Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
iScience ; 27(4): 109366, 2024 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-38510127

RESUMEN

Triple-negative breast cancer (TNBC) contributes greatly to mortality of breast cancer, demanding new targetable options. We have shown that TNBC patients have high ΔNp63 expression in tumors. However, the function of ΔNp63 in established TNBC is yet to be explored. In current studies, targeting ΔNp63 with inducible CRISPR knockout and Histone deacetylase inhibitor Quisinostat showed that ΔNp63 is important for tumor progression and metastasis in established tumors by promoting myeloid-derived suppressor cell (MDSC) survival through tumor necrosis factor alpha. Decreasing ΔNp63 levels are associated with decreased CD4+ and FOXP3+ T-cells but increased CD8+ T-cells. RNA sequencing analysis indicates that loss of ΔNp63 alters multiple MDSC properties such as lipid metabolism, chemotaxis, migration, and neutrophil degranulation besides survival. We further demonstrated that targeting ΔNp63 sensitizes chemotherapy. Overall, we showed that ΔNp63 reprograms the MDSC-mediated immunosuppressive functions in TNBC, highlighting the benefit of targeting ΔNp63 in chemotherapy-resistant TNBC.

5.
Nat Commun ; 12(1): 432, 2021 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-33462238

RESUMEN

Development of chemoresistance in breast cancer patients greatly increases mortality. Thus, understanding mechanisms underlying breast cancer resistance to chemotherapy is of paramount importance to overcome this clinical challenge. Although activated Notch receptors have been associated with chemoresistance in cancer, the specific Notch ligands and their molecular mechanisms leading to chemoresistance in breast cancer remain elusive. Using conditional knockout and reporter mouse models, we demonstrate that tumor cells expressing the Notch ligand Dll1 is important for tumor growth and metastasis and bear similarities to tumor-initiating cancer cells (TICs) in breast cancer. RNA-seq and ATAC-seq using reporter models and patient data demonstrated that NF-κB activation is downstream of Dll1 and is associated with a chemoresistant phenotype. Finally, pharmacological blocking of Dll1 or NF-κB pathway completely sensitizes Dll1+ tumors to chemotherapy, highlighting therapeutic avenues for chemotherapy resistant breast cancer patients in the near future.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proteínas de Unión al Calcio/metabolismo , Resistencia a Antineoplásicos/genética , Proteínas de la Membrana/metabolismo , Subunidad p50 de NF-kappa B/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Benzamidas/farmacología , Benzamidas/uso terapéutico , Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Unión al Calcio/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Doxorrubicina/farmacología , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Subunidad p50 de NF-kappa B/antagonistas & inhibidores , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , RNA-Seq , Receptores Notch/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
6.
Nat Cell Biol ; 22(5): 591-602, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32284542

RESUMEN

Triple-negative breast cancer (TNBC) is characterized by a high degree of immune infiltrate in the tumour microenvironment, which may influence the fate of TNBC cells. We reveal that loss of the tumour suppressive transcription factor Elf5 in TNBC cells activates intrinsic interferon-γ (IFN-γ) signalling, promoting tumour progression and metastasis. Mechanistically, we find that loss of the Elf5-regulated ubiquitin ligase FBXW7 ensures stabilization of its putative protein substrate IFN-γ receptor 1 (IFNGR1) at the protein level in TNBC. Elf5low tumours show enhanced IFN-γ signalling accompanied by an increase of immunosuppressive neutrophils within the tumour microenvironment and increased programmed death ligand 1 expression. Inactivation of either programmed death ligand 1 or IFNGR1 elicited a robust anti-tumour and/or anti-metastatic effect. A positive correlation between ELF5 and FBXW7 expression and a negative correlation between ELF5, FBXW7 and IFNGR1 expression in the tumours of patients with TNBC strongly suggest that this signalling axis could be exploited for patient stratification and immunotherapeutic treatment strategies for Elf5low patients with TNBC.


Asunto(s)
Proliferación Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Interferón gamma/metabolismo , Metástasis de la Neoplasia/patología , Receptores de Interferón/metabolismo , Factores de Transcripción/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Ratones , Ratones Endogámicos BALB C , Transducción de Señal/fisiología , Microambiente Tumoral/fisiología , Receptor de Interferón gamma
7.
Cancer Cell ; 36(5): 528-544.e10, 2019 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-31631026

RESUMEN

H3K27M mutations resulting in epigenetic dysfunction are frequently observed in diffuse intrinsic pontine glioma (DIPGs), an incurable pediatric cancer. We conduct a CRISPR screen revealing that knockout of KDM1A encoding lysine-specific demethylase 1 (LSD1) sensitizes DIPG cells to histone deacetylase (HDAC) inhibitors. Consistently, Corin, a bifunctional inhibitor of HDACs and LSD1, potently inhibits DIPG growth in vitro and in xenografts. Mechanistically, Corin increases H3K27me3 levels suppressed by H3K27M histones, and simultaneously increases HDAC-targeted H3K27ac and LSD1-targeted H3K4me1 at differentiation-associated genes. Corin treatment induces cell death, cell-cycle arrest, and a cellular differentiation phenotype and drives transcriptional changes correlating with increased survival time in DIPG patients. These data suggest a strategy for treating DIPG by simultaneously inhibiting LSD1 and HDACs.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Tronco Encefálico/tratamiento farmacológico , Glioma/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Histona Demetilasas/antagonistas & inhibidores , Animales , Antineoplásicos/uso terapéutico , Neoplasias del Tronco Encefálico/genética , Neoplasias del Tronco Encefálico/mortalidad , Neoplasias del Tronco Encefálico/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular Tumoral , Cromatina/metabolismo , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Glioma/genética , Glioma/mortalidad , Glioma/patología , Código de Histonas/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Histona Desacetilasas/metabolismo , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Histonas/metabolismo , Humanos , Ratones , Mutación , Puente/patología , RNA-Seq , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Clin Invest ; 128(11): 5095-5109, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30295647

RESUMEN

Triple-negative breast cancer (TNBC) is particularly aggressive, with enhanced incidence of tumor relapse, resistance to chemotherapy, and metastases. As the mechanistic basis for this aggressive phenotype is unclear, treatment options are limited. Here, we showed an increased population of myeloid-derived immunosuppressor cells (MDSCs) in TNBC patients compared with non-TNBC patients. We found that high levels of the transcription factor ΔNp63 correlate with an increased number of MDSCs in basal TNBC patients, and that ΔNp63 promotes tumor growth, progression, and metastasis in human and mouse TNBC cells. Furthermore, we showed that MDSC recruitment to the primary tumor and metastatic sites occurs via direct ΔNp63-dependent activation of the chemokines CXCL2 and CCL22. CXCR2/CCR4 inhibitors reduced MDSC recruitment, angiogenesis, and metastasis, highlighting a novel treatment option for this subset of TNBC patients. Finally, we found that MDSCs secrete prometastatic factors such as MMP9 and chitinase 3-like 1 to promote TNBC cancer stem cell function, thereby identifying a nonimmunologic role for MDSCs in promoting TNBC progression. These findings identify a unique crosstalk between ΔNp63+ TNBC cells and MDSCs that promotes tumor progression and metastasis, which could be exploited in future combined immunotherapy/chemotherapy strategies for TNBC patients.


Asunto(s)
Células Supresoras de Origen Mieloide/inmunología , Células Madre Neoplásicas/inmunología , Factores de Transcripción/inmunología , Neoplasias de la Mama Triple Negativas/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Quimiocina CCL22/genética , Quimiocina CCL22/inmunología , Quimiocina CXCL2/genética , Quimiocina CXCL2/inmunología , Proteína 1 Similar a Quitinasa-3/genética , Proteína 1 Similar a Quitinasa-3/inmunología , Femenino , Humanos , Células MCF-7 , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Células Supresoras de Origen Mieloide/patología , Metástasis de la Neoplasia , Células Madre Neoplásicas/patología , Receptores CCR4/genética , Receptores CCR4/inmunología , Receptores CXCR4/genética , Receptores CXCR4/inmunología , Factores de Transcripción/genética , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/terapia , Proteínas Supresoras de Tumor/genética
9.
Nat Rev Mol Cell Biol ; 16(12): 705-10, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26507168

RESUMEN

DNA N(6)-adenine methylation (N(6)-methyladenine; 6mA) in prokaryotes functions primarily in the host defence system. The prevalence and significance of this modification in eukaryotes had been unclear until recently. Here, we discuss recent publications documenting the presence of 6mA in Chlamydomonas reinhardtii, Drosophila melanogaster and Caenorhabditis elegans; consider possible roles for this DNA modification in regulating transcription, the activity of transposable elements and transgenerational epigenetic inheritance; and propose 6mA as a new epigenetic mark in eukaryotes.


Asunto(s)
Adenina/análogos & derivados , Caenorhabditis elegans/genética , Chlamydomonas reinhardtii/genética , Metilación de ADN , Drosophila melanogaster/genética , Epigénesis Genética , Adenina/química , Animales , ADN/química , ADN/genética , Elementos Transponibles de ADN/genética , Marcadores Genéticos , Filogenia , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/clasificación , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/metabolismo , Transcripción Genética/genética
10.
Cell ; 161(4): 868-78, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25936839

RESUMEN

In mammalian cells, DNA methylation on the fifth position of cytosine (5mC) plays an important role as an epigenetic mark. However, DNA methylation was considered to be absent in C. elegans because of the lack of detectable 5mC, as well as homologs of the cytosine DNA methyltransferases. Here, using multiple approaches, we demonstrate the presence of adenine N(6)-methylation (6mA) in C. elegans DNA. We further demonstrate that this modification increases trans-generationally in a paradigm of epigenetic inheritance. Importantly, we identify a DNA demethylase, NMAD-1, and a potential DNA methyltransferase, DAMT-1, which regulate 6mA levels and crosstalk between methylations of histone H3K4 and adenines and control the epigenetic inheritance of phenotypes associated with the loss of the H3K4me2 demethylase spr-5. Together, these data identify a DNA modification in C. elegans and raise the exciting possibility that 6mA may be a carrier of heritable epigenetic information in eukaryotes.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/genética , Metilación de ADN , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/metabolismo , Adenina/metabolismo , Animales , Caenorhabditis elegans/fisiología , Proteínas de Caenorhabditis elegans/genética , Fertilidad , Histonas/metabolismo , Mutación , Oxidorreductasas N-Desmetilantes/genética , Oxidorreductasas N-Desmetilantes/metabolismo , Filogenia , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica)/genética
11.
Cancer Cell ; 26(3): 358-373, 2014 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-25203322

RESUMEN

Metastatic dissemination is often initiated by the reactivation of an embryonic development program referred to as epithelial-mesenchymal transition (EMT). The transcription factor SNAIL promotes EMT and elicits associated pathological characteristics such as invasion, metastasis, and stemness. To better understand the posttranslational regulation of SNAIL, we performed a luciferase-based, genome-wide E3 ligase siRNA library screen and identified SCF-FBXO11 as an important E3 that targets SNAIL for ubiquitylation and degradation. Furthermore, we discovered that SNAIL degradation by FBXO11 is dependent on Ser-11 phosphorylation of SNAIL by protein kinase D1 (PKD1). FBXO11 blocks SNAIL-induced EMT, tumor initiation, and metastasis in multiple breast cancer models. These findings establish the PKD1-FBXO11-SNAIL axis as a mechanism of posttranslational regulation of EMT and cancer metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proteínas F-Box/metabolismo , Neoplasias Pulmonares/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Canales Catiónicos TRPP/fisiología , Factores de Transcripción/metabolismo , Ubiquitinación , Animales , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Línea Celular Tumoral , Supervivencia sin Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Humanos , Estimación de Kaplan-Meier , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/secundario , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Fosforilación , Estructura Terciaria de Proteína , Proteolisis , Factores de Transcripción de la Familia Snail
12.
Cancer Cell ; 26(1): 92-105, 2014 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-24981741

RESUMEN

The Metadherin gene (MTDH) is prevalently amplified in breast cancer and associated with poor prognosis; however, its functional contribution to tumorigenesis is poorly understood. Using mouse models representing different subtypes of breast cancer, we demonstrated that MTDH plays a critical role in mammary tumorigenesis by regulating oncogene-induced expansion and activities of tumor-initiating cells (TICs), whereas it is largely dispensable for normal development. Mechanistically, MTDH supports the survival of mammary epithelial cells under oncogenic/stress conditions by interacting with and stabilizing Staphylococcal nuclease domain-containing 1 (SND1). Silencing MTDH or SND1 individually or disrupting their interaction compromises tumorigenenic potential of TICs in vivo. This functional significance of MTDH-SND1 interaction is further supported by clinical analysis of human breast cancer samples.


Asunto(s)
Neoplasias de la Mama/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Glándulas Mamarias Animales/metabolismo , Proteínas de la Membrana/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Nucleares/metabolismo , 9,10-Dimetil-1,2-benzantraceno , Animales , Neoplasias de la Mama/inducido químicamente , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/virología , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Transformación Celular Viral , Endonucleasas , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Glándulas Mamarias Animales/patología , Glándulas Mamarias Animales/virología , Virus del Tumor Mamario del Ratón/genética , Virus del Tumor Mamario del Ratón/patogenicidad , Acetato de Medroxiprogesterona , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Invasividad Neoplásica , Células Madre Neoplásicas/patología , Proteínas Nucleares/genética , Fenotipo , Unión Proteica , Interferencia de ARN , Proteínas de Unión al ARN , Factores de Tiempo , Transfección , Células Tumorales Cultivadas
13.
Cell Res ; 22(9): 1339-55, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22688892

RESUMEN

Bone is the one of the most common sites of distant metastasis of solid tumors. Secreted proteins are known to influence pathological interactions between metastatic cancer cells and the bone stroma. To comprehensively profile secreted proteins associated with bone metastasis, we used quantitative and non-quantitative mass spectrometry to globally analyze the secretomes of nine cell lines of varying bone metastatic ability from multiple species and cancer types. By comparing the secretomes of parental cells and their bone metastatic derivatives, we identified the secreted proteins that were uniquely associated with bone metastasis in these cell lines. We then incorporated bioinformatic analyses of large clinical metastasis datasets to obtain a list of candidate novel bone metastasis proteins of several functional classes that were strongly associated with both clinical and experimental bone metastasis. Functional validation of selected proteins indicated that in vivo bone metastasis can be promoted by high expression of (1) the salivary cystatins CST1, CST2, and CST4; (2) the plasminogen activators PLAT and PLAU; or (3) the collagen functionality proteins PLOD2 and COL6A1. Overall, our study has uncovered several new secreted mediators of bone metastasis and therefore demonstrated that secretome analysis is a powerful method for identification of novel biomarkers and candidate therapeutic targets.


Asunto(s)
Neoplasias Óseas/metabolismo , Proteoma/análisis , Animales , Biomarcadores de Tumor/análisis , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Línea Celular Tumoral , Colágeno Tipo VI/metabolismo , Biología Computacional , Humanos , Espectrometría de Masas , Ratones , Neoplasias/patología , Activadores Plasminogénicos/metabolismo , Procolágeno-Lisina 2-Oxoglutarato 5-Dioxigenasa/metabolismo , Proteómica , Cistatinas Salivales/metabolismo
14.
Breast Cancer Res ; 13(2): 206, 2011 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-21457525

RESUMEN

The advent of genomic profiling technology has brought about revolutionary changes in our understanding of breast cancer metastasis. Gene expression analyses of primary tumors have been used to predict metastatic propensity with high accuracy. Animal models of metastasis additionally offer a platform to experimentally dissect components of the metastasis genetic program. Recent integrated studies have synergized clinical bioinformatic analyses with advanced experimental methodology and begun to uncover the identities and dynamics of signaling programs driving breast cancer metastasis. Such functional genomics studies hold great promise for understanding the genetic basis of metastasis and improving therapeutics for advanced diseases.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Metástasis de la Neoplasia , Transducción de Señal , Animales , Mama/metabolismo , Mama/patología , Neoplasias de la Mama/genética , Femenino , Perfilación de la Expresión Génica , Genómica , Humanos , Ratones , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Transducción de Señal/genética
15.
J Biol Chem ; 286(22): 19982-92, 2011 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-21478147

RESUMEN

Metastasis is the deadliest and most poorly understood feature of malignant diseases. Recent work has shown that Metadherin (MTDH) is overexpressed in over 40% of breast cancer patients and promotes metastasis and chemoresistance in experimental models of breast cancer progression. Here we applied mass spectrometry-based screen to identify staphylococcal nuclease domain-containing 1 (SND1) as a candidate MTDH-interacting protein. After confirming the interaction between SND1 and MTDH, we tested the role of SND1 in breast cancer and found that it strongly promotes lung metastasis. SND1 was further shown to promote resistance to apoptosis and to regulate the expression of genes associated with metastasis and chemoresistance. Analyses of breast cancer clinical microarray data indicated that high expression of SND1 in primary tumors is strongly associated with reduced metastasis-free survival in multiple large scale data sets. Thus, we have uncovered SND1 as a novel MTDH-interacting protein and shown that it is a functionally and clinically significant mediator of metastasis.


Asunto(s)
Neoplasias de la Mama/metabolismo , Moléculas de Adhesión Celular/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Moléculas de Adhesión Celular/genética , Línea Celular , Supervivencia sin Enfermedad , Perros , Endonucleasas , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Proteínas de la Membrana , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas de Unión al ARN , Tasa de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA