Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Invest Dermatol ; 144(3): 601-611, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37739336

RESUMEN

Premature hair graying occurs owing to the depletion of melanocyte stem cells in the hair follicle, which can be accelerated by stress caused by genetic or environmental factors. However, the connection between stress and melanocyte stem cell loss is not fully understood. MicroRNAs are molecules that control gene expression by regulating mRNA stability and translation and are produced by the enzyme Dicer, which is repressed under stress. In this study, using 2 mouse genetic models and human and mouse cell lines, we found that the inactivation of Dicer in melanocytes leads to misplacement of these cells within the hair follicle, resulting in a lack of melanin transfer to keratinocytes in the growing hair and the exhaustion of the melanocyte stem cell pool. We also show that miR-92b, which regulates ItgaV mRNA and protein levels, plays a role in altering melanocyte migration. Overall, our findings suggest that the Dicer-miR92b-ItgaV pathway serves as a major signaling pathway linking stress to premature hair greying.


Asunto(s)
Color del Cabello , Melanocitos , Ratones , Humanos , Animales , Color del Cabello/genética , Melanocitos/metabolismo , Melaninas/metabolismo , Cabello , Folículo Piloso
2.
J Invest Dermatol ; 143(4): 538-544.e2, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36958885

RESUMEN

The establishment of consistent genetically modified mouse melanoma models and cell lines is of paramount importance for prevention and treatment. In this study, we review the different mouse melanoma cell lines that have been established. After careful molecular characterization of the established mouse melanoma cell lines, modification of the genome, microenvironment, or even the environment using appropriate in cellulo and in vivo assays may reveal novel genetic and nongenetic changes. These murine melanoma cell lines with defined genetic mutations allow the testing of innovative therapies based on chemistry, physics, and biology using alternative methods. In addition to the fundamental aspects, these results are important for humans because of the relevance of these murine melanoma cell lines to human disease.


Asunto(s)
Melanoma , Humanos , Ratones , Animales , Línea Celular Tumoral , Melanoma/genética , Modelos Animales de Enfermedad , Microambiente Tumoral/genética
3.
Cancers (Basel) ; 14(3)2022 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-35158973

RESUMEN

G-protein-coupled receptors (GPCRs) serve prominent roles in melanocyte lineage physiology, with an impact at all stages of development, as well as on mature melanocyte functions. GPCR ligands are present in the skin and regulate melanocyte homeostasis, including pigmentation. The role of GPCRs in the regulation of pigmentation and, consequently, protection against external aggression, such as ultraviolet radiation, has long been established. However, evidence of new functions of GPCRs directly in melanomagenesis has been highlighted in recent years. GPCRs are coupled, through their intracellular domains, to heterotrimeric G-proteins, which induce cellular signaling through various pathways. Such signaling modulates numerous essential cellular processes that occur during melanomagenesis, including proliferation and migration. GPCR-associated signaling in melanoma can be activated by the binding of paracrine factors to their receptors or directly by activating mutations. In this review, we present melanoma-associated alterations of GPCRs and their downstream signaling and discuss the various preclinical models used to evaluate new therapeutic approaches against GPCR activity in melanoma. Recent striking advances in our understanding of the structure, function, and regulation of GPCRs will undoubtedly broaden melanoma treatment options in the future.

4.
Development ; 149(2)2022 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-34878101

RESUMEN

The canonical Wnt/ß-catenin pathway governs a multitude of developmental processes in various cell lineages, including the melanocyte lineage. Indeed, ß-catenin regulates transcription of Mitf-M, the master regulator of this lineage. The first wave of melanocytes to colonize the skin is directly derived from neural crest cells, whereas the second wave of melanocytes is derived from Schwann cell precursors (SCPs). We investigated the influence of ß-catenin in the development of melanocytes of the first and second waves by generating mice expressing a constitutively active form of ß-catenin in cells expressing tyrosinase. Constitutive activation of ß-catenin did not affect the development of truncal melanoblasts but led to marked hyperpigmentation of the paws. By activating ß-catenin at various stages of development (E8.5-E11.5), we showed that the activation of ß-catenin in bipotent SCPs favored melanoblast specification at the expense of Schwann cells in the limbs within a specific temporal window. Furthermore, in vitro hyperactivation of the Wnt/ß-catenin pathway, which is required for melanocyte development, induces activation of Mitf-M, in turn repressing FoxD3 expression. In conclusion, ß-catenin overexpression promotes SCP cell fate decisions towards the melanocyte lineage.


Asunto(s)
Diferenciación Celular , Melanocitos/metabolismo , Células de Schwann/citología , beta Catenina/metabolismo , Animales , Línea Celular Tumoral , Linaje de la Célula , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Melanocitos/citología , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción Asociado a Microftalmía/genética , Factor de Transcripción Asociado a Microftalmía/metabolismo , Estabilidad Proteica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Células de Schwann/metabolismo , Vía de Señalización Wnt , beta Catenina/genética
5.
Nat Commun ; 12(1): 3707, 2021 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-34140478

RESUMEN

While the major drivers of melanoma initiation, including activation of NRAS/BRAF and loss of PTEN or CDKN2A, have been identified, the role of key transcription factors that impose altered transcriptional states in response to deregulated signaling is not well understood. The POU domain transcription factor BRN2 is a key regulator of melanoma invasion, yet its role in melanoma initiation remains unknown. Here, in a BrafV600E PtenF/+ context, we show that BRN2 haplo-insufficiency promotes melanoma initiation and metastasis. However, metastatic colonization is less efficient in the absence of Brn2. Mechanistically, BRN2 directly induces PTEN expression and in consequence represses PI3K signaling. Moreover, MITF, a BRN2 target, represses PTEN transcription. Collectively, our results suggest that on a PTEN heterozygous background somatic deletion of one BRN2 allele and temporal regulation of the other allele elicits melanoma initiation and progression.


Asunto(s)
Carcinogénesis/metabolismo , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Genes Supresores de Tumor , Proteínas de Homeodominio/metabolismo , Melanoma/metabolismo , Factores del Dominio POU/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Estudios de Cohortes , Variaciones en el Número de Copia de ADN , Progresión de la Enfermedad , Técnicas de Silenciamiento del Gen , Haploinsuficiencia , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Melanoma/genética , Melanoma/mortalidad , Melanoma/secundario , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Análisis por Micromatrices , Factor de Transcripción Asociado a Microftalmía/metabolismo , Mutación , Factores del Dominio POU/genética , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , ARN Interferente Pequeño , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/secundario , Melanoma Cutáneo Maligno
6.
Commun Biol ; 4(1): 423, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33772156

RESUMEN

Skin pigmentation is dependent on cellular processes including melanosome biogenesis, transport, maturation and transfer to keratinocytes. However, how the cells finely control these processes in space and time to ensure proper pigmentation remains unclear. Here, we show that a component of the cytoplasmic dynein complex, Dynlt3, is required for efficient melanosome transport, acidity and transfer. In Mus musculus melanocytes with decreased levels of Dynlt3, pigmented melanosomes undergo a more directional motion, leading to their peripheral location in the cell. Stage IV melanosomes are more acidic, but still heavily pigmented, resulting in a less efficient melanosome transfer. Finally, the level of Dynlt3 is dependent on ß-catenin activity, revealing a function of the Wnt/ß-catenin signalling pathway during melanocyte and skin pigmentation, by coupling the transport, positioning and acidity of melanosomes required for their transfer.


Asunto(s)
Dineínas/genética , Melanocitos/metabolismo , Melanosomas/fisiología , Animales , Dineínas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Pigmentación de la Piel
8.
Pigment Cell Melanoma Res ; 33(3): 426-434, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31679174

RESUMEN

The Cre/loxP system is a powerful tool that has allowed the study of the effects of specific genes of interest in various biological settings. The Tyr::CreERT2 system allows for the targeted expression and activity of the Cre enzyme in the melanocyte lineage following treatment with tamoxifen, thus providing spatial and temporal control of the expression of specific target genes. Two independent transgenic mouse models, each containing a Tyr::CreERT2 transgene, have been generated and are widely used to study melanocyte transformation. In this study, we performed whole genome sequencing (WGS) on genomic DNA from the two Tyr::CreERT2 mouse models and identified their sites of integration in the C57BL/6 genome. Based on these results, we designed PCR primers to accurately, and efficiently, genotype transgenic mice. Finally, we discussed some of the advantages of each transgenic mouse model.


Asunto(s)
Integrasas/metabolismo , Análisis de Secuencia de ADN , Transgenes , Animales , Cruzamientos Genéticos , Femenino , Sitios Genéticos , Genoma , Genotipo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos
9.
Development ; 145(12)2018 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-29769218

RESUMEN

To distribute and establish the melanocyte lineage throughout the skin and other developing organs, melanoblasts undergo several rounds of proliferation, accompanied by migration through complex environments and differentiation. Melanoblast migration requires interaction with extracellular matrix of the epidermal basement membrane and with surrounding keratinocytes in the developing skin. Migration has been characterized by measuring speed, trajectory and directionality of movement, but there are many unanswered questions about what motivates and defines melanoblast migration. Here, we have established a general mathematical model to simulate the movement of melanoblasts in the epidermis based on biological data, assumptions and hypotheses. Comparisons between experimental data and computer simulations reinforce some biological assumptions, and suggest new ideas for how melanoblasts and keratinocytes might influence each other during development. For example, it appears that melanoblasts instruct each other to allow a homogeneous distribution in the tissue and that keratinocytes may attract melanoblasts until one is stably attached to them. Our model reveals new features of how melanoblasts move and, in particular, suggest that melanoblasts leave a repulsive trail behind them as they move through the skin.


Asunto(s)
Movimiento Celular/fisiología , Simulación por Computador , Queratinocitos/metabolismo , Melanocitos/citología , Piel/embriología , Animales , Membrana Basal/metabolismo , Adhesión Celular/fisiología , Matriz Extracelular/metabolismo , Melanocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Teóricos
10.
Pigment Cell Melanoma Res ; 31(3): 423-431, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29224244

RESUMEN

Genetically engineered mouse models offer essential opportunities to investigate the mechanisms of initiation and progression in melanoma. Here, we report a new simplified histopathology classification of mouse melanocytic lesions in Tyr::NRASQ61K derived models, using an interactive decision tree that produces homogeneous categories. Reproducibility for this classification system was evaluated on a panel of representative cases of murine melanocytic lesions by pathologists and basic scientists. Reproducibility, measured as inter-rater agreement between evaluators using a modified Fleiss' kappa statistic, revealed a very good agreement between observers. Should this new simplified classification be adopted, it would create a robust system of communication between researchers in the field of mouse melanoma models.


Asunto(s)
Melanoma , Proteínas de Unión al GTP Monoméricas , Mutación Missense , Sustitución de Aminoácidos , Animales , Melanoma/clasificación , Melanoma/genética , Melanoma/metabolismo , Melanoma/patología , Ratones , Ratones Transgénicos , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo
11.
Oncotarget ; 8(19): 32270-32291, 2017 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-28416759

RESUMEN

Plakoglobin (also known as γ-catenin) is a member of the Armadillo family of proteins and a paralog of ß-catenin. Plakoglobin is a component of both the adherens junctions and desmosomes, and therefore plays a vital role in the regulation of cell-cell adhesion. Similar to ß-catenin, plakoglobin is capable of participating in cell signaling in addition to its role in cell-cell adhesion. In this context, ß-catenin has a well-documented oncogenic potential as a component of the Wnt signaling pathway. In contrast, while some studies have suggested a tumor promoting activity of plakoglobin in a cell/malignancy specific context, it generally acts as a tumor/metastasis suppressor. How plakoglobin acts as a growth/metastasis inhibitory protein has remained, until recently, unclear. Recent evidence suggests that plakoglobin may suppress tumorigenesis and metastasis by multiple mechanisms, including the suppression of oncogenic signaling, interactions with various proteins involved in tumorigenesis and metastasis, and the regulation of the expression of genes involved in these processes. This review is primarily focused on various mechanisms by which plakoglobin may inhibit tumorigenesis and metastasis.


Asunto(s)
Adhesión Celular , Transformación Celular Neoplásica/metabolismo , Neoplasias/metabolismo , gamma Catenina/metabolismo , Uniones Adherentes/metabolismo , Animales , Adhesión Celular/genética , Transformación Celular Neoplásica/genética , Desarrollo Embrionario/genética , Regulación de la Expresión Génica , Humanos , Complejos Multiproteicos/metabolismo , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/patología , Células Neoplásicas Circulantes/metabolismo , Células Madre Neoplásicas/metabolismo , Unión Proteica , Transducción de Señal , Células Madre/metabolismo , gamma Catenina/genética
12.
Pigment Cell Melanoma Res ; 29(5): 524-40, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27311806

RESUMEN

ß-catenin is known as an Armadillo protein that regulates gene expression following WNT pathway activation. However, WNT-independent pathways also activate ß-catenin. During the establishment of the melanocyte lineage, ß-catenin plays an important role. In the context of physiopathology, ß-catenin is activated genetically or transiently in various cancers, including melanoma, where it can be found in the nucleus of tumors. In this review, we discuss alternative pathways that activate ß-catenin independent of WNTs and highlight what is known regarding these pathways in melanoma. We also discuss the role of ß-catenin as a transcriptional regulator in various cell types, with emphasis on the different transcription factors it associates with independent of WNT induction. Finally, the role of WNT-independent ß-catenin in melanocyte development and melanomagenesis is also discussed.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Melanoma/patología , Factores de Transcripción/metabolismo , beta Catenina/metabolismo , Animales , Humanos , Melanoma/metabolismo , Transducción de Señal , Vía de Señalización Wnt
13.
Nat Commun ; 6: 8093, 2015 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-26307673

RESUMEN

Loss of the tumour suppressor PTEN is frequent in human melanoma, results in MAPK activation, suppresses senescence and mediates metastatic behaviour. How PTEN loss mediates these effects is unknown. Here we show that loss of PTEN in epithelial and melanocytic cell lines induces the nuclear localization and transcriptional activation of ß-catenin independent of the PI3K-AKT-GSK3ß axis. The absence of PTEN leads to caveolin-1 (CAV1)-dependent ß-catenin transcriptional modulation in vitro, cooperates with NRAS(Q61K) to initiate melanomagenesis in vivo and induces efficient metastasis formation associated with E-cadherin internalization. The CAV1-ß-catenin axis is mediated by a feedback loop in which ß-catenin represses transcription of miR-199a-5p and miR-203, which suppress the levels of CAV1 mRNA in melanoma cells. These data reveal a mechanism by which loss of PTEN increases CAV1-mediated dissociation of ß-catenin from membranous E-cadherin, which may promote senescence bypass and metastasis.


Asunto(s)
Cadherinas/metabolismo , Caveolina 1/genética , Melanocitos/metabolismo , Melanoma/genética , Fosfohidrolasa PTEN/genética , Neoplasias Cutáneas/genética , Activación Transcripcional/genética , beta Catenina/metabolismo , Animales , Western Blotting , Línea Celular Tumoral , Retroalimentación Fisiológica , GTP Fosfohidrolasas/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Humanos , Inmunohistoquímica , Melanoma/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , MicroARNs , Microscopía Fluorescente , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Neoplasias Cutáneas/metabolismo
14.
PLoS One ; 8(11): e78388, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24260116

RESUMEN

Plakoglobin (γ-catenin) is a homolog of ß-catenin with dual adhesive and signaling functions. Plakoglobin participates in cell-cell adhesion as a component of the adherens junction and desmosomes whereas its signaling function is mediated by its interactions with various intracellular protein partners. To determine the role of plakoglobin during tumorigenesis and metastasis, we expressed plakoglobin in the human tongue squamous cell carcinoma (SCC9) cells and compared the mRNA profiles of parental SCC9 cells and their plakoglobin-expressing transfectants (SCC9-PG). We observed that the mRNA levels of SATB1, the oncogenic chromatin remodeling factor, were decreased approximately 3-fold in SCC9-PG cells compared to parental SCC9 cells. Here, we showed that plakoglobin decreased levels of SATB1 mRNA and protein in SCC9-PG cells and that plakoglobin and p53 associated with the SATB1 promoter. Plakoglobin expression also resulted in decreased SATB1 promoter activity. These results were confirmed following plakoglobin expression in the very low plakoglobin expressing and invasive mammary carcinoma cell line MDA-MB-231 cells (MDA-231-PG). In addition, knockdown of endogenous plakoglobin in the non-invasive mammary carcinoma MCF-7 cells (MCF-7-shPG) resulted in increased SATB1 mRNA and protein. Plakoglobin expression also resulted in increased mRNA and protein levels of the metastasis suppressor Nm23-H1, a SATB1 target gene. Furthermore, the levels of various SATB1 target genes involved in tumorigenesis and metastasis were altered in MCF-7-shPG cells relative to parental MCF-7 cells. Finally, plakoglobin expression resulted in decreased in vitro proliferation, migration and invasion in different carcinoma cell lines. Together with the results of our previous studies, the data suggests that plakoglobin suppresses tumorigenesis and metastasis through the regulation of genes involved in these processes.


Asunto(s)
Neoplasias de la Mama/metabolismo , Movimiento Celular , Proliferación Celular , Desmoplaquinas/biosíntesis , Regulación Neoplásica de la Expresión Génica , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Desmoplaquinas/genética , Regulación hacia Abajo , Femenino , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteínas de Neoplasias/genética , gamma Catenina
15.
J Cell Sci ; 126(Pt 14): 3031-42, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23687381

RESUMEN

Plakoglobin (γ-catenin), a constituent of the adherens junction and desmosomes, has signaling capabilities typically associated with tumor/metastasis suppression through mechanisms that remain undefined. To determine the role of plakoglobin during tumorigenesis and metastasis, we expressed plakoglobin in human tongue squamous cell carcinoma (SCC9) cells and compared the mRNA profiles of parental SCC9 cells and their plakoglobin-expressing transfectants (SCC9-PG). We detected several p53-target genes whose levels were altered upon plakoglobin expression. In this study, we identified the p53 regulated tumor suppressor 14-3-3σ as a direct plakoglobin-p53 target gene. Coimmunoprecipitation experiments revealed that plakoglobin and p53 interact, and chromatin immunoprecipitation and electrophoretic mobility shift assays revealed that plakoglobin and p53 associate with the 14-3-3σ promoter. Furthermore, luciferase reporter assays showed that p53 transcriptional activity is increased in the presence of plakoglobin. Finally, knockdown of plakoglobin in MCF-7 cells followed by luciferase assays confirmed that p53 transcriptional activity is enhanced in the presence of plakoglobin. Our data suggest that plakoglobin regulates gene expression in conjunction with p53 and that plakoglobin may regulate p53 transcriptional activity, which may account, in part, for the tumor/metastasis suppressor activity of plakoglobin.


Asunto(s)
Proteínas 14-3-3/genética , Carcinogénesis/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteína p53 Supresora de Tumor/metabolismo , gamma Catenina/metabolismo , Carcinogénesis/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Células MCF-7 , Regiones Promotoras Genéticas/genética , Unión Proteica , ARN Interferente Pequeño/genética , Activación Transcripcional/genética , Transgenes/genética , gamma Catenina/genética
16.
Int J Cell Biol ; 2012: 189521, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22481945

RESUMEN

Plakoglobin (γ-catenin) is a member of the Armadillo family of proteins and a homolog of ß-catenin. As a component of both the adherens junctions and desmosomes, plakoglobin plays a pivotal role in the regulation of cell-cell adhesion. Furthermore, similar to ß-catenin, plakoglobin is capable of participating in cell signaling. However, unlike ß-catenin that has well-documented oncogenic potential through its involvement in the Wnt signaling pathway, plakoglobin generally acts as a tumor/metastasis suppressor. The exact roles that plakoglobin plays during tumorigenesis and metastasis are not clear; however, recent evidence suggests that it may regulate gene expression, cell proliferation, apoptosis, invasion, and migration. In this paper, we describe plakoglobin, its discovery and characterization, its role in regulating cell-cell adhesion, and its signaling capabilities in regulation of tumorigenesis and metastasis.

17.
Breast Cancer Res Treat ; 121(2): 527-38, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19787450

RESUMEN

The complexity of breast cancer biology makes it challenging to analyze large datasets of clinicopathologic and molecular attributes, toward identifying the key prognostic features and producing systems capable of predicting which patients are likely to relapse. We applied machine-learning techniques to analyze a set of well-characterized primary breast cancers, which specified the abundance and localization of various junctional proteins. We hypothesized that disruption of junctional complexes would lead to the cytoplasmic/nuclear redistribution of the protein components and their potential interactions with growth-regulating molecules, which would promote relapse, and that machine-learning techniques could use the subcellular locations of these proteins, together with standard clinicopathological data, to produce an efficient prognostic classifier. We used immunohistochemistry to assess the expression and subcellular distribution of six junctional proteins, in addition to a panel of eight standard clinical features and concentrations of four "growth-regulating" proteins, to produce a database involving 36 features, over 66 primary invasive ductal breast carcinomas. A machine-learning system was applied to this clinicopathologic dataset to produce a decision-tree classifier that could predict whether a novel breast cancer patient would relapse. We show that this decision-tree classifier, which incorporates a combination of only four features (nuclear alpha- and beta-catenin levels, the total level of PTEN and the number of involved axillary lymph nodes), is able to correctly classify patient outcomes essentially 80% of the time. Further, this classifier is significantly better than classifiers based on any subgroup of these 36 features. This study demonstrates that autonomous machine-learning techniques are able to generate simple and efficient decision-tree prognostic classifiers from a wide variety of clinical, pathologic and biomarker data, and unlike other analytic methods, suggest testable biologic relationships among explicitly identified key variables. The decision-tree classifier resulting from these analytic methods is sufficiently simple and should be widely applicable to a spectrum of clinical cancer settings. Further, the subcellular distribution of junctional proteins, which influences growth regulatory pathways involved in locoregional and metastatic relapse of breast cancer, helped to identify which patients would relapse while their total concentration did not. This emphasizes the need to evaluate the subcellular distribution of junctional proteins in assessing their contribution to tumor progression.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Conexinas/metabolismo , Recurrencia Local de Neoplasia/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/patología , Carcinoma Ductal de Mama/patología , Árboles de Decisión , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Pronóstico
18.
Breast Cancer Res Treat ; 118(3): 605-21, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19002577

RESUMEN

We recently showed that estrogen withdrawal from the ERalpha(+), high Bcl-2-expressing breast carcinoma cells (MCF-7B) reduced Bcl-2 protein levels while increasing cell-cell adhesion, and junction formation. Here we compared these cells with the ERalpha(+) and low Bcl-2-expressing MCF-7 cells and with the normal mammary epithelial cell line MCF-10-2A not expressing ERalpha or Bcl-2. All cell lines expressed normal HER2. Antiestrogen (Tamoxifen and ICI 182,780) treatment increased Bcl-2 levels in both MCF-7 and -7B cells and led to the formation of acinar structures. This treatment led to the dissociation of junctions and redistribution of junctional components to the cytoplasm in MCF-10-2A and -7 cells, while in MCF-7B cells junctional proteins redistributed to membranes. Antiestrogen treatment decreased PI3K/Akt activation and increased ERK activation regardless of ERalpha status. IGF-1R was inactivated in the antiestrogen-treated MCF-7 cells while it was activated in MCF-7B cells. Our data show that Tamoxifen and ICI 182,780 can induce growth inhibitory effects via the sustained activation/inactivation of signaling pathways that regulate cell survival, cell death and differentiation in the absence of ERalpha. Furthermore, Bcl-2 overexpression may alter the functional interactions among these pathways in response to antiestrogens, which also may provide a potential explanation for the observation that Bcl-2 overexpressing tumors have a better prognosis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Estradiol/análogos & derivados , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Transducción de Señal/efectos de los fármacos , Tamoxifeno/farmacología , Western Blotting , Adhesión Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Conexinas/efectos de los fármacos , Estradiol/farmacología , Receptor alfa de Estrógeno/efectos de los fármacos , Receptor alfa de Estrógeno/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Fulvestrant , Humanos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Receptor IGF Tipo 1/efectos de los fármacos , Receptor IGF Tipo 1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA