dependent on hypoxia inducible factor 1α (HIF1α), a known regulator of the hypoxic response. In vitro invasion assays showed that a hypoxic environment increases the invasiveness of proliferative melanoma cell cultures in a HIF1α-dependent manner. In contrast, invasive phenotype melanoma cells showed no increase in invasive potential upon exposure to hypoxia. Thus, exposure of proliferative melanoma cells to hypoxic microenvironments is sufficient, in a HIF1α-dependent manner, to downregulate melanocytic marker expression and increase their invasive potential.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia/patología , Melanoma/patología , Neoplasias Cutáneas/patología , Microambiente Tumoral/fisiología , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Heterogeneidad Genética , Humanos , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Antígeno MART-1/genética , Antígeno MART-1/metabolismo , Melanoma/genética , Melanoma/metabolismo , Invasividad Neoplásica , Fenotipo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Células Tumorales Cultivadas
17.
Pigment Cell Melanoma Res ; 25(3): 343-53, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22336146

RESUMEN

There is growing evidence that the metastatic spread of melanoma is driven not by a linear increase in tumorigenic aggressiveness, but rather by switching back and forth between two different phenotypes of metastatic potential. In vitro these phenotypes are respectively defined by the characteristics of strong proliferation/weak invasiveness and weak proliferation/strong invasiveness. Melanoma cell phenotype is tightly linked to gene expression. Taking advantage of this, we have developed a gene expression-based tool for predicting phenotype called Heuristic Online Phenotype Prediction. We demonstrate the predictive utility of this tool by comparing phenotype-specific signatures with measurements of characteristics of melanoma phenotype-specific biology in different melanoma cell lines and short-term cultures. We further show that 86% of 536 tested melanoma lines and short-term cultures are significantly associated with the phenotypes we describe. These findings reinforce the concept that a two-state system, as described by the phenotype switching model, underlies melanoma progression.


Asunto(s)
Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Perfilación de la Expresión Génica , Melanoma/clasificación , Neoplasias Cutáneas/clasificación , Proliferación Celular , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Genes Relacionados con las Neoplasias/genética , Estudios de Asociación Genética , Humanos , Melanoma/genética , Melanoma/patología , Análisis por Micromatrices , Invasividad Neoplásica , Fenotipo , Reproducibilidad de los Resultados , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Distribución Tisular , Células Tumorales Cultivadas/clasificación
18.
Pigment Cell Melanoma Res ; 24(4): 631-42, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21599871

RESUMEN

Recent observations suggest that melanoma cells drive disease progression by switching back and forth between phenotypic states of proliferation and invasion. Phenotype switching has been linked to changes in Wnt signalling, and we therefore looked for cell phenotype-specific differences in the levels and activity of ß-catenin and its LEF/TCF co-factors. We found that while cytosolic ß-catenin distribution is phenotype-specific (membrane-associated in proliferative cells and cytosolic in invasive cells), its nuclear distribution and activity is not. Instead, the expression patterns of two ß-catenin co-factors, LEF1 and TCF4, are both phenotype-specific and inversely correlated. LEF1 is preferentially expressed by differentiated/proliferative phenotype cells and TCF4 by dedifferentiated/invasive phenotype cells. Knock-down experiments confirmed that these co-factors are important for the phenotype-specific expression of M-MITF, WNT5A and other genes and that LEF1 suppresses TCF4 expression independently of ß-catenin. Our data show that melanoma cell phenotype switching behaviour is regulated by differential LEF1/TCF4 activity.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Melanoma/genética , Melanoma/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Factores de Transcripción/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Proliferación Celular/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Modelos Biológicos , Invasividad Neoplásica , Fenotipo , Factor de Transcripción 4 , Factores de Transcripción/metabolismo , Proteínas Wnt/farmacología , beta Catenina/metabolismo
19.
Pigment Cell Melanoma Res ; 24(2): 326-33, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21176117

RESUMEN

Oncogenic mutations within the MAPK pathway are frequent in melanoma, and targeting of MAPK signaling has yielded spectacular responses in a significant number of patients that last for several months before relapsing. We investigated the effects of two different inhibitors of MAPK signaling in proliferative and invasive melanoma cell cultures with various mutations in the MAPK pathway. Proliferative melanoma cells were more susceptible to pathway inhibition than invasive phenotype cells, irrespective of BRAF mutation status, while invasive phenotype cell response was dependent on BRAF mutation status. Critically, MAPK pathway inhibition of proliferative phenotype cells resulted in acquisition of invasive phenotype characteristics. These results show that melanoma cell phenotype is an important factor in MAPK pathway inhibition response. This suggests that while current therapeutic strategies target proliferative melanoma cells, future approaches should also account for the invasive phenotype population.


Asunto(s)
Melanoma/genética , Melanoma/patología , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Fenotipo , Proteínas Proto-Oncogénicas B-raf/genética , Quinasas raf/antagonistas & inhibidores , Adulto , Anciano , Proliferación Celular , Inhibidores Enzimáticos/uso terapéutico , Femenino , Perfilación de la Expresión Génica , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Análisis por Micromatrices , Persona de Mediana Edad , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación , Invasividad Neoplásica , Proteínas Proto-Oncogénicas B-raf/metabolismo
20.
Melanoma Res ; 20(4): 349-55, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20526217

RESUMEN

To date there is no effective therapy for metastatic melanoma and at the molecular level the disease progression is poorly understood. A recent study by our group led to the development of a novel phenotype switching model for melanoma progression, wherein cells transition back-and-forth between states of proliferation and invasion to drive disease progression. To explore the model's clinical relevance we interrogated phenotype-specific expression patterns in human melanoma patient material. A matched primary/metastasis pair from a human melanoma patient was obtained and immunohistochemically stained for proliferative and invasive phenotype markers. These were also stained for hypoxia and blood vessel markers. Proliferative phenotype markers Melan-A and Mitf showed consistent anti-correlation with invasive phenotype marker Wnt5A and hypoxia marker Glut-1. These also correlated with observed intra-tumoural vascularization patterns. Similar pattern distributions were present in both primary and metastasis samples. Strikingly, we observed that late phase metastatic melanoma cells adopt morphologies and behaviours identical to very early phase cells. The expression patterns observed closely matched expectations derived from previous in vitro and xenografting experiments. These results highlight the likelihood that disease progression involves melanoma cells retaining the capacity to regulate the expression of metastatic potential critical factors according to changing microenvironmental conditions.


Asunto(s)
Melanoma/patología , Neoplasias Cutáneas/patología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Progresión de la Enfermedad , Femenino , Neoplasias de la Vesícula Biliar/genética , Neoplasias de la Vesícula Biliar/metabolismo , Neoplasias de la Vesícula Biliar/patología , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Antígeno MART-1 , Melanoma/genética , Melanoma/metabolismo , Melanoma/secundario , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA