Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 10(7): 778-85, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19483719

RESUMEN

Studies have suggested involvement of interleukin 17 (IL-17) in autoimmune diseases, although its effect on B cell biology has not been clearly established. Here we demonstrate that IL-17 alone or in combination with B cell-activating factor controlled the survival and proliferation of human B cells and their differentiation into immunoglobulin-secreting cells. This effect was mediated mainly through the nuclear factor-kappaB-regulated transcription factor Twist-1. In support of the relevance of our observations and the potential involvement of IL-17 in B cell biology, we found that the serum of patients with systemic lupus erythematosus had higher concentrations of IL-17 than did the serum of healthy people and that IL-17 abundance correlated with the disease severity of systemic lupus erythematosus.


Asunto(s)
Factor Activador de Células B/farmacología , Linfocitos B/efectos de los fármacos , Interleucina-17/sangre , Interleucina-17/farmacología , Lupus Eritematoso Sistémico/sangre , Antígenos CD19/metabolismo , Apoptosis/efectos de los fármacos , Linfocitos B/citología , Linfocitos B/metabolismo , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Humanos , Immunoblotting , Inmunoglobulinas/metabolismo , Interleucina-17/genética , Lupus Eritematoso Sistémico/patología , Masculino , Antígenos de Histocompatibilidad Menor , FN-kappa B/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo
2.
Nucleic Acids Res ; 44(11): 5470-89, 2016 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-27151200

RESUMEN

The TWIST1 bHLH transcription factor controls embryonic development and cancer processes. Although molecular and genetic analyses have provided a wealth of data on the role of bHLH transcription factors, very little is known on the molecular mechanisms underlying their binding affinity to the E-box sequence of the promoter. Here, we used an in silico model of the TWIST1/E12 (TE) heterocomplex and performed molecular dynamics (MD) simulations of its binding to specific (TE-box) and modified E-box sequences. We focused on (i) active E-box and inactive E-box sequences, on (ii) modified active E-box sequences, as well as on (iii) two box sequences with modified adjacent bases the AT- and TA-boxes. Our in silico models were supported by functional in vitro binding assays. This exploration highlighted the predominant role of protein side-chain residues, close to the heart of the complex, at anchoring the dimer to DNA sequences, and unveiled a shift towards adjacent ((-1) and (-1*)) bases and conserved bases of modified E-box sequences. In conclusion, our study provides proof of the predictive value of these MD simulations, which may contribute to the characterization of specific inhibitors by docking approaches, and their use in pharmacological therapies by blocking the tumoral TWIST1/E12 function in cancers.


Asunto(s)
Elementos E-Box , Modelos Moleculares , Factor de Transcripción 3/química , Proteína 1 Relacionada con Twist/química , Secuencia de Bases , Sitios de Unión , Línea Celular , Humanos , Enlace de Hidrógeno , Conformación Molecular , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica , Factor de Transcripción 3/metabolismo , Proteína 1 Relacionada con Twist/metabolismo
3.
BMC Struct Biol ; 17(1): 6, 2017 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-28521820

RESUMEN

BACKGROUND: The bHLH transcription factor TWIST1 plays a key role in the embryonic development and in tumorigenesis. Some loss-of-function mutations of the TWIST1 gene have been shown to cause an autosomal dominant craniosynostosis, known as the Saethre-Chotzen syndrome (SCS). Although the functional impacts of many TWIST1 mutations have been experimentally reported, little is known on the molecular mechanisms underlying their loss-of-function. In a previous study, we highlighted the predictive value of in silico molecular dynamics (MD) simulations in deciphering the molecular function of TWIST1 residues. RESULTS: Here, since the substitution of the arginine 154 amino acid by a glycine residue (R154G) is responsible for the SCS phenotype and the substitution of arginine 154 by a proline experimentally decreases the dimerizing ability of TWIST1, we investigated the molecular impact of this point mutation using MD approaches. Consistently, MD simulations highlighted a clear decrease in the stability of the α-helix during the dimerization of the mutated R154P TWIST1/E12 dimer compared to the wild-type TE complex, which was further confirmed in vitro using immunoassays. CONCLUSIONS: Our study demonstrates that MD simulations provide a structural explanation for the loss-of-function associated with the SCS TWIST1 mutation and provides a proof of concept of the predictive value of these MD simulations. This in silico methodology could be used to determine reliable pharmacophore sites, leading to the application of docking approaches in order to identify specific inhibitors of TWIST1 complexes.


Asunto(s)
Simulación de Dinámica Molecular , Proteínas Nucleares/química , Proteínas Nucleares/genética , Mutación Puntual , Factor de Transcripción 3/química , Proteína 1 Relacionada con Twist/química , Proteína 1 Relacionada con Twist/genética , Secuencia de Aminoácidos , Animales , Arginina/química , Arginina/genética , Simulación por Computador , Cristalografía por Rayos X , Glicina/química , Glicina/genética , Humanos , Ratones , Fosforilación , Conformación Proteica , Multimerización de Proteína , Homología de Secuencia , Factor de Transcripción 3/genética
4.
PLoS Genet ; 8(5): e1002723, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22654675

RESUMEN

The epithelial-mesenchymal transition (EMT) is an embryonic transdifferentiation process consisting of conversion of polarized epithelial cells to motile mesenchymal ones. EMT-inducing transcription factors are aberrantly expressed in multiple tumor types and are known to favor the metastatic dissemination process. Supporting oncogenic activity within primary lesions, the TWIST and ZEB proteins can prevent cells from undergoing oncogene-induced senescence and apoptosis by abolishing both p53- and RB-dependent pathways. Here we show that they also downregulate PP2A phosphatase activity and efficiently cooperate with an oncogenic version of H-RAS in malignant transformation of human mammary epithelial cells. Thus, by down-regulating crucial tumor suppressor functions, EMT inducers make cells particularly prone to malignant conversion. Importantly, by analyzing transformed cells generated in vitro and by characterizing novel transgenic mouse models, we further demonstrate that cooperation between an EMT inducer and an active form of RAS is sufficient to trigger transformation of mammary epithelial cells into malignant cells exhibiting all the characteristic features of claudin-low tumors, including low expression of tight and adherens junction genes, EMT traits, and stem cell-like characteristics. Claudin-low tumors are believed to be the most primitive breast malignancies, having arisen through transformation of an early epithelial precursor with inherent stemness properties and metaplastic features. Challenging this prevailing view, we propose that these aggressive tumors arise from cells committed to luminal differentiation, through a process driven by EMT inducers and combining malignant transformation and transdifferentiation.


Asunto(s)
Neoplasias de la Mama , Transformación Celular Neoplásica , Claudinas , Transición Epitelial-Mesenquimal , Glándulas Mamarias Humanas/metabolismo , Proteína Fosfatasa 2 , Proteína 1 Relacionada con Twist/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Diferenciación Celular , Línea Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Claudinas/genética , Claudinas/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Genes ras , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Humanas/citología , Ratones , Ratones Transgénicos , Proteína Fosfatasa 2/antagonistas & inhibidores , Proteína Fosfatasa 2/metabolismo , Proteína de Retinoblastoma/metabolismo , Telomerasa/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
5.
Mol Cancer ; 13: 213, 2014 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-25216750

RESUMEN

INTRODUCTION: Tumor-initiating cells (TICs), aka "cancer stem cells", are believed to fuel tumors and to sustain therapy resistance and systemic metastasis. Breast cancer is the first human carcinoma in which a subpopulation of cells displaying a specific CD44+/CD24-/low/ESA+ antigenic phenotype was found to have TIC properties. However, CD44+/CD24-/low/ESA+ is not a universal marker phenotype of TICs in all breast cancer subtypes. The aim of this study was to identify novel antigens with which to isolate the TIC population of the basal-A/basal-like breast cancer cell lines. METHODS: We used polychromatic flow-cytometry to characterize the cell surface of several breast cancer cell lines that may represent different tumor molecular subtypes. We next used fluorescence-activated cell sorting to isolate the cell subpopulations of interest from the cell lines. Finally, we explored the stem-like and tumorigenic properties of the sorted cell subpopulations using complementary in vitro and in vivo approaches: mammosphere formation assays, soft-agar colony assays, and tumorigenic assays in NOD/SCID mice. RESULTS: The CD44+/CD24+ subpopulation of the BRCA1-mutated basal-A/basal-like cell line HCC1937 is enriched in several stemness markers, including the ABCG2 transporter (i.e., the CD338 antigen). Consistently, CD338-expressing cells were also enriched in CD24 expression, suggesting that coexpression of these two antigenic markers may segregate TICs in this cell line. In support of ABCG2 expression in TICs, culturing of HCC1937 cells in ultra-low adherent conditions to enrich them in precursor/stem-cells resulted in an increase in CD338-expressing cells. Furthermore, CD338-expressing cells, unlike their CD338-negative counterparts, displayed stemness and transformation potential, as assessed in mammosphere and colony formation assays. Lastly, CD338-expressing cells cultured in ultra-low adherent conditions maintained the expression of CD326/EpCAM and CD49f/α6-integrin, which is a combination of antigens previously assigned to luminal progenitors. CONCLUSION: Collectively, our data suggest that CD338 expression is specific to the tumor-initiating luminal progenitor subpopulation of BRCA1-mutated cells and is a novel antigen with which to sort this subpopulation.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteína BRCA1/metabolismo , Neoplasias de la Mama/patología , Citometría de Flujo , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/patología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Animales , Antígeno CD24/metabolismo , Adhesión Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Ratones Endogámicos NOD , Ratones SCID , Esferoides Celulares/patología
6.
Semin Cancer Biol ; 21(6): 392-6, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21986518

RESUMEN

The epithelial to mesenchymal transition (EMT) is a latent embryonic process which can be aberrantly reactivated during tumor progression. It is generally viewed as one of the main forces driving metastatic dissemination, by providing cells with invasive and motility capabilities. The aberrant reactivation of embryonic EMT inducers has now been additionally linked to escape from senescence and apoptosis, which suggests a role in tumor initiation. This oncogenic potential relies on the ability of EMT inducers to neutralize both the RB and p53 oncosuppressive pathways. RB and p53 have recently been described as key factors in the maintenance of epithelial morphology, which suggests an unexpected and intimate crosstalk between EMT and the corresponding safety programs. In this review, we attempt to understand how these two cell processes are interlinked and might facilitate cell transformation and tumor initiation.


Asunto(s)
Transición Epitelial-Mesenquimal , Escape del Tumor , Progresión de la Enfermedad , Humanos , Neoplasias/patología
7.
Cancers (Basel) ; 15(3)2023 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-36765930

RESUMEN

Reactive oxygen species (ROS) are considered to be the main drivers of inflammatory bowel disease. We investigated whether this permanent insult compels intestinal stem cells to develop strategies to dampen the deleterious effects of ROS. As an adverse effect, this adaptation process may increase their tolerance to oncogenic insults and facilitate their neoplastic transformation. We submitted immortalized human colonic epithelial cells to either a mimic of chronic inflammation or to a chemical peroxide, analyzed how they adapted to stress, and addressed the biological relevance of these observations in databases. We demonstrated that cells adapt to chronic-inflammation-associated oxidative stress in vitro through a partial genetic reprogramming. Through a gene set enrichment analysis, we showed that this program is recurrently active in the intestinal mucosae of Crohn's and ulcerative colitis disease patients and evolves alongside disease progression. Based on a previously reported characterization of intestinal stem and precursor cells using tracing experiments, we lastly confirmed the activation of the program in intestinal precursor cells during murine colorectal cancer development. This adaptive process is thus likely to play a role in the progression of Crohn's and ulcerative disease, and potentially in the initiation of colorectal cancer.

8.
Mol Oncol ; 2023 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-37452637

RESUMEN

Nutrient availability is a key determinant of tumor cell behavior. While nutrient-rich conditions favor proliferation and tumor growth, scarcity, and particularly glutamine starvation, promotes cell dedifferentiation and chemoresistance. Here, linking ribosome biogenesis plasticity with tumor cell fate, we uncover that the amino acid sensor general control non-derepressible 2 (GCN2; also known as eIF-2-alpha kinase 4) represses the expression of the precursor of ribosomal RNA (rRNA), 47S, under metabolic stress. We show that blockade of GCN2 triggers cell death by an irremediable nucleolar stress and subsequent TP53-mediated apoptosis in patient-derived models of colon adenocarcinoma (COAD). In nutrient-rich conditions, a cell-autonomous GCN2 activity supports cell proliferation by stimulating 47S rRNA transcription, independently of the canonical integrated stress response (ISR) axis. Impairment of GCN2 activity prevents nuclear translocation of methionyl-tRNA synthetase (MetRS), resulting in nucleolar stress, mTORC1 inhibition and, ultimately, autophagy induction. Inhibition of the GCN2-MetRS axis drastically improves the cytotoxicity of RNA polymerase I (RNA pol I) inhibitors, including the first-line chemotherapy oxaliplatin, on patient-derived COAD tumoroids. Our data thus reveal that GCN2 differentially controls ribosome biogenesis according to the nutritional context. Furthermore, pharmacological co-inhibition of the two GCN2 branches and RNA pol I activity may represent a valuable strategy for elimination of proliferative and metabolically stressed COAD cells.

9.
Cancers (Basel) ; 14(11)2022 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-35681770

RESUMEN

The kynurenine pathway has been highlighted as a gatekeeper of immune-privileged sites through its ability to generate from tryptophan a set of immunosuppressive metabolic intermediates. It additionally constitutes an important source of cellular NAD+ for the organism. Hijacking of its immunosuppressive functions, as recurrently observed in multiple cancers, facilitates immune evasion and promotes tumor development. Based on these observations, researchers have focused on characterizing indoleamine 2,3-dioxygenase (IDO1), the main enzyme catalyzing the first and limiting step of the pathway, and on developing therapies targeting it. Unfortunately, clinical trials studying IDO1 inhibitors have thus far not met expectations, highlighting the need to unravel this complex signaling pathway further. Recent advances demonstrate that these metabolites additionally promote tumor growth, metastatic dissemination and chemoresistance by a combination of paracrine and autocrine effects. Production of NAD+ also contributes to cancer progression by providing cancer cells with enhanced plasticity, invasive properties and chemoresistance. A comprehensive survey of this complexity is challenging but necessary to achieve medical success.

10.
Oncotarget ; 9(57): 30979-30996, 2018 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-30123421

RESUMEN

Our previous work demonstrated a key function of the thyroid hormone nuclear receptor TRα1, a T3-modulated transcription factor, in controlling intestinal development and homeostasis via the Wnt and Notch pathways. Importantly, increased expression of TRα1 in the intestinal epithelium in a mutated Apc genetic background (vil-TRα1/Apc+/1638N mice) accelerated tumorigenesis and contributed to a more aggressive tumor phenotype compared to that of the Apc mutants alone. Therefore, the aim of this study was to determine the relevance of this synergistic effect in human colorectal cancers and to gain insights into the mechanisms involved. We analyzed cohorts of patients by in silico and experimental approaches and observed increased TRα1 expression and a significant correlation between TRα1 levels and Wnt activity. TRα1 loss-of-function and gain-of-function in Caco2 cell lines not only confirmed that TRα1 levels control Wnt activity but also demonstrated the role of TRα1 in regulating cell proliferation and migration. Finally, upon investigation of the molecular mechanisms responsible for the Wnt-TRα1 association, we described the repression by TRα1 of several Wnt inhibitors, including Frzb, Sox17 and Wif1. In conclusion, our results underline an important functional interplay between the thyroid hormone nuclear receptor TRα1 and the canonical Wnt pathway in intestinal cancer initiation and progression. More importantly, we show for the first time that the expression of TRα1 is induced in human colorectal cancers.

11.
J Mol Biol ; 358(2): 498-508, 2006 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-16527309

RESUMEN

The MYND domain (named after myeloid translocation protein 8, Nervy, and DEAF-1) is a conserved zinc binding domain. It is defined by seven conserved cysteine residues and a single histidine residue that are arranged in a C4-C2HC consensus. MYND domains exist in a large number of proteins that play important roles in development or are associated with cancers and have been shown to mediate protein-protein interactions, mainly in the context of transcriptional regulation. We have determined the three-dimensional structure of the MYND domain from human deformed epidermal autoregulatory factor-1 (DEAF-1). The structure reveals a novel zinc binding fold, in which the C4-C2HC motif forms two sequential zinc binding sites. The first and second zinc binding modules comprise a small beta hairpin and two short alpha helices, respectively. The sequential topology of the two zinc binding sites is distinct from the cross-brace PHD and RING finger folds but has some resemblance to LIM domains. The structure reveals that the MYND domain is a novel member of the treble-clef family of zinc binding domains. The MYND domains of BS69 and BOP bind ligands comprising a PXLXP peptide motif. On the basis of the solution structure of the DEAF-1 MYND domain we calculated a homology model of the MYND domain of the BS69 tumor suppressor. A mutational analysis of the BS69 MYND domain indicates that positively charged residues located on one face of its MYND domain are crucial for the molecular interactions of BS69. Different binding specificities of MYND domains may depend on distinct surface charge distributions.


Asunto(s)
Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Secuencia de Aminoácidos , Proteínas de Unión al ADN , Humanos , Ligandos , Datos de Secuencia Molecular , Mutación , Proteínas Nucleares/genética , Fragmentos de Péptidos/metabolismo , Unión Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Factores de Transcripción , Zinc/metabolismo
12.
Nat Med ; 23(5): 568-578, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28394329

RESUMEN

Chromosomal instability (CIN), a feature of most adult neoplasms from their early stages onward, is a driver of tumorigenesis. However, several malignancy subtypes, including some triple-negative breast cancers, display a paucity of genomic aberrations, thus suggesting that tumor development may occur in the absence of CIN. Here we show that the differentiation status of normal human mammary epithelial cells dictates cell behavior after an oncogenic event and predetermines the genetic routes toward malignancy. Whereas oncogene induction in differentiated cells induces massive DNA damage, mammary stem cells are resistant, owing to a preemptive program driven by the transcription factor ZEB1 and the methionine sulfoxide reductase MSRB3. The prevention of oncogene-induced DNA damage precludes induction of the oncosuppressive p53-dependent DNA-damage response, thereby increasing stem cells' intrinsic susceptibility to malignant transformation. In accord with this model, a subclass of breast neoplasms exhibit unique pathological features, including high ZEB1 expression, a low frequency of TP53 mutations and low CIN.


Asunto(s)
Neoplasias de la Mama/genética , Carcinoma/genética , Diferenciación Celular/genética , Células Epiteliales/metabolismo , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica/genética , Metionina Sulfóxido Reductasas/genética , Células Madre/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Neoplasias de la Mama/metabolismo , Carcinoma/metabolismo , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Inmunoprecipitación de Cromatina , Daño del ADN , Células Epiteliales/citología , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Humanos , Immunoblotting , Glándulas Mamarias Humanas/citología , Metionina Sulfóxido Reductasas/metabolismo , Ratones Endogámicos NOD , Persona de Mediana Edad , Especies Reactivas de Oxígeno , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Secuencia de ADN , Células Madre/citología , Análisis de Matrices Tisulares , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Adulto Joven , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
13.
Neoplasia ; 18(5): 317-327, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27237323

RESUMEN

The TWIST1 embryonic transcription factor displays biphasic functions during the course of carcinogenesis. It facilitates the escape of cells from oncogene-induced fail-safe programs (senescence, apoptosis) and their consequent neoplastic transformation. Additionally, it promotes the epithelial-to-mesenchymal transition and the initiation of the metastatic spread of cancer cells. Interestingly, cancer cells recurrently remain dependent on TWIST1 for their survival and/or proliferation, making TWIST1 their Achilles' heel. TWIST1 has been reported to form either homodimeric or heterodimeric complexes mainly in association with the E bHLH class I proteins. These complexes display distinct, sometimes even antagonistic, functions during development and unequal prometastatic functions in prostate cancer cells. Using a tethered dimer strategy, we successively assessed the ability of TWIST1 dimers to cooperate with an activated version of RAS in human mammary epithelial cell transformation, to provide mice with the ability to spontaneously develop breast tumors, and lastly to maintain a senescence program at a latent state in several breast cancer cell lines. We demonstrate that the TWIST1-E12 complex, unlike the homodimer, is an oncogenic form of TWIST1 in mammary epithelial cells and that efficient binding of both partners is a prerequisite for its activity. The detection of the heterodimer in human premalignant lesions by a proximity ligation assay, at a stage preceding the initiation of the metastatic cascade, is coherent with such an oncogenic function. TWIST1-E protein heterodimeric complexes may thus constitute the main active forms of TWIST1 with regard to senescence inhibition over the time course of breast tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Células Epiteliales/metabolismo , Glándulas Mamarias Humanas/metabolismo , Factor de Transcripción 3/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Línea Celular Tumoral , Proliferación Celular , Senescencia Celular/genética , Células Epiteliales/patología , Expresión Génica , Humanos , Glándulas Mamarias Humanas/patología , Mutación , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Unión Proteica , Multimerización de Proteína , Factor de Transcripción 3/genética , Proteína 1 Relacionada con Twist/genética
14.
EMBO Mol Med ; 8(10): 1143-1161, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27596438

RESUMEN

Targeted therapies with MAPK inhibitors (MAPKi) are faced with severe problems of resistance in BRAF-mutant melanoma. In parallel to the acquisition of genetic mutations, melanoma cells may also adapt to the drugs through phenotype switching. The ZEB1 transcription factor, a known inducer of EMT and invasiveness, is now considered as a genuine oncogenic factor required for tumor initiation, cancer cell plasticity, and drug resistance in carcinomas. Here, we show that high levels of ZEB1 expression are associated with inherent resistance to MAPKi in BRAFV600-mutated cell lines and tumors. ZEB1 levels are also elevated in melanoma cells with acquired resistance and in biopsies from patients relapsing while under treatment. ZEB1 overexpression is sufficient to drive the emergence of resistance to MAPKi by promoting a reversible transition toward a MITFlow/p75high stem-like and tumorigenic phenotype. ZEB1 inhibition promotes cell differentiation, prevents tumorigenic growth in vivo, sensitizes naive melanoma cells to MAPKi, and induces cell death in resistant cells. Overall, our results demonstrate that ZEB1 is a major driver of melanoma cell plasticity, driving drug adaptation and phenotypic resistance to MAPKi.


Asunto(s)
Antineoplásicos/farmacología , Plasticidad de la Célula , Resistencia a Medicamentos , Melanocitos/efectos de los fármacos , Melanocitos/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Adaptación Fisiológica , Línea Celular Tumoral , Humanos , Melanoma/tratamiento farmacológico , Melanoma/fisiopatología
15.
Cancer Res ; 75(20): 4335-50, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26282171

RESUMEN

The transcription accessory factor TIF1γ/TRIM33/RFG7/PTC7/Ectodermin functions as a tumor suppressor that promotes development and cellular differentiation. However, its precise function in cancer has been elusive. In the present study, we report that TIF1γ inactivation causes cells to accumulate chromosomal defects, a hallmark of cancer, due to attenuations in the spindle assembly checkpoint and the post-mitotic checkpoint. TIF1γ deficiency also caused a loss of contact growth inhibition and increased anchorage-independent growth in vitro and in vivo. Clinically, reduced TIF1γ expression in human tumors correlated with an increased rate of genomic rearrangements. Overall, our work indicates that TIF1γ exerts its tumor-suppressive functions in part by promoting chromosomal stability.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Inestabilidad Cromosómica , Regulación Neoplásica de la Expresión Génica , Mitosis/genética , Neoplasias/genética , Neoplasias/metabolismo , Factores de Transcripción/metabolismo , Animales , Carcinoma in Situ , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación hacia Abajo , Transición Epitelial-Mesenquimal/genética , Silenciador del Gen , Humanos , Ratones , Ratones Noqueados , Neoplasias/patología , Ploidias , Huso Acromático/metabolismo
16.
FEBS Lett ; 521(1-3): 36-8, 2002 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-12067721

RESUMEN

The LIM only protein Lmo2 plays an important role in hematopoiesis and leukemogenesis. Lmo2 acts as a bridging molecule between components of hematopoietic gene regulatory protein complexes. We used the yeast two-hybrid system to identify novel Lmo2 interacting proteins and found that the AF6 protein binds to Lmo2. AF6 is a recurrent fusion partner of MLL, the human homolog of Drosophila trithorax chromatin remodeling protein that is involved in childhood leukemia and mixed lineage leukemia. Our data support the notion that recurrent fusion partners of chimeric MLL proteins recruit hematopoietic gene regulatory complexes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila , Cinesinas/metabolismo , Metaloproteínas/metabolismo , Miosinas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proto-Oncogenes , Factores de Transcripción , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular , Proteínas de Unión al ADN/genética , N-Metiltransferasa de Histona-Lisina , Humanos , Células Jurkat , Cinesinas/genética , Proteínas con Dominio LIM , Metaloproteínas/genética , Proteína de la Leucemia Mieloide-Linfoide , Miosinas/genética , Proteínas Proto-Oncogénicas/genética , Codorniz , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
19.
J Bone Miner Res ; 29(8): 1886-99, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24619707

RESUMEN

The transcription factor TWIST1 induces epithelial-mesenchymal transition and/or escape to the oncogenic-induced failsafe program, facilitating the intravasation of breast cancer cells in the systemic circulation and their dissemination to the lungs. Its involvement in breast cancer bone metastasis is unknown. To address this question, human osteotropic MDA-MB-231/B02 breast cancer cells were stably transfected with a Tet-inducible vector encoding for TWIST1, whose expression was specifically repressed in the presence of doxycycline (dox). The intra-arterial inoculation of transfectants expressing TWIST1 in immunodeficient mice substantially increased the extent of osteolytic lesions in these animals, being 50% larger than that of animals bearing mock-transfected tumors, as determined by radiography. This difference was accompanied by a sharp reduction of the bone volume (indicating a higher bone destruction) and a twofold increase in the tumor volume compared with mice bearing mock-transfected tumors, as determined by histomorphometry. Importantly, the suppression of TWIST1 expression in MDA-MB-231/B02 cells in the presence of dox abolished the stimulatory effect of TWIST1 on bone metastasis formation in vivo. Additionally, examination of the bone marrow from untreated and dox-treated animals on day 7 after tumor cell inoculation, at which time there was no evidence of radiographic osteolytic lesions, revealed that the number of tumor cell colonies that were recovered from the bone marrow of untreated mice was dramatically increased compared with that of dox-fed animals. In vitro, TWIST1 expression promoted tumor cell invasion and enhanced microRNA 10b (miR-10b) expression, a proinvasive factor, but was dispensable for growth of tumor cells. In vivo, the repression of miR-10b substantially decreased the presence of TWIST1-expressing breast cancer cells in the bone marrow. Overall, these results establish that TWIST1 facilitates breast cancer bone metastasis formation through a mechanism dependent of miR-10b, which leads to increase tumor burden and bone destruction.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares/genética , Proteína 1 Relacionada con Twist/genética , Animales , Western Blotting , Diferenciación Celular , Línea Celular Tumoral , Doxiciclina/farmacología , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Metástasis de la Neoplasia , Proteínas Nucleares/metabolismo , Osteoclastos/citología , Proteína 1 Relacionada con Twist/metabolismo
20.
PLoS One ; 9(3): e92254, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24638100

RESUMEN

By fostering cell commitment to the epithelial-to-mesenchymal transition (EMT), SNAIL proteins endow cells with motility, thereby favoring the metastatic spread of tumor cells. Whether the phenotypic change additionally facilitates tumor initiation has never been addressed. Here we demonstrate that when a SNAIL protein is ectopically produced in non-transformed mammary epithelial cells, the cells are protected from anoikis and proliferate under low-adherence conditions: a hallmark of cancer cells. The three SNAIL proteins show unequal oncogenic potential, strictly correlating with their ability to promote EMT. SNAIL3 especially behaves as a poor EMT-inducer comforting the concept that the transcription factor functionally diverges from its two related proteins.


Asunto(s)
Transformación Celular Neoplásica/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Transición Epitelial-Mesenquimal , Glándulas Mamarias Humanas/patología , Factores de Transcripción/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Adhesión Celular/genética , Supervivencia Celular/genética , Transformación Celular Neoplásica/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción de la Familia Snail , Telomerasa/metabolismo , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA