Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Endocrinol (Lausanne) ; 15: 1312357, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38654924

RESUMEN

RASopathies are syndromes caused by congenital defects in the Ras/mitogen-activated protein kinase (MAPK) pathway genes, with a population prevalence of 1 in 1,000. Patients are typically identified in childhood based on diverse characteristic features, including cryptorchidism (CR) in >50% of affected men. As CR predisposes to spermatogenic failure (SPGF; total sperm count per ejaculate 0-39 million), we hypothesized that men seeking infertility management include cases with undiagnosed RASopathies. Likely pathogenic or pathogenic (LP/P) variants in 22 RASopathy-linked genes were screened in 521 idiopathic SPGF patients (including 155 CR cases) and 323 normozoospermic controls using exome sequencing. All 844 men were recruited to the ESTonian ANDrology (ESTAND) cohort and underwent identical andrological phenotyping. RASopathy-specific variant interpretation guidelines were used for pathogenicity assessment. LP/P variants were identified in PTPN11 (two), SOS1 (three), SOS2 (one), LZTR1 (one), SPRED1 (one), NF1 (one), and MAP2K1 (one). The findings affected six of 155 cases with CR and SPGF, three of 366 men with SPGF only, and one (of 323) normozoospermic subfertile man. The subgroup "CR and SPGF" had over 13-fold enrichment of findings compared to controls (3.9% vs. 0.3%; Fisher's exact test, p = 5.5 × 10-3). All ESTAND subjects with LP/P variants in the Ras/MAPK pathway genes presented congenital genitourinary anomalies, skeletal and joint conditions, and other RASopathy-linked health concerns. Rare forms of malignancies (schwannomatosis and pancreatic and testicular cancer) were reported on four occasions. The Genetics of Male Infertility Initiative (GEMINI) cohort (1,416 SPGF cases and 317 fertile men) was used to validate the outcome. LP/P variants in PTPN11 (three), LZTR1 (three), and MRAS (one) were identified in six SPGF cases (including 4/31 GEMINI cases with CR) and one normozoospermic man. Undiagnosed RASopathies were detected in total for 17 ESTAND and GEMINI subjects, 15 SPGF patients (10 with CR), and two fertile men. Affected RASopathy genes showed high expression in spermatogenic and testicular somatic cells. In conclusion, congenital defects in the Ras/MAPK pathway genes represent a new congenital etiology of syndromic male infertility. Undiagnosed RASopathies were especially enriched among patients with a history of cryptorchidism. Given the relationship between RASopathies and other conditions, infertile men found to have this molecular diagnosis should be evaluated for known RASopathy-linked health concerns, including specific rare malignancies.


Asunto(s)
Infertilidad Masculina , Humanos , Masculino , Infertilidad Masculina/genética , Infertilidad Masculina/diagnóstico , Adulto , Proteínas ras/genética , Criptorquidismo/genética , Criptorquidismo/complicaciones , Secuenciación del Exoma , Mutación
2.
Int J Cancer ; 141(10): 2002-2013, 2017 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-28710771

RESUMEN

Intratumoral heterogeneity at the genetic, epigenetic, transcriptomic, and morphologic levels is a commonly observed phenomenon in many aggressive cancer types. Clonal evolution during tumor formation and in response to therapeutic intervention can be predicted utilizing reverse engineering approaches on detailed genomic snapshots of heterogeneous patient tumor samples. In this study, we developed an extensive dataset for a GBM case via the generation of polyclonal and monoclonal glioma stem cell lines from initial diagnosis, and from multiple sections of distant tumor locations of the deceased patient's brain following tumor recurrence. Our analyses revealed the tissue-wide expansion of a new clone in the recurrent tumor and chromosome 7 gain and chromosome 10 loss as repeated genomic events in primary and recurrent disease. Moreover, chromosome 7 gain and chromosome 10 loss produced similar alterations in mRNA expression profiles in primary and recurrent tumors despite possessing other highly heterogeneous and divergent genomic alterations between the tumors. We identified ETV1 and CDK6 as putative candidate genes, and NFKB (complex), IL1B, IL6, Akt and VEGF as potential signaling regulators, as potentially central downstream effectors of chr7 gain and chr10 loss. Finally, the differences caused by the transcriptomic shift following gain of chromosome 7 and loss of chromosome 10 were consistent with those generally seen in GBM samples compared to normal brain in large-scale patient-tumor data sets.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Cromosomas Humanos Par 10/genética , Cromosomas Humanos Par 7/genética , Glioma/genética , Recurrencia Local de Neoplasia/genética , Células Madre Neoplásicas/metabolismo , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Aberraciones Cromosómicas , Perfilación de la Expresión Génica , Genómica/métodos , Glioma/patología , Xenoinjertos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/patología , Pronóstico
3.
Sci Rep ; 7: 43605, 2017 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-28256619

RESUMEN

Glioblastoma, the most common primary malignant brain tumor, harbors a small population of tumor initiating cells (glioblastoma stem cells) that have many properties similar to neural stem cells. To investigate common regulatory networks in both neural and glioblastoma stem cells, we subjected both cell types to in-vitro differentiation conditions and measured global gene-expression changes using gene expression microarrays. Analysis of enriched transcription factor DNA-binding sites in the promoters of differentially expressed genes was used to reconstruct regulatory networks involved in differentiation. Computational predictions, which were biochemically validated, show an extensive overlap of regulatory circuitry between cell types including a network centered on the transcription factor KLF4. We further demonstrate that EGR1, a transcription factor previously shown to be downstream of the MAPK pathway, regulates KLF4 expression and that KLF4 in turn transcriptionally activates NOTCH as well as SOX2. These results demonstrate how known genomic alterations in glioma that induce constitutive activation of MAPK are transcriptionally linked to master regulators essential for neural stem cell identify.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Células Madre Neoplásicas/metabolismo , Células-Madre Neurales/metabolismo , Animales , Sitios de Unión , Biomarcadores , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Biología Computacional/métodos , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Estudio de Asociación del Genoma Completo , Glioblastoma/patología , Humanos , Factor 4 Similar a Kruppel , Ratones , Clasificación del Tumor , Unión Proteica , Transducción de Señal , Factores de Transcripción/metabolismo , Transcriptoma
4.
PLoS One ; 9(11): e111783, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25365423

RESUMEN

Gliomas are mostly incurable secondary to their diffuse infiltrative nature. Thus, specific therapeutic targeting of invasive glioma cells is an attractive concept. As cells exit the tumor mass and infiltrate brain parenchyma, they closely interact with a changing micro-environmental landscape that sustains tumor cell invasion. In this study, we used a unique microarray profiling approach on a human glioma stem cell (GSC) xenograft model to explore gene expression changes in situ in Invading Glioma Cells (IGCs) compared to tumor core, as well as changes in host cells residing within the infiltrated microenvironment relative to the unaffected cortex. IGCs were found to have reduced expression of genes within the extracellular matrix compartment, and genes involved in cell adhesion, cell polarity and epithelial to mesenchymal transition (EMT) processes. The infiltrated microenvironment showed activation of wound repair and tissue remodeling networks. We confirmed by protein analysis the downregulation of EMT and polarity related genes such as CD44 and PARD3 in IGCs, and EFNB3, a tissue-remodeling agent enriched at the infiltrated microenvironment. OLIG2, a proliferation regulator and glioma progenitor cell marker upregulated in IGCs was found to function in enhancing migration and stemness of GSCs. Overall, our results unveiled a more comprehensive picture of the complex and dynamic cell autonomous and tumor-host interactive pathways of glioma invasion than has been previously demonstrated. This suggests targeting of multiple pathways at the junction of invading tumor and microenvironment as a viable option for glioma therapy.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Proteínas de Neoplasias/biosíntesis , Microambiente Tumoral , Adulto , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Femenino , Glioma/genética , Glioma/patología , Xenoinjertos , Humanos , Masculino , Ratones , Ratones SCID , Invasividad Neoplásica , Proteínas de Neoplasias/genética , Trasplante de Neoplasias
5.
PLoS One ; 9(4): e94045, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24728236

RESUMEN

In vitro and in vivo models are widely used in cancer research. Characterizing the similarities and differences between a patient's tumor and corresponding in vitro and in vivo models is important for understanding the potential clinical relevance of experimental data generated with these models. Towards this aim, we analyzed the genomic aberrations, DNA methylation and transcriptome profiles of five parental tumors and their matched in vitro isolated glioma stem cell (GSC) lines and xenografts generated from these same GSCs using high-resolution platforms. We observed that the methylation and transcriptome profiles of in vitro GSCs were significantly different from their corresponding xenografts, which were actually more similar to their original parental tumors. This points to the potentially critical role of the brain microenvironment in influencing methylation and transcriptional patterns of GSCs. Consistent with this possibility, ex vivo cultured GSCs isolated from xenografts showed a tendency to return to their initial in vitro states even after a short time in culture, supporting a rapid dynamic adaptation to the in vitro microenvironment. These results show that methylation and transcriptome profiles are highly dependent on the microenvironment and growth in orthotopic sites partially reverse the changes caused by in vitro culturing.


Asunto(s)
Glioma/genética , Células Madre Neoplásicas/metabolismo , Animales , Metilación de ADN/genética , Metilación de ADN/fisiología , Femenino , Humanos , Técnicas In Vitro , Ratones , Ratones SCID , Polimorfismo de Nucleótido Simple/genética , Análisis de Componente Principal , Estudios Prospectivos , Células Tumorales Cultivadas
6.
PLoS One ; 7(12): e51407, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23236496

RESUMEN

Histone methylation regulates normal stem cell fate decisions through a coordinated interplay between histone methyltransferases and demethylases at lineage specific genes. Malignant transformation is associated with aberrant accumulation of repressive histone modifications, such as polycomb mediated histone 3 lysine 27 (H3K27me3) resulting in a histone methylation mediated block to differentiation. The relevance, however, of histone demethylases in cancer remains less clear. We report that JMJD3, a H3K27me3 demethylase, is induced during differentiation of glioblastoma stem cells (GSCs), where it promotes a differentiation-like phenotype via chromatin dependent (INK4A/ARF locus activation) and chromatin independent (nuclear p53 protein stabilization) mechanisms. Our findings indicate that deregulation of JMJD3 may contribute to gliomagenesis via inhibition of the p53 pathway resulting in a block to terminal differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Glioblastoma/fisiopatología , Histona Demetilasas con Dominio de Jumonji/metabolismo , Células Madre Neoplásicas/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Western Blotting , Cartilla de ADN/genética , Histonas/metabolismo , Humanos , Inmunohistoquímica , Inmunoprecipitación , Luciferasas , Espectrometría de Masas , Ratones , Ratones SCID , Estabilidad Proteica , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
J Natl Cancer Inst ; 103(15): 1162-78, 2011 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-21771732

RESUMEN

BACKGROUND: Tumor cell invasion is the principal cause of treatment failure and death among patients with malignant gliomas. Connective tissue growth factor (CTGF) has been previously implicated in cancer metastasis and invasion in various tumors. We explored the mechanism of CTGF-mediated glioma cell infiltration and examined potential therapeutic targets. METHODS: Highly infiltrative patient-derived glioma tumor-initiating or tumor stem cells (TIC/TSCs) were harvested and used to explore a CTGF-induced signal transduction pathway via luciferase reporter assays, chromatin immunoprecipitation (ChIP), real-time polymerase chain reaction, and immunoblotting. Treatment of TIC/TSCs with small-molecule inhibitors targeting integrin ß1 (ITGB1) and the tyrosine kinase receptor type A (TrkA), and short hairpin RNAs targeting CTGF directly were used to reduce the levels of key protein components of CTGF-induced cancer infiltration. TIC/TSC infiltration was examined in real-time cell migration and invasion assays in vitro and by immunohistochemistry and in situ hybridization in TIC/TSC orthotopic xenograft mouse models (n = 30; six mice per group). All statistical tests were two-sided. RESULTS: Treatment of TIC/TSCs with CTGF resulted in CTGF binding to ITGB1-TrkA receptor complexes and nuclear factor kappa B (NF-κB) transcriptional activation as measured by luciferase reporter assays (mean relative luciferase activity, untreated vs CTGF(200 ng/mL): 0.53 vs 1.87, difference = 1.34, 95% confidence interval [CI] = 0.69 to 2, P < .001). NF-κB activation resulted in binding of ZEB-1 to the E-cadherin promoter as demonstrated by ChIP analysis with subsequent E-cadherin suppression (fold increase in ZEB-1 binding to the E-cadherin promoter region: untreated + ZEB-1 antibody vs CTGF(200 ng/mL) + ZEB-1 antibody: 1.5 vs 6.4, difference = 4.9, 95% CI = 4.8 to 5.0, P < .001). Immunohistochemistry and in situ hybridization revealed that TrkA is selectively expressed in the most infiltrative glioma cells in situ and that the surrounding reactive astrocytes secrete CTGF. CONCLUSION: A CTGF-rich microenvironment facilitates CTGF-ITGB1-TrkA complex activation in TIC/TSCs, thereby increasing the invasiveness of malignant gliomas.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Glioma/metabolismo , Glioma/patología , Proteínas de Homeodominio/metabolismo , Integrina beta1/metabolismo , FN-kappa B/metabolismo , Células Madre Neoplásicas/metabolismo , Receptor trkA/metabolismo , Factores de Transcripción/metabolismo , Animales , Unión Competitiva , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular , Inmunoprecipitación de Cromatina , Modelos Animales de Enfermedad , Activación Enzimática , Citometría de Flujo , Transferencia Resonante de Energía de Fluorescencia , Técnica del Anticuerpo Fluorescente , Humanos , Immunoblotting , Inmunohistoquímica , Hibridación in Situ , Radioisótopos de Yodo , Luciferasas/metabolismo , Ratones , Microscopía Confocal , Invasividad Neoplásica , Factor de Crecimiento Nervioso/metabolismo , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Trasplante Heterólogo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
8.
Cancer Res ; 70(10): 4112-22, 2010 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-20460517

RESUMEN

The integrity of DNA is constantly challenged throughout the life of a cell by both endogenous and exogenous stresses. A well-organized rapid damage response and proficient DNA repair, therefore, become critically important for maintaining genomic stability and cell survival. When DNA is damaged, the DNA damage response (DDR) can be initiated by alterations in chromosomal structure and histone modifications, such as the phosphorylation of the histone H2AX (the phosphorylated form is referred to as gamma-H2AX). gamma-H2AX plays a crucial role in recruiting DDR factors to damage sites for accurate DNA repair. On repair completion, gamma-H2AX must then be reverted to H2AX by dephosphorylation for attenuation of the DDR. Here, we report that the wild-type p53-induced phosphatase 1 (Wip1) phosphatase, which is often overexpressed in a variety of tumors, effectively dephosphorylates gamma-H2AX in vitro and in vivo. Ectopic expression of Wip1 significantly reduces the level of gamma-H2AX after ionizing as well as UV radiation. Forced premature dephosphorylation of gamma-H2AX by Wip1 disrupts recruitment of important DNA repair factors to damaged sites and delays DNA damage repair. Additionally, deletion of Wip1 enhances gamma-H2AX levels in cells undergoing constitutive oncogenic stress. Taken together, our studies show that Wip1 is an important mammalian phosphatase for gamma-H2AX and shows an additional mechanism for Wip1 in the tumor surveillance network.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias del Colon/patología , Daño del ADN , Embrión de Mamíferos/patología , Fibroblastos/patología , Histonas/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Animales , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Células Cultivadas , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Ensayo Cometa , Reparación del ADN , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación , Rayos Infrarrojos , Ratones , Mutagénesis Sitio-Dirigida , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/genética , Fosforilación , Proteína Fosfatasa 2C , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Rayos Ultravioleta , Rayos X
9.
J Biol Chem ; 283(13): 8080-8, 2008 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-18218636

RESUMEN

The nucleosome-binding protein HMGN1 affects the structure and function of chromatin; however, its role in regulating specific gene expression in living cells is not fully understood. Here we use embryonic fibroblasts from Hmgn1(+/+) and Hmgn1(-/-) mice to examine the effect of HMGN1 on the heat shock-induced transcriptional activation of Hsp70, a well characterized gene known to undergo a rapid chromatin re-structuring during transcriptional activation. We find that loss of HMGN1 decreases the levels of Hsp70 transcripts at the early stages of heat shock. HMGN1 enhances the rate of heat shockinduced changes in the Hsp70 chromatin but does not affect the chromatin structure before induction, an indication that it does not predispose the gene to rapid activation. Heat shock elevates the levels of H3K14 acetylation in the Hsp70 chromatin of wild type cells more efficiently than in the chromatin of Hmgn1(-/-) cells, whereas treatment with histone deacetylase inhibitors abrogates the effects of HMGN1 on the heat shock response. We suggest that HMGN1 enhances the rate of heat shock-induced H3K14 acetylation in the Hsp70 promoter, thereby enhancing the rate of chromatin remodeling and the subsequent transcription during the early rounds of Hsp70 activation when the gene is still associated with histones in a nucleosomal conformation.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Cromosomas/metabolismo , Proteína HMGN1/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Respuesta al Choque Térmico , Acetilación , Animales , Células Cultivadas , Cromosomas/genética , Proteína HMGN1/deficiencia , Proteína HMGN1/genética , Proteínas HSP70 de Choque Térmico/genética , Histonas/metabolismo , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/genética
10.
Biochemistry ; 45(50): 15092-9, 2006 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-17154547

RESUMEN

Here we demonstrate that HMGN1, a nuclear protein that binds specifically to nucleosomes, modulates the level of histone H2A phosphorylation. In Hmgn1-/- cells, loss of HMGN1 elevates the steady-state levels of H2AS1ph throughout the cell cycle. In vitro, HMGN1 reduces the rate of Rsk2- and Msk1-mediated phosphorylation of nucleosomal, but not free, histone H2A. HMGN1 inhibits H2A phosphorylation by binding to nucleosomes since an HMGN mutant, which cannot bind to chromatin, does not inhibit the Rsk2- mediated H2A phosphorylation. HMGN2 also inhibits H2A phosphorylation, suggesting that the inhibition of H2A phosphorylation is not specific to only one member of this protein family. Thus, the present data add modifications of histone H2A to the list of histone modifications affected by HMGN proteins. It supports the suggestion that structural chromatin binding proteins can modify the whole profile of post-translational modifications of core histones.


Asunto(s)
Proteína HMGN1/metabolismo , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Animales , Ciclo Celular/genética , Línea Celular , Proteína HMGN1/genética , Ratones , Mutación , Nucleosomas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Serina/metabolismo
11.
Cancer Biol Ther ; 4(10): 1154-8, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16258255

RESUMEN

Wip1 is an amplified oncogene whose deletion causes a tumor resistant phenotype in mice. These observations provide justification for a search for Wip1 chemical inhibitors as potential anticancer drugs. Here we report a group of Wip1 inhibitors with anticancer properties both in vitro and in vivo. In vitro, inactivation of Wip1 reduces the proliferation rate of breast cancer cell lines and enhances growth inhibition caused by doxorubicin. In vivo, administration of Wip1 inhibitors decreases proliferation of xenograph tumors and tumors developed in MMTV-c-Neu transgenic mice. We propose that these agents may serve as lead compounds for the development of anticancer drugs targeting Wip1 phosphatase.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Adenoviridae/genética , Animales , Neoplasias de la Mama/prevención & control , Línea Celular Transformada , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transformación Celular Viral , Embrión de Mamíferos/citología , Inhibidores Enzimáticos/química , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Genes ras , Humanos , Ratones , Ratones Transgénicos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Conformación Molecular , Trasplante de Neoplasias , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/aislamiento & purificación , Fosforilación/efectos de los fármacos , Proteína Fosfatasa 2C , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Nat Immunol ; 6(4): 396-402, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15735649

RESUMEN

The p38 MAP kinase (MAPK) is phosphorylated and activated by upstream MAPK kinases. T cells have an alternative pathway in which T cell receptor-activated tyrosine kinase Zap70 phosphorylates p38 on Tyr323. Mice lacking Gadd45alpha, a small p38-binding molecule, develop a lupus-like autoimmune disease. Here we show that resting T cells but not B cells from Gadd45a(-/-) mice had spontaneously increased p38 activity in the absence of 'upstream' MAPK kinase activation. The p38 from resting Gadd45a(-/-) T cells was spontaneously phosphorylated on Tyr323, and its activity was specifically inhibited by recombinant Gadd45alpha in vitro. Thus, constitutive activation of T cell p38 through the alternative pathway is prevented by Gadd45alpha, the absence of which results in p38 activation, T cell hyperproliferation and autoimmunity.


Asunto(s)
Proteínas de Ciclo Celular/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Proteínas Nucleares/inmunología , Linfocitos T/enzimología , Linfocitos T/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Animales , Proteínas de Ciclo Celular/metabolismo , Activación Enzimática , Ratones , Ratones Noqueados , Proteínas Nucleares/metabolismo , Fosforilación , Proteínas Tirosina Quinasas/inmunología , Proteínas Tirosina Quinasas/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Tirosina/inmunología , Tirosina/metabolismo , Proteína Tirosina Quinasa ZAP-70 , Proteínas Quinasas p38 Activadas por Mitógenos/inmunología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
13.
Oncogene ; 23(10): 1829-37, 2004 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-14647429

RESUMEN

The p53-regulated growth arrest and DNA damage-inducible gene product Gadd45a has been recently identified as a key factor protecting the epidermis against ultraviolet radiation (UVR)-induced skin tumors by activating p53 via the stress mitogen-activated protein kinase (MAPK) signaling pathway. Herein we identify Gadd45a as an important negative regulator of two oncogenes commonly over-expressed in epithelial tumors: the p53 homologue DeltaNp63alpha and beta-catenin. DeltaNp63alpha is one of the several p63 isoforms and is the predominant species expressed in basal epidermal keratinocytes. DeltaNp63alpha lacks the N-terminal transactivation domain and behaves as a dominant-negative factor blocking expression of several p53-effector genes. DeltaNp63alpha also associates with and blocks activation of the adenomatous polyposis coli (APC) destruction complex that targets free cytoplasmic beta-catenin for degradation. While most beta-catenin protein is localized to the cell membrane and is involved in cell-cell adhesion, accumulation of free cytoplasmic beta-catenin will translocate into the nucleus where it functions in a bipartite transcription factor complex, whose targets include invasion and metastasis promoting endopeptidases, matrix metalloproteinases (MMP). We show that Gadd45a not only directly associates with two components of the APC complex, namely protein phosphatase 2A (PP2A) and glycogen synthase kinase 3beta (GSK3beta) but also promotes GSK3beta dephosphorylation at Ser9, which is essential for GSK3beta activation, and resultant activation of the APC destruction complex. We demonstrate that lack of Gadd45a not only prevents DeltaNp63alpha suppression and GSK3beta dephosphorylation but also prevents free cytoplasmic beta-catenin degradation after UV irradiation. The inability of Gadd45a-null keratinocytes to suppress beta-catenin may contribute to the resulting observation of increased MMP expression and activity along with significantly faster keratinocyte migration in Matrigel in vitro and accelerated wound closure in vivo. Furthermore, epidermal keratinocytes treated with p38 MAPK inhibitors, both in vivo and in vitro, behave very similarly to Gadd45a-null keratinocytes after UVR. Similarly, Trp53-null mice are unable to attenuate DeltaNp63alpha expression in epidermal keratinocytes after such stress. These findings demonstrate a dependence on Gadd45a-mediated p38 MAPK and p53 activation for proper modulation of DeltaNp63alpha, GSK3beta, and beta-catenin after irradiation. Taken together, our results indicate that Gadd45a is able to repress DeltaNp63alpha, beta-catenin, and consequently MMP expression by two means: by maintaining UVR-induced p38 MAPK and p53 activation and also by associating with the APC complex. This implicates Gadd45a in the negative regulation of cell migration, and invasion.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Proteínas de Ciclo Celular , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Queratinocitos/citología , Metaloproteinasas de la Matriz/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas , Transactivadores/genética , Animales , Regulación de la Expresión Génica/efectos de la radiación , Queratinocitos/enzimología , Queratinocitos/efectos de la radiación , Sistema de Señalización de MAP Quinasas/genética , Ratones , Neoplasias Inducidas por Radiación/genética , Neoplasias Inducidas por Radiación/prevención & control , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/prevención & control , Rayos Ultravioleta , beta Catenina , Proteínas Quinasas p38 Activadas por Mitógenos
14.
Proc Natl Acad Sci U S A ; 99(21): 13510-5, 2002 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12368475

RESUMEN

Helix-hairpin-helix (HhH) is a widespread motif involved in sequence-nonspecific DNA binding. The majority of HhH motifs function as DNA-binding modules with typical occurrence of one HhH motif or one or two (HhH)(2) domains in proteins. We recently identified 24 HhH motifs in DNA topoisomerase V (Topo V). Although these motifs are dispensable for the topoisomerase activity of Topo V, their removal narrows the salt concentration range for topoisomerase activity tenfold. Here, we demonstrate the utility of Topo V's HhH motifs for modulating DNA-binding properties of the Stoffel fragment of TaqDNA polymerase and Pfu DNA polymerase. Different HhH cassettes fused with either NH(2) terminus or COOH terminus of DNA polymerases broaden the salt concentration range of the polymerase activity significantly (up to 0.5 M NaCl or 1.8 M potassium glutamate). We found that anions play a major role in the inhibition of DNA polymerase activity. The resistance of initial extension rates and the processivity of chimeric polymerases to salts depend on the structure of added HhH motifs. Regardless of the type of the construct, the thermal stability of chimeric Taq polymerases increases under the optimal ionic conditions, as compared with that of TaqDNA polymerase or its Stoffel fragment. Our approach to raise the salt tolerance, processivity, and thermostability of Taq and Pfu DNA polymerases may be applied to all pol1- and polB-type polymerases, as well as to other DNA processing enzymes.


Asunto(s)
ADN Polimerasa Dirigida por ADN/química , Secuencias de Aminoácidos , Secuencia de Bases , Betaína , Sitios de Unión , ADN/biosíntesis , ADN/genética , ADN-Topoisomerasas de Tipo I/química , ADN-Topoisomerasas de Tipo I/genética , ADN-Topoisomerasas de Tipo I/metabolismo , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Activación Enzimática , Estabilidad de Enzimas , Glutamatos , Técnicas In Vitro , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Análisis de Secuencia de ADN , Cloruro de Sodio , Especificidad por Sustrato , Polimerasa Taq/química , Polimerasa Taq/genética , Polimerasa Taq/metabolismo , Temperatura
15.
Proc Natl Acad Sci U S A ; 99(7): 4644-9, 2002 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-11930014

RESUMEN

We have determined the complete 1,694,969-nt sequence of the GC-rich genome of Methanopyrus kandleri by using a whole direct genome sequencing approach. This approach is based on unlinking of genomic DNA with the ThermoFidelase version of M. kandleri topoisomerase V and cycle sequencing directed by 2'-modified oligonucleotides (Fimers). Sequencing redundancy (3.3x) was sufficient to assemble the genome with less than one error per 40 kb. Using a combination of sequence database searches and coding potential prediction, 1,692 protein-coding genes and 39 genes for structural RNAs were identified. M. kandleri proteins show an unusually high content of negatively charged amino acids, which might be an adaptation to the high intracellular salinity. Previous phylogenetic analysis of 16S RNA suggested that M. kandleri belonged to a very deep branch, close to the root of the archaeal tree. However, genome comparisons indicate that, in both trees constructed using concatenated alignments of ribosomal proteins and trees based on gene content, M. kandleri consistently groups with other archaeal methanogens. M. kandleri shares the set of genes implicated in methanogenesis and, in part, its operon organization with Methanococcus jannaschii and Methanothermobacter thermoautotrophicum. These findings indicate that archaeal methanogens are monophyletic. A distinctive feature of M. kandleri is the paucity of proteins involved in signaling and regulation of gene expression. Also, M. kandleri appears to have fewer genes acquired via lateral transfer than other archaea. These features might reflect the extreme habitat of this organism.


Asunto(s)
Euryarchaeota/genética , Genoma Arqueal , Secuencia de Bases , Euryarchaeota/clasificación , Euryarchaeota/metabolismo , Datos de Secuencia Molecular , Operón , Filogenia
16.
J Biol Chem ; 277(7): 4959-65, 2002 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11733530

RESUMEN

Topoisomerase V (Topo V) is a type IB (eukaryotic-like) DNA topoisomerase. It was discovered in the hyperthermophilic prokaryote Methanopyrus kandleri and is the only topoisomerase with associated apurinic/apyrimidinic (AP) site-processing activities. The structure of Topo V in the free and DNA-bound states was probed by limited proteolysis at 37 degrees C and 80 degrees C. The Topo V protein is comprised of (i) a 44-kDa NH(2)-terminal core subdomain, which contains the active site tyrosine residue for topoisomerase activity, (ii) an immediately adjacent 16-kDa subdomain that contains degenerate helix-hairpin-helix (HhH) motifs, (iii) a protease-sensitive 18-kDa HhH "hinge" region, and (iv) a 34-kDa COOH-terminal HhH domain. Three truncated Topo V polypeptides comprising the NH(2)-terminal 44-kDa and 16-kDa domains (Topo61), the 44-, 16-, and 18-kDa domains (Topo78), and the COOH-terminal 34-kDa domain (Topo34) were cloned, purified, and characterized. Both Topo61 and Topo78 are active topoisomerases, but in contrast to Topo V these enzymes are inhibited by high salt concentrations. Topo34 has strong DNA-binding ability but shows no topoisomerase activity. Finally, we demonstrate that Topo78 and Topo34 possess AP lyase activities that are important in base excision DNA repair. Thus, Topo V has at least two active sites capable of processing AP DNA. The significance of multiple HhH motifs for the Topo V processivity is discussed.


Asunto(s)
Reparación del ADN , ADN-Topoisomerasas de Tipo I/química , Secuencias de Aminoácidos , Sitios de Unión , ADN/química , ADN/metabolismo , Escherichia coli/metabolismo , Plásmidos/metabolismo , Reacción en Cadena de la Polimerasa , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/metabolismo , Sales (Química)/farmacología , Serina Endopeptidasas/farmacología , Temperatura , Termolisina/farmacología , Tripsina/farmacología , Tirosina/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...