Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 63(3): 526-38, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27453044

RESUMEN

Intratumor genetic heterogeneity underlies the ability of tumors to evolve and adapt to different environmental conditions. Using CRISPR/Cas9 technology and specific DNA barcodes, we devised a strategy to recapitulate and trace the emergence of subpopulations of cancer cells containing a mutation of interest. We used this approach to model different mechanisms of lung cancer cell resistance to EGFR inhibitors and to assess effects of combined drug therapies. By overcoming intrinsic limitations of current approaches, CRISPR-barcoding also enables investigation of most types of genetic modifications, including repair of oncogenic driver mutations. Finally, we used highly complex barcodes inserted at a specific genome location as a means of simultaneously tracing the fates of many thousands of genetically labeled cancer cells. CRISPR-barcoding is a straightforward and highly flexible method that should greatly facilitate the functional investigation of specific mutations, in a context that closely mimics the complexity of cancer.


Asunto(s)
Biomarcadores de Tumor/genética , Sistemas CRISPR-Cas , Carcinoma de Pulmón de Células no Pequeñas/genética , ADN de Neoplasias/genética , Edición Génica/métodos , Heterogeneidad Genética , Neoplasias Pulmonares/genética , Oncogenes , Mutación Puntual , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Linaje de la Célula , Células Clonales/efectos de los fármacos , Células Clonales/metabolismo , Células Clonales/patología , Análisis Mutacional de ADN , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Predisposición Genética a la Enfermedad , Células HCT116 , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Células MCF-7 , Masculino , Ratones SCID , Reacción en Cadena de la Polimerasa Multiplex , Fenotipo , Inhibidores de Proteínas Quinasas/farmacología , Factores de Tiempo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Carcinogenesis ; 43(6): 528-537, 2022 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-35239955

RESUMEN

There is increased incidence of prostate cancer (PC) among World Trade Center (WTC)-exposed responders and community members, with preliminary evidence suggestive of more aggressive disease. While previous research is supportive of differences in DNA methylation and gene expression as a consequence of WTC exposure, as measured in blood of healthy individuals, the epigenetics of WTC PC tissues has yet to be explored. Patients were recruited from the World Trade Center Health Program. Non-WTC PC samples were frequency matched on age, race/ethnicity and Gleason score. Bisulfite-treated DNA was extracted from tumor tissue blocks and used to assess global DNA methylation with the MethylationEPIC BeadChip. Differential and pathway enrichment analyses were conducted. RNA from the same tumor blocks was used for gene expression analysis to further support DNA methylation findings. Methylation data were generated for 28 samples (13 WTC and 15 non-WTC). Statistically significant differences in methylation were observed for 3,586 genes; on average WTC samples were statistically significantly more hypermethylated (P = 0.04131). Pathway enrichment analysis revealed hypermethylation in epithelial mesenchymal transition (EMT), hypoxia, mitotic spindle, TNFA signaling via NFKB, WNT signaling, and TGF beta signaling pathways in WTC compared to non-WTC samples. The androgen response, G2M and MYC target pathways were hypomethylated. These results correlated well with RNA gene expression. In conclusion, long-term epigenic changes associated with WTC dust exposure were observed in PC tissues. These occurred in genes of critical pathways, likely increasing prostate tumorigenesis potential. This warrants analysis of larger WTC groups and other cancer types.


Asunto(s)
Neoplasias de la Próstata , Ataques Terroristas del 11 de Septiembre , Metilación de ADN/genética , Polvo , Humanos , Masculino , Neoplasias de la Próstata/genética , ARN
3.
Genes Dev ; 27(17): 1868-85, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24013501

RESUMEN

The p53 tumor suppressor is a transcription factor that mediates varied cellular responses. The C terminus of p53 is subjected to multiple and diverse post-translational modifications. An attractive hypothesis is that differing sets of combinatorial modifications therein determine distinct cellular outcomes. To address this in vivo, a Trp53(ΔCTD/ΔCTD) mouse was generated in which the endogenous p53 is targeted and replaced with a truncated mutant lacking the C-terminal 24 amino acids. These Trp53(ΔCTD/ΔCTD) mice die within 2 wk post-partum with hematopoietic failure and impaired cerebellar development. Intriguingly, the C terminus acts via three distinct mechanisms to control p53-dependent gene expression depending on the tissue. First, in the bone marrow and thymus, the C terminus dampens p53 activity. Increased senescence in the Trp53(ΔCTD/ΔCTD) bone marrow is accompanied by up-regulation of Cdkn1 (p21). In the thymus, the C-terminal domain negatively regulates p53-dependent gene expression by inhibiting promoter occupancy. Here, the hyperactive p53(ΔCTD) induces apoptosis via enhanced expression of the proapoptotic Bbc3 (Puma) and Pmaip1 (Noxa). In the liver, a second mechanism prevails, since p53(ΔCTD) has wild-type DNA binding but impaired gene expression. Thus, the C terminus of p53 is needed in liver cells at a step subsequent to DNA binding. Finally, in the spleen, the C terminus controls p53 protein levels, with the overexpressed p53(ΔCTD) showing hyperactivity for gene expression. Thus, the C terminus of p53 regulates gene expression via multiple mechanisms depending on the tissue and target, and this leads to specific phenotypic effects in vivo.


Asunto(s)
Regulación de la Expresión Génica , Genes p53/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/genética , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Senescencia Celular/genética , Cerebelo/crecimiento & desarrollo , Cerebelo/metabolismo , Técnicas de Sustitución del Gen , Crecimiento y Desarrollo/genética , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Unión Proteica , Procesamiento Proteico-Postraduccional , Eliminación de Secuencia/genética , Timocitos/citología , Timocitos/metabolismo , Factores de Tiempo
4.
J Immunol ; 201(6): 1727-1734, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30068593

RESUMEN

Glatiramer acetate (GA; Copaxone) is a copolymer therapeutic that is approved by the Food and Drug Administration for the relapsing-remitting form of multiple sclerosis. Despite an unclear mechanism of action, studies have shown that GA promotes protective Th2 immunity and stimulates release of cytokines that suppress autoimmunity. In this study, we demonstrate that GA interacts with murine paired Ig-like receptor B (PIR-B) on myeloid-derived suppressor cells and suppresses the STAT1/NF-κB pathways while promoting IL-10/TGF-ß cytokine release. In inflammatory bowel disease models, GA enhanced myeloid-derived suppressor cell-dependent CD4+ regulatory T cell generation while reducing proinflammatory cytokine secretion. Human monocyte-derived macrophages responded to GA by reducing TNF-α production and promoting CD163 expression typical of alternative maturation despite the presence of GM-CSF. Furthermore, GA competitively interacts with leukocyte Ig-like receptors B (LILRBs), the human orthologs of PIR-B. Because GA limited proinflammatory activation of myeloid cells, therapeutics that target LILRBs represent novel treatment modalities for autoimmune indications.


Asunto(s)
Antígenos CD/inmunología , Acetato de Glatiramer/farmacología , Células Supresoras de Origen Mieloide/inmunología , Receptores Inmunológicos/inmunología , Animales , Antígenos CD/genética , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Citocinas/genética , Citocinas/inmunología , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células Supresoras de Origen Mieloide/patología , FN-kappa B/genética , FN-kappa B/inmunología , Receptores Inmunológicos/genética , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Células Th2/inmunología , Células Th2/patología
5.
Genes Dev ; 24(19): 2157-68, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20837657

RESUMEN

Abelson (Abl) family tyrosine kinases have been implicated in cell morphogenesis, adhesion, motility, and oncogenesis. Using a candidate approach for genes involved in planar cell polarity (PCP) signaling, we identified Drosophila Abl (dAbl) as a modulator of Frizzled(Fz)/PCP signaling. We demonstrate that dAbl positively regulates the Fz/Dishevelled (Dsh) PCP pathway without affecting canonical Wnt/Wg-Fz signaling. Genetic dissection suggests that Abl functions via Fz/Dsh signaling in photoreceptor R3 specification, a well-established Fz-PCP signaling readout. Molecular analysis shows that dAbl binds and phosphorylates Dsh on Tyr473 within the DEP domain. This phosphorylation event on Dsh is functionally critical, as the equivalent DshY473F mutant is nonfunctional in PCP signaling and stable membrane association, although it rescues canonical Wnt signaling. Strikingly, mouse embryonic fibroblasts (MEFs) deficient for Abl1 and Abl2/Arg genes also show reduced Dvl2 phosphorylation as compared with control MEFs, and this correlates with a change in subcellular localization of endogenous Dvl2. As in Drosophila, such Abl-deficient MEFs show no change in canonical Wnt signaling. Taken together, our results argue for a conserved role of Abl family members in the positive regulation of Dsh activity toward Fz-Dsh/PCP signaling by Dsh phosphorylation.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Polaridad Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Receptores Frizzled/metabolismo , Fosfoproteínas/metabolismo , Células Fotorreceptoras de Invertebrados/citología , Proteínas Tirosina Quinasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Proteínas Dishevelled , Drosophila melanogaster/citología , Drosophila melanogaster/enzimología , Fenotipo , Fosforilación , Unión Proteica
6.
Genes Dev ; 24(22): 2517-30, 2010 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21078818

RESUMEN

Wnt ligands signal through ß-catenin and are critically involved in cell fate determination and stem/progenitor self-renewal. Wnts also signal through ß-catenin-independent or noncanonical pathways that regulate crucial events during embryonic development. The mechanism of noncanonical receptor activation and how Wnts trigger canonical as opposed to noncanonical signaling have yet to be elucidated. We demonstrate here that prototype canonical Wnt3a and noncanonical Wnt5a ligands specifically trigger completely unrelated endogenous coreceptors-LRP5/6 and Ror1/2, respectively-through a common mechanism that involves their Wnt-dependent coupling to the Frizzled (Fzd) coreceptor and recruitment of shared components, including dishevelled (Dvl), axin, and glycogen synthase kinase 3 (GSK3). We identify Ror2 Ser 864 as a critical residue phosphorylated by GSK3 and required for noncanonical receptor activation by Wnt5a, analogous to the priming phosphorylation of low-density receptor-related protein 6 (LRP6) in response to Wnt3a. Furthermore, this mechanism is independent of Ror2 receptor Tyr kinase functions. Consistent with this model of Wnt receptor activation, we provide evidence that canonical and noncanonical Wnts exert reciprocal pathway inhibition at the cell surface by competition for Fzd binding. Thus, different Wnts, through their specific coupling and phosphorylation of unrelated coreceptors, activate completely distinct signaling pathways.


Asunto(s)
Proteínas Wnt/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Membrana Celular/metabolismo , Receptores Frizzled/metabolismo , Humanos , Proteínas Relacionadas con Receptor de LDL/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad , Ratones , Fosforilación , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteína Wnt-5a , Proteína Wnt3 , Proteína Wnt3A
7.
Bioconjug Chem ; 28(5): 1413-1421, 2017 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-28316241

RESUMEN

Active targeting of nanoparticles through surface functionalization is a common strategy to enhance tumor delivery specificity. However, active targeting strategies tend to work against long polyethylene glycol's shielding effectiveness and associated favorable pharmacokinetics. To overcome these limitations, we developed a matrix metalloproteinase-2 sensitive surface-converting polyethylene glycol coating. This coating prevents nanoparticle-cell interaction in the bloodstream, but, once exposed to matrix metalloproteinase-2, i.e., when the nanoparticles accumulate within the tumor interstitium, the converting polyethylene glycol coating is cleaved, and targeting ligands become available for binding to tumor cells. In this study, we applied a comprehensive multimodal imaging strategy involving optical, nuclear, and magnetic resonance imaging methods to evaluate this coating approach in a breast tumor mouse model. The data obtained revealed that this surface-converting coating enhances the nanoparticle's blood half-life and tumor accumulation and ultimately results in improved tumor-cell targeting. Our results show that this enzyme-specific surface-converting coating ensures a high cell-targeting specificity without compromising favorable nanoparticle pharmacokinetics.


Asunto(s)
Neoplasias de la Mama/patología , Imagen por Resonancia Magnética/métodos , Metaloproteinasa 2 de la Matriz/metabolismo , Imagen Multimodal/métodos , Nanopartículas/administración & dosificación , Espectrofotometría Infrarroja/métodos , Animales , Neoplasias de la Mama/metabolismo , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Metaloproteinasa 2 de la Matriz/química , Ratones , Ratones Desnudos , Nanopartículas/química , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Mol Cell ; 36(3): 379-92, 2009 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-19917247

RESUMEN

The p53 tumor suppressor protein has a well-established role in cell-fate decision-making processes. However, recent discoveries indicate that p53 has a non-tumor-suppressive role. Here we identify guanidinoacetate methyltransferase (GAMT), an enzyme involved in creatine synthesis, as a p53 target gene and a key downstream effector of adaptive response to nutrient stress. We show that GAMT is not only involved in p53-dependent apoptosis in response to genotoxic stress but is important for apoptosis induced by glucose deprivation. Additionally, p53-->GAMT upregulates fatty acid oxidation (FAO) induced by glucose starvation, utilizing this pathway as an alternate ATP-generating energy source. These results highlight that p53-dependent regulation of GAMT allows cells to maintain energy levels sufficient to undergo apoptosis or survival under conditions of nutrient stress. The p53-->GAMT pathway represents a new link between cellular stress responses and processes of creatine synthesis and FAO, demonstrating a further role of p53 in cellular metabolism.


Asunto(s)
Apoptosis/fisiología , Guanidinoacetato N-Metiltransferasa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adenosina Trifosfato/metabolismo , Animales , Antineoplásicos Fitogénicos/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Western Blotting , Línea Celular Tumoral , Creatina/biosíntesis , Daño del ADN , Etopósido/farmacología , Ácidos Grasos/metabolismo , Rayos gamma , Regulación de la Expresión Génica , Glucosa/farmacología , Guanidinoacetato N-Metiltransferasa/genética , Células HCT116 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Estrés Oxidativo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/genética
9.
Nucleic Acids Res ; 43(4): 2177-87, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25653158

RESUMEN

Eukaryotic cells carry two genomes, nuclear (nDNA) and mitochondrial (mtDNA), which are ostensibly decoupled in their replication, segregation and inheritance. It is increasingly appreciated that heteroplasmy, the occurrence of multiple mtDNA haplotypes in a cell, plays an important biological role, but its features are not well understood. Accurately determining the diversity of mtDNA has been difficult, due to the relatively small amount of mtDNA in each cell (<1% of the total DNA), the intercellular variability of mtDNA content and mtDNA pseudogenes (Numts) in nDNA. To understand the nature of heteroplasmy, we developed Mseek, a novel technique to purify and sequence mtDNA. Mseek yields high purity (>90%) mtDNA and its ability to detect rare variants is limited only by sequencing depth, providing unprecedented sensitivity and specificity. Using Mseek, we confirmed the ubiquity of heteroplasmy by analyzing mtDNA from a diverse set of cell lines and human samples. Applying Mseek to colonies derived from single cells, we find heteroplasmy is stably maintained in individual daughter cells over multiple cell divisions. We hypothesized that the stability of heteroplasmy could be facilitated by intercellular exchange of mtDNA. We explicitly demonstrate this exchange by co-culturing cell lines with distinct mtDNA haplotypes. Our results shed new light on the maintenance of heteroplasmy and provide a novel platform to investigate features of heteroplasmy in normal and diseased states.


Asunto(s)
ADN Mitocondrial/metabolismo , Análisis de Secuencia de ADN/métodos , Transporte Biológico , Línea Celular , Línea Celular Tumoral , ADN Mitocondrial/química , ADN Mitocondrial/aislamiento & purificación , Variación Genética , Haplotipos , Humanos
10.
PLoS Genet ; 9(8): e1003603, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23966864

RESUMEN

The role of Wnt signaling in embryonic development and stem cell maintenance is well established and aberrations leading to the constitutive up-regulation of this pathway are frequent in several types of human cancers. Upon ligand-mediated activation, Wnt receptors promote the stabilization of ß-catenin, which translocates to the nucleus and binds to the T-cell factor/lymphoid enhancer factor (TCF/LEF) family of transcription factors to regulate the expression of Wnt target genes. When not bound to ß-catenin, the TCF/LEF proteins are believed to act as transcriptional repressors. Using a specific lentiviral reporter, we identified hematopoietic tumor cells displaying constitutive TCF/LEF transcriptional activation in the absence of ß-catenin stabilization. Suppression of TCF/LEF activity in these cells mediated by an inducible dominant-negative TCF4 (DN-TCF4) inhibited both cell growth and the expression of Wnt target genes. Further, expression of TCF1 and LEF1, but not TCF4, stimulated TCF/LEF reporter activity in certain human cell lines independently of ß-catenin. By a complementary approach in vivo, TCF1 mutants, which lacked the ability to bind to ß-catenin, induced Xenopus embryo axis duplication, a hallmark of Wnt activation, and the expression of the Wnt target gene Xnr3. Through generation of different TCF1-TCF4 fusion proteins, we identified three distinct TCF1 domains that participate in the ß-catenin-independent activity of this transcription factor. TCF1 and LEF1 physically interacted and functionally synergized with members of the activating transcription factor 2 (ATF2) family of transcription factors. Moreover, knockdown of ATF2 expression in lymphoma cells phenocopied the inhibitory effects of DN-TCF4 on the expression of target genes associated with the Wnt pathway and on cell growth. Together, our findings indicate that, through interaction with ATF2 factors, TCF1/LEF1 promote the growth of hematopoietic malignancies in the absence of ß-catenin stabilization, thus establishing a new mechanism for TCF1/LEF1 transcriptional activity distinct from that associated with canonical Wnt signaling.


Asunto(s)
Factor de Transcripción Activador 2/genética , Carcinogénesis/genética , Factor Nuclear 1-alfa del Hepatocito/genética , Neoplasias/genética , beta Catenina/genética , Factor de Transcripción Activador 2/metabolismo , Animales , Línea Celular Tumoral , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Humanos , Neoplasias/patología , Regiones Promotoras Genéticas , Transducción de Señal , Activación Transcripcional/genética , Vía de Señalización Wnt/genética , Xenopus laevis
12.
Proc Natl Acad Sci U S A ; 108(47): 18937-42, 2011 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-22084066

RESUMEN

p53 functions as a central node for organizing whether the cell responds to stress with apoptosis or cell cycle arrest; however, the molecular events that lead to apoptotic responses are not completely understood. Here, we identified p90 (also called Coiled-Coil Domain Containing 8) as a unique regulator for p53. p90 has no obvious effects on either the levels of p53 or p53-mediated cell cycle arrest but is specifically required for p53-mediated apoptosis upon DNA damage. Notably, p90 is crucial for Tip60-dependent p53 acetylation at Lys120, therefore facilitating activation of the proapoptotic targets. These studies indicate that p90 is a critical cofactor for p53-mediated apoptosis through promoting Tip60-mediated p53 acetylation.


Asunto(s)
Apoptosis/genética , Proteínas Portadoras/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Daño del ADN , Histona Acetiltransferasas/metabolismo , Complejos Multiproteicos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Secuencia de Aminoácidos , Secuencia de Bases , Western Blotting , Proteínas Portadoras/genética , Línea Celular , Humanos , Inmunoprecipitación , Lisina Acetiltransferasa 5 , Datos de Secuencia Molecular , Complejos Multiproteicos/genética , Oligonucleótidos/genética , Interferencia de ARN , Análisis de Secuencia de ADN
14.
Ann Surg ; 257(3): 548-54, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23011390

RESUMEN

OBJECTIVE: To determine the incidence of Wnt pathway activation in patients with stage I NSCLC and its influence on lung cancer recurrence. BACKGROUND: Despite resection, the 5-year recurrence with localized stage I nonsmall cell lung cancer (NSCLC) is 18.4%-24%. Aberrant Wnt signaling activation plays an important role in a wide variety of tumor types. However, there is not much known about the role the Wnt pathway plays in patients with stage I lung cancer. METHODS: Tumor and normal lung tissues from 55 patients following resection for stage I NSCLC were subjected to glutathione S-transferase (GST) E-cadherin pulldown and immunoblot analysis to assess levels of uncomplexed ß-catenin, a reliable measure of Wnt signaling activation. The ß-catenin gene was also screened for oncogenic mutations in tumors with activated Wnt signaling. Cancer recurrence rates were correlated in a blinded manner in patients with Wnt pathway-positive and -negative tumors. RESULTS: Tumors in 20 patients (36.4%) scored as Wnt positive, with only 1 exhibiting a ß-catenin oncogenic mutation. Patients with Wnt-positive tumors experienced a significantly higher rate of overall cancer recurrence than those with Wnt-negative tumors (30.0% vs. 5.7%, P = 0.02), with 25.0% exhibiting distal tumor recurrence compared with 2.9% in the Wnt-negative group (P = 0.02). CONCLUSIONS: Wnt pathway activation occurred in a substantial fraction of Stage I NSCLCs, which was rarely due to mutations. Moreover, Wnt pathway activation was associated with a significantly higher rate of tumor recurrence. These findings suggest that Wnt pathway activation reflects a more aggressive tumor phenotype and identifies patients who may benefit from more aggressive therapy in addition to resection.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares/genética , Recurrencia Local de Neoplasia/genética , Estadificación de Neoplasias , Proteínas Wnt/genética , Anciano , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , ADN de Neoplasias/genética , Femenino , Humanos , Incidencia , Pulmón/metabolismo , Pulmón/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Masculino , Mutación , Recurrencia Local de Neoplasia/epidemiología , Recurrencia Local de Neoplasia/metabolismo , Estudios Retrospectivos , Transducción de Señal/genética , Estados Unidos/epidemiología , Proteínas Wnt/metabolismo
15.
J Immunol ; 187(12): 6428-36, 2011 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-22105999

RESUMEN

Several direct target genes of the p53 tumor suppressor have been identified within pathways involved in viral sensing, cytokine production, and inflammation, suggesting a potential role of p53 in antiviral immunity. The increasing need to identify immune factors to devise host-targeted therapies against pandemic influenza A virus (IAV) led us to investigate the role of endogenous wild-type p53 on the immune response to IAV. We observed that the absence of p53 resulted in delayed cytokine and antiviral gene responses in lung and bone marrow, decreased dendritic cell activation, and reduced IAV-specific CD8(+) T cell immunity. Consequently, p53(-/-) mice showed a more severe IAV-induced disease compared with their wild-type counterparts. These findings establish that p53 influences the antiviral response to IAV, affecting both innate and adaptive immunity. Thus, in addition to its established functions as a tumor suppressor gene, p53 serves as an IAV host antiviral factor that might be modulated to improve anti-IAV therapy and vaccines.


Asunto(s)
Inmunidad Adaptativa , Regulación Viral de la Expresión Génica/inmunología , Inmunidad Innata , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H3N2 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Proteína p53 Supresora de Tumor/fisiología , Proteínas Reguladoras y Accesorias Virales/fisiología , Inmunidad Adaptativa/genética , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Dendríticas/patología , Inmunidad Innata/genética , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/patología , Neumonía Viral/inmunología , Neumonía Viral/metabolismo , Neumonía Viral/patología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteínas Reguladoras y Accesorias Virales/deficiencia , Proteínas Reguladoras y Accesorias Virales/genética
16.
Cell Death Differ ; 30(5): 1097-1154, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37100955

RESUMEN

Apoptosis is a form of regulated cell death (RCD) that involves proteases of the caspase family. Pharmacological and genetic strategies that experimentally inhibit or delay apoptosis in mammalian systems have elucidated the key contribution of this process not only to (post-)embryonic development and adult tissue homeostasis, but also to the etiology of multiple human disorders. Consistent with this notion, while defects in the molecular machinery for apoptotic cell death impair organismal development and promote oncogenesis, the unwarranted activation of apoptosis promotes cell loss and tissue damage in the context of various neurological, cardiovascular, renal, hepatic, infectious, neoplastic and inflammatory conditions. Here, the Nomenclature Committee on Cell Death (NCCD) gathered to critically summarize an abundant pre-clinical literature mechanistically linking the core apoptotic apparatus to organismal homeostasis in the context of disease.


Asunto(s)
Apoptosis , Caspasas , Animales , Humanos , Apoptosis/genética , Muerte Celular , Caspasas/genética , Caspasas/metabolismo , Carcinogénesis , Mamíferos/metabolismo
17.
J Biol Chem ; 286(20): 17672-81, 2011 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-21398698

RESUMEN

DDR1 (discoidin domain receptor tyrosine kinase 1) kinase s highly expressed in a variety of human cancers and occasionally mutated in lung cancer and leukemia. It is now clear that aberrant signaling through the DDR1 receptor is closely associated with various steps of tumorigenesis, although little is known about the molecular mechanism(s) underlying the role of DDR1 in cancer. Besides the role of DDR1 in tumorigenesis, we previously identified DDR1 kinase as a transcriptional target of tumor suppressor p53. DDR1 is functionally activated as determined by its tyrosine phosphorylation, in response to p53-dependent DNA damage. In this study, we report the characterization of the Notch1 protein as an interacting partner of DDR1 receptor, as determined by tandem affinity protein purification. Upon ligand-mediated DDR1 kinase activation, Notch1 was activated, bound to DDR1, and activated canonical Notch1 targets, including Hes1 and Hey2. Moreover, DDR1 ligand (collagen I) treatment significantly increased the active form of Notch1 receptor in the nuclear fraction, whereas DDR1 knockdown cells show little or no increase of the active form of Notch1 in the nuclear fraction, suggesting a novel intracellular mechanism underlying autocrine activation of wild-type Notch signaling through DDR1. DDR1 activation suppressed genotoxic-mediated cell death, whereas Notch1 inhibition by a γ-secretase inhibitor, DAPT, enhanced cell death in response to stress. Moreover, the DDR1 knockdown cancer cells showed the reduced transformed phenotypes in vitro and in vivo xenograft studies. The results suggest that DDR1 exerts prosurvival effect, at least in part, through the functional interaction with Notch1.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor Notch1/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Núcleo Celular/genética , Núcleo Celular/metabolismo , Supervivencia Celular/fisiología , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Daño del ADN/fisiología , Dipéptidos/farmacología , Receptor con Dominio Discoidina 1 , Activación Enzimática/fisiología , Técnicas de Silenciamiento del Gen , Células HEK293 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Fosforilación/fisiología , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/genética , Receptor Notch1/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor de Transcripción HES-1 , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
18.
J Cell Sci ; 123(Pt 15): 2605-12, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20605919

RESUMEN

Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature-aging syndrome caused by a dominant mutation in the gene encoding lamin A, which leads to an aberrantly spliced and processed protein termed progerin. Previous studies have shown that progerin induces early senescence associated with increased DNA-damage signaling and that telomerase extends HGPS cellular lifespan. We demonstrate that telomerase extends HGPS cellular lifespan by decreasing progerin-induced DNA-damage signaling and activation of p53 and Rb pathways that otherwise mediate the onset of premature senescence. We show further that progerin-induced DNA-damage signaling is localized to telomeres and is associated with telomere aggregates and chromosomal aberrations. Telomerase amelioration of DNA-damage signaling is relatively rapid, requires both its catalytic and DNA-binding functions, and correlates in time with the acquisition by HGPS cells of the ability to proliferate. All of these findings establish that HGPS premature cellular senescence results from progerin-induced telomere dysfunction.


Asunto(s)
Senescencia Celular/fisiología , Proteínas Nucleares/metabolismo , Progeria/metabolismo , Precursores de Proteínas/metabolismo , Telomerasa/metabolismo , Telómero/metabolismo , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/fisiopatología , Línea Celular , Senescencia Celular/genética , Inmunoprecipitación de Cromatina , Daño del ADN/genética , Daño del ADN/fisiología , Citometría de Flujo , Humanos , Immunoblotting , Lamina Tipo A , Microscopía Confocal , Proteínas Nucleares/genética , Progeria/genética , Precursores de Proteínas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología , Telomerasa/genética
19.
Cancer Cell ; 6(5): 497-506, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15542433

RESUMEN

Autocrine Wnt signaling in the mouse mammary tumor virus model was the first identified mechanism of canonical pathway activation in cancer. In search of this transformation mechanism in human cancer cells, we identified breast and ovarian tumor lines with upregulation of the uncomplexed transcriptionally active form of beta-catenin without mutations afflicting downstream components. Extracellular Wnt antagonists FRP1 and DKK1 caused a dramatic downregulation of beta-catenin levels in these tumor cells associated with alteration of biological properties and increased expression of epithelial differentiation markers. Colorectal carcinoma cells with knockout of the mutant beta-catenin allele retained upregulated beta-catenin levels, which also could be inhibited by these Wnt antagonists. Together, these findings establish the involvement of autocrine Wnt signaling in human cancer cells.


Asunto(s)
Neoplasias/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas de la Ataxia Telangiectasia Mutada , Comunicación Autocrina/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Proteínas de Ciclo Celular/farmacología , Neoplasias del Colon , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular , Proteína Reguladora de Respuesta a la Leucina , Neoplasias Ováricas/metabolismo , Proteínas Serina-Treonina Quinasas/farmacología , Proteínas/farmacología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Transducción de Señal , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Proteínas Wnt , beta Catenina
20.
Nat Commun ; 13(1): 703, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-35121738

RESUMEN

Rho family mechano-signaling through the actin cytoskeleton positively regulates physiological TEAD/YAP transcription, while the evolutionarily conserved Hippo tumor suppressor pathway antagonizes this transcription through YAP cytoplasmic localization/degradation. The mechanisms responsible for oncogenic dysregulation of these pathways, their prevalence in tumors, as well as how such dysregulation can be therapeutically targeted are not resolved. We demonstrate that p53 DNA contact mutants in human tumors, indirectly hyperactivate RhoA/ROCK1/actomyosin signaling, which is both necessary and sufficient to drive oncogenic TEAD/YAP transcription. Moreover, we demonstrate that recurrent lesions in the Hippo pathway depend on physiological levels of ROCK1/actomyosin signaling for oncogenic TEAD/YAP transcription. Finally, we show that ROCK inhibitors selectively antagonize proliferation and motility of human tumors with either mechanism. Thus, we identify a cancer driver paradigm and a precision medicine approach for selective targeting of human malignancies driven by TEAD/YAP transcription through mechanisms that either upregulate or depend on homeostatic RhoA mechano-signaling.


Asunto(s)
Proteínas de Ciclo Celular/genética , Neoplasias/genética , Transducción de Señal/genética , Factores de Transcripción de Dominio TEA/genética , Factores de Transcripción/genética , Quinasas Asociadas a rho/genética , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Vía de Señalización Hippo/efectos de los fármacos , Vía de Señalización Hippo/genética , Humanos , Ratones SCID , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Factores de Transcripción de Dominio TEA/metabolismo , Factores de Transcripción/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA