Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 117(32): 19415-19424, 2020 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-32719125

RESUMEN

Synthetic lethality strategies for cancer therapy exploit cancer-specific genetic defects to identify targets that are uniquely essential to the survival of tumor cells. Here we show RAD27/FEN1, which encodes flap endonuclease 1 (FEN1), a structure-specific nuclease with roles in DNA replication and repair, and has the greatest number of synthetic lethal interactions with Saccharomyces cerevisiae genome instability genes, is a druggable target for an inhibitor-based approach to kill cancers with defects in homologous recombination (HR). The vulnerability of cancers with HR defects to FEN1 loss was validated by studies showing that small-molecule FEN1 inhibitors and FEN1 small interfering RNAs (siRNAs) selectively killed BRCA1- and BRCA2-defective human cell lines. Furthermore, the differential sensitivity to FEN1 inhibition was recapitulated in mice, where a small-molecule FEN1 inhibitor reduced the growth of tumors established from drug-sensitive but not drug-resistant cancer cell lines. FEN1 inhibition induced a DNA damage response in both sensitive and resistant cell lines; however, sensitive cell lines were unable to recover and replicate DNA even when the inhibitor was removed. Although FEN1 inhibition activated caspase to higher levels in sensitive cells, this apoptotic response occurred in p53-defective cells and cell killing was not blocked by a pan-caspase inhibitor. These results suggest that FEN1 inhibitors have the potential for therapeutically targeting HR-defective cancers such as those resulting from BRCA1 and BRCA2 mutations, and other genetic defects.


Asunto(s)
Antineoplásicos/farmacología , Endonucleasas de ADN Solapado/antagonistas & inhibidores , Recombinación Homóloga/efectos de los fármacos , Neoplasias/genética , Animales , Proteína BRCA1/deficiencia , Proteína BRCA1/genética , Proteína BRCA2/deficiencia , Proteína BRCA2/genética , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Reparación del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Endonucleasas de ADN Solapado/genética , Inestabilidad Genómica/genética , Humanos , Ratones , Neoplasias/tratamiento farmacológico , ARN Interferente Pequeño/farmacología , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Bibliotecas de Moléculas Pequeñas/farmacología , Mutaciones Letales Sintéticas , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Mol Cell ; 43(4): 663-72, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21855804

RESUMEN

The LXCXE peptide motif facilitates interaction between the RB tumor suppressor and a large number of cellular proteins that are expected to impinge on diverse biological processes. In vitro and in vivo analyses demonstrated that LXCXE binding function is dispensable for RB promoter association and control of basal gene expression. Dependence on this function of RB is unmasked after DNA damage, wherein LXCXE binding is essential for exerting control over E2F3 and suppressing cell-cycle progression in the presence of genotoxic stress. Gene expression profiling revealed that the transcriptional program coordinated by this specific aspect of RB is associated with progression of human hepatocellular carcinoma and poor disease outcome. Consistent with these findings, biological challenge revealed a requirement for LXCXE binding in suppression of genotoxin-initiated hepatocellular carcinoma in vivo. Together, these studies establish an essential role of the LXCXE binding motif for RB-mediated transcriptional control, response to genotoxic insult, and tumor suppression.


Asunto(s)
Daño del ADN , Regulación de la Expresión Génica , Proteína de Retinoblastoma/fisiología , Transcripción Genética , Secuencias de Aminoácidos , Animales , Sitios de Unión , Línea Celular , Cromatina/metabolismo , Humanos , Ratones , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Proteína de Retinoblastoma/química , Proteína de Retinoblastoma/metabolismo
3.
Yale J Biol Med ; 92(4): 771-779, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31866794

RESUMEN

The cell death response to DNA damage is discussed in this Perspectives piece with cancer as the backdrop because DNA damaging agents (DDA) are widely used to treat cancer. From decades of clinical results, we learn that DDA have cured some cancers but their toxicity is temporary in most cancers due to emergence of DDA-resistant cancer cells. Investigation of DDA-activated genes, proteins, and pathways, known collectively as the DNA damage response (DDR), has uncovered the inner workings of DDR that protect the genome to sustain life. Paradoxically, however, DDR can also activate death. Current knowledge on DDA-activated death and hypotheses for how DDR may determine when and where to execute death are discussed. Given that cancer cells suffer from DDR defects, which account for their initial sensitivity to DDA, future therapeutic development may exploit those cancer-specific DDR defects to selectively create death-inducing DNA lesions, without using DDA, to kill DDA-resistant cancers.


Asunto(s)
Apoptosis , Daño del ADN , Animales , Puntos de Control del Ciclo Celular , Reparación del ADN , Humanos , Necroptosis , Neoplasias/genética , Neoplasias/patología
4.
J Immunol ; 191(2): 837-47, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23772033

RESUMEN

The p53 protein has not only important tumor suppressor activity but also additional immunological and other functions, whose nature and extent are just beginning to be recognized. In this article, we show that p53 has a novel inflammation-promoting action in the intestinal tract, because loss of p53 or the upstream activating kinase, ATM, protects against acute intestinal inflammation in murine models. Mechanistically, deficiency in p53 leads to increased survival of epithelial cells and lamina propria macrophages, higher IL-6 expression owing to enhanced glucose-dependent NF-κB activation, and increased mucosal STAT3 activation. Blockade or loss of IL-6 signaling reverses the protective effects of p53 deficiency. Conversely, IL-6 treatment protects against acute colitis in a manner dependent on STAT3 signaling and induction of cytoprotective factors in epithelial cells. Together, these results indicate that p53 promotes inflammation in the intestinal tract through suppression of epithelium-protective factors, thus significantly expanding the spectrum of physiological and immunological p53 activities unrelated to cancer formation.


Asunto(s)
Colitis/inmunología , Colitis/prevención & control , Inflamación/inmunología , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada , Células de la Médula Ósea/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Colitis/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Dendríticas/metabolismo , Activación Enzimática , Células Epiteliales/metabolismo , Inflamación/prevención & control , Interleucina-6/biosíntesis , Interleucina-6/farmacología , Interleucinas/biosíntesis , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , FN-kappa B/metabolismo , Estrés Oxidativo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Interleucina-22
5.
Am J Physiol Cell Physiol ; 307(2): C180-9, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24848114

RESUMEN

Enteropathogenic Escherichia coli (EPEC) and Citrobacter rodentium are attaching-and-effacing (A/E) pathogens that cause intestinal inflammation and diarrhea. The bacteria adhere to the intestinal epithelium, destroy microvilli, and induce actin-filled membranous pedestals but do not invade the mucosa. Adherence leads to activation of several host cell kinases, including FYN, n-SRC, YES, ABL, and ARG, phosphorylation of the bacterial translocated intimin receptor, and actin polymerization and pedestal formation in cultured cells. However, marked functional redundancy appears to exist between kinases, and their physiological importance in A/E pathogen infections has remained unclear. To address this question, we employed a novel dynamic in vitro infection model that mimics transient and short-term interactions in the intestinal tract. Screening of a kinase inhibitor library and RNA interference experiments in vitro revealed that ABL and platelet-derived growth factor (PDGF) receptor (PDGFR) kinases, as well as p38 MAP kinase, have unique, indispensable roles in early attachment of EPEC to epithelial cells under dynamic infection conditions. Studies with mutant EPEC showed that the attachment functions of ABL and PDGFR were independent of the intimin receptor but required bacterial bundle-forming pili. Furthermore, inhibition of ABL and PDGFR with imatinib protected against infection of mice with modest loads of C. rodentium, whereas the kinases were dispensable for high inocula or late after infection. These results indicate that ABL and PDGFR have indispensable roles in early A/E pathogen attachment to intestinal epithelial cells and for in vivo infection with limiting inocula but are not required for late intimate bacterial attachment or high inoculum infections.


Asunto(s)
Adhesión Bacteriana/fisiología , Escherichia coli Enteropatógena/metabolismo , Células Epiteliales/fisiología , Proteínas Oncogénicas v-abl/metabolismo , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Animales , Línea Celular , Escherichia coli Enteropatógena/citología , Escherichia coli Enteropatógena/fisiología , Infecciones por Escherichia coli/microbiología , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas v-abl/genética , Inhibidores de Proteínas Quinasas/farmacología , Receptores del Factor de Crecimiento Derivado de Plaquetas/genética
6.
Cancer Cell ; 9(6): 417-8, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16766259

RESUMEN

The DNA mismatch repair (MMR) system maintains genome integrity by correcting replication errors. MMR also stimulates checkpoint and cell death responses to DNA damage suggested by the resistance of MMR-defective tumor cells to several chemotherapeutic agents. MMR-dependent cytotoxic response may result from futile repair; however, MMR-mediated apoptosis has been genetically separated from its repair function. In a recent issue of Molecular Cell, Yoshioka and coworkers show that MMR complexes (MutSalpha and MutLalpha) are required for the recruitment of ATR-ATRIP to sites of alkylation damage, demonstrating that MMR complexes can function as sensors in DNA damage signal transduction.


Asunto(s)
Apoptosis , Daño del ADN , Reparación del ADN , Exodesoxirribonucleasas/metabolismo , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/fisiología , Fosfoproteínas/metabolismo , Transducción de Señal , Proteínas Adaptadoras Transductoras de Señales , Alquilación , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Disparidad de Par Base , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN , Humanos , Proteína MutS de Unión a los Apareamientos Incorrectos del ADN/genética , Proteínas Serina-Treonina Quinasas/metabolismo
7.
EMBO Rep ; 12(2): 164-71, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21212806

RESUMEN

Despite having distinct expression patterns and phenotypes in mutant mice, the myogenic regulatory factors Myf5 and MyoD have been considered to be functionally equivalent. Here, we report that these factors have a different response to DNA damage, due to the presence in MyoD and absence in Myf5 of a consensus site for Abl-mediated tyrosine phosphorylation that inhibits MyoD activity in response to DNA damage. Genotoxins failed to repress skeletal myogenesis in MyoD-null embryos; reintroduction of wild-type MyoD, but not mutant Abl phosphorylation-resistant MyoD, restored the DNA-damage-dependent inhibition of muscle differentiation. Conversely, introduction of the Abl-responsive phosphorylation motif converts Myf5 into a DNA-damage-sensitive transcription factor. Gene-dosage-dependent reduction of Abl kinase activity in MyoD-expressing cells attenuated the DNA-damage-dependent inhibition of myogenesis. The presence of a DNA-damage-responsive phosphorylation motif in vertebrate, but not in invertebrate MyoD suggests an evolved response to environmental stress, originated from basic helix-loop-helix gene duplication in vertebrate myogenesis.


Asunto(s)
Desarrollo de Músculos/efectos de los fármacos , Mutágenos/toxicidad , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Evolución Biológica , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Reactivos de Enlaces Cruzados/toxicidad , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Etopósido/toxicidad , Femenino , Técnicas de Silenciamiento del Gen , Metilmetanosulfonato/toxicidad , Ratones/embriología , Mitomicina/toxicidad , Proteína MioD/genética , Factor 5 Regulador Miogénico/genética , Fosforilación , Embarazo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-abl/fisiología , Interferencia de ARN , Somitos/efectos de los fármacos , Somitos/metabolismo , Proteínas Supresoras de Tumor/metabolismo
8.
Nat Rev Cancer ; 3(2): 130-8, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12563312

RESUMEN

Recent studies have shown that RB can inhibit apoptosis, independently of its ability to block cell proliferation. This poses the question of how cells choose to grow or to die when RB becomes inactivated. RB is phosphorylated following mitogenic stimulation, but it is degraded in response to death stimuli. Most sporadic cancers also inactivate RB by phosphorylation, rather than losing RB entirely--possibly to exploit the survival advantage conferred by RB under stress. Drawing from the different mechanisms of RB inactivation, we propose two models for ways in which cells use RB to make the choice of life versus death.


Asunto(s)
Apoptosis/fisiología , Proteína de Retinoblastoma/fisiología , Animales , Ciclo Celular , Supervivencia Celular , Humanos , Ratones , Ratones Noqueados , Neoplasias/mortalidad , Neoplasias/fisiopatología , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética
9.
Nat Genet ; 32(4): 585-93, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12415271

RESUMEN

Cell-cycle checkpoints help to protect the genomes of proliferating cells under genotoxic stress. In multicellular organisms, cell proliferation is often directed toward differentiation during development and throughout adult homeostasis. To prevent the formation of differentiated cells with genetic instability, we hypothesized that genotoxic stress may trigger a differentiation checkpoint. Here we show that exposure to genotoxic agents causes a reversible inhibition of myogenic differentiation. Muscle-specific gene expression is suppressed by DNA-damaging agents if applied prior to differentiation induction but not after the differentiation program is established. The myogenic determination factor, MyoD (encoded by Myod1), is a target of the differentiation checkpoint in myoblasts. The inhibition of MyoD by DNA damage requires a functional c-Abl tyrosine kinase (encoded by Abl1), but occurs in cells deficient for p53 (transformation-related protein 53, encoded by Trp53) or c-Jun (encoded by the oncogene Jun). These results support the idea that genotoxic stress can regulate differentiation, and identify a new biological function for DNA damage-activated signaling network.


Asunto(s)
Daño del ADN , Mioblastos/metabolismo , Células 3T3 , Animales , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Cisplatino/farmacología , Reparación del ADN , Etopósido/farmacología , Metilmetanosulfonato/farmacología , Ratones , Fibras Musculares Esqueléticas/citología , Fibras Musculares Esqueléticas/efectos de los fármacos , Mutágenos/farmacología , Proteína MioD/efectos de los fármacos , Proteína MioD/genética , Proteína MioD/metabolismo , Mioblastos/citología , Mioblastos/efectos de los fármacos , Miogenina/efectos de los fármacos , Miogenina/metabolismo , Cadenas Pesadas de Miosina/efectos de los fármacos , Cadenas Pesadas de Miosina/metabolismo , Fosforilación , Mutación Puntual , Proteínas Proto-Oncogénicas c-abl/metabolismo , Proteínas Proto-Oncogénicas c-abl/fisiología , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-jun/fisiología , Radiación Ionizante , Activación Transcripcional/efectos de los fármacos , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/fisiología , Tirosina/metabolismo
10.
Proc Natl Acad Sci U S A ; 106(37): 15762-7, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19717431

RESUMEN

The histone H3 variant CENP-A is required for epigenetic specification of centromere identity through a loading mechanism independent of DNA sequence. Using multiphoton absorption and DNA cleavage at unique sites by I-SceI endonuclease, we demonstrate that CENP-A is rapidly recruited to double-strand breaks in DNA, along with three components (CENP-N, CENP-T, and CENP-U) associated with CENP-A at centromeres. The centromere-targeting domain of CENP-A is both necessary and sufficient for recruitment to double-strand breaks. CENP-A accumulation at DNA breaks is enhanced by active non-homologous end-joining but does not require DNA-PKcs or Ligase IV, and is independent of H2AX. Thus, induction of a double-strand break is sufficient to recruit CENP-A in human and mouse cells. Finally, since cell survival after radiation-induced DNA damage correlates with CENP-A expression level, we propose that CENP-A may have a function in DNA repair.


Asunto(s)
Autoantígenos/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Roturas del ADN de Doble Cadena , Animales , Autoantígenos/química , Autoantígenos/genética , Transporte Biológico Activo , Línea Celular , Centrómero/metabolismo , Proteína A Centromérica , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Daño del ADN/fisiología , Reparación del ADN/fisiología , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Histonas/metabolismo , Humanos , Cinética , Ratones , Modelos Biológicos , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
11.
Nat Cell Biol ; 6(1): 3-7, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14704671

RESUMEN

Auto-inhibition describes the capacity of proteins to adopt a self-imposed latent conformation. Recently, a crystal structure of the Abl tyrosine kinase has revealed its ability to auto-inhibit. However, a separate body of work suggests that other cellular proteins also inhibit Abl. To reconcile the crystal structure with Abl inhibitors, I propose that Abl is controlled by cellular 'co-inhibitors' that bind Abl, stabilizing the auto-inhibited conformation. The implication of co-inhibition on Abl function is discussed.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Proteínas Proto-Oncogénicas c-abl/química , Proteínas Proto-Oncogénicas c-abl/fisiología , Animales , Ratones , Estructura Molecular , Unión Proteica/fisiología , Conformación Proteica
12.
Nat Cell Biol ; 4(10): 757-65, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12360286

RESUMEN

The retinoblastoma tumour suppressor protein RB is cleaved by caspases during apoptosis. Here we have mutated the caspase cleavage site in the carboxy terminus of the murine Rb protein in the mouse germ line to create the Rb-MI allele. After endotoxic shock, expression of Rb-MI inhibits apoptosis in the intestines, but not in the spleen, and promotes the survival of male mice. Fibroblasts expressing Rb-MI protein are protected from apoptosis induced by the tumour-necrosis factor-alpha type I receptor (TNFRI) but remain sensitive to cell death induced by DNA damage. Correspondingly, the release of cytochrome c and the activation of caspase-3 induced by TNFRI, but not by DNA damage, are defective in cells expressing Rb-MI. Our results highlight the importance of Rb cleavage in TNFRI-induced apoptosis.


Asunto(s)
Antígenos CD/metabolismo , Apoptosis/genética , Caspasas/metabolismo , Células Eucariotas/metabolismo , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Células 3T3 , Alelos , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Caspasa 3 , Grupo Citocromo c/efectos de los fármacos , Grupo Citocromo c/metabolismo , Células Eucariotas/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Lipopolisacáridos , Ratones , Ratones Noqueados , Mutación/genética , Receptores del Factor de Necrosis Tumoral/antagonistas & inhibidores , Receptores Tipo I de Factores de Necrosis Tumoral , Receptores Tipo II del Factor de Necrosis Tumoral , Retina/crecimiento & desarrollo , Retina/metabolismo , Retina/efectos de la radiación , Choque Séptico/inducido químicamente , Choque Séptico/genética , Choque Séptico/metabolismo , Transducción de Señal/genética , Toxinas Biológicas , Factor de Necrosis Tumoral alfa/metabolismo , Factor de Necrosis Tumoral alfa/farmacología
13.
Proc Natl Acad Sci U S A ; 105(37): 13993-8, 2008 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-18768816

RESUMEN

Mismatch repair (MMR) corrects replication errors during DNA synthesis. The mammalian MMR proteins also activate cell cycle checkpoints and apoptosis in response to persistent DNA damage. MMR-deficient cells are resistant to cisplatin, a DNA cross-linking agent used in chemotherapy, because of impaired activation of apoptotic pathways. It is shown that postmeiotic segregation 2 (PMS2), an MMR protein, is required for cisplatin-induced activation of p73, a member of the p53 family of transcription factors with proapoptotic activity. The human PMS2 is highly polymorphic, with at least 12 known nonsynonymous codon changes identified. We show here that the PMS2(R20Q) variant is defective in activating p73-dependent apoptotic response to cisplatin. When expressed in Pms2-deficient mouse fibroblasts, human PMS2(R20Q) but not PMS2 interfered with the apoptotic response to cisplatin. Correspondingly, PMS2 but not PMS2(R20Q) enhanced the cytotoxic effect of cisplatin measured by clonogenic survival. Because PMS2(R20Q) lacks proapoptotic activity, this polymorphic allele may modulate tumor responses to cisplatin among cancer patients.


Asunto(s)
Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Apoptosis , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Polimorfismo de Nucleótido Simple/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adenosina Trifosfatasas/deficiencia , Animales , Apoptosis/efectos de los fármacos , Arginina/genética , Arginina/metabolismo , Línea Celular , Chlorocebus aethiops , Cisplatino/farmacología , Daño del ADN/genética , Enzimas Reparadoras del ADN/deficiencia , Proteínas de Unión al ADN/deficiencia , Regulación de la Expresión Génica , Glutamina/genética , Glutamina/metabolismo , Humanos , Ratones , Ratones Noqueados , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto , Homólogo 1 de la Proteína MutL , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
14.
Proc Natl Acad Sci U S A ; 105(46): 17967-72, 2008 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-19004799

RESUMEN

During blast crisis of chronic myelogenous leukemia (CML), abnormal granulocyte macrophage progenitors (GMP) with nuclear beta-catenin acquire self-renewal potential and may function as leukemic stem cells (Jamieson et al. N Engl J Med, 2004). To develop a mouse model for CML-initiating GMP, we expressed p210(BCR-ABL) in an established line of E2A-knockout mouse BM cells that retain pluripotency in ex vivo culture. Expression of BCR-ABL in these cells reproducibly stimulated myeloid expansion in culture and generated leukemia-initiating cells specifically in the GMP compartment. The leukemogenic GMP displayed higher levels of beta-catenin activity than either the nontransformed GMP or the transformed nonGMP, both in culture and in transplanted mouse BM. Although E2A-deficiency may have contributed to the formation of leukemogenic GMP, restoration of E2A-function did not reverse BCR-ABL-induced transformation. These results provide further evidence that BCR-ABL-transformed GMP with abnormal beta-catenin activity can function as leukemic stem cells.


Asunto(s)
Transformación Celular Neoplásica/patología , Proteínas de Fusión bcr-abl/metabolismo , Células Progenitoras de Granulocitos y Macrófagos/patología , Células Progenitoras Mieloides/patología , Células Madre Neoplásicas/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/deficiencia , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , beta Catenina/metabolismo
15.
Proc Natl Acad Sci U S A ; 104(52): 20708-12, 2007 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-18093921

RESUMEN

An effective, noninvasive means of selecting cells based on their phase within the cell cycle is an important capability for biological research. Current methods of producing synchronous cell populations, however, tend to disrupt the natural physiology of the cell or suffer from low synchronization yields. In this work, we report a microfluidic device that utilizes the dielectrophoresis phenomenon to synchronize cells by exploiting the relationship between the cell's volume and its phase in the cell cycle. The dielectrophoresis activated cell synchronizer (DACSync) device accepts an asynchronous mixture of cells at the inlet, fractionates the cell populations according to the cell-cycle phase (G(1)/S and G(2)/M), and elutes them through different outlets. The device is gentle and efficient; it utilizes electric fields that are 1-2 orders of magnitude below those used in electroporation and enriches asynchronous tumor cells in the G(1) phase to 96% in one round of sorting, in a continuous flow manner at a throughput of 2 x 10(5) cells per hour per microchannel. This work illustrates the feasibility of using laminar flow and electrokinetic forces for the efficient, noninvasive separation of living cells.


Asunto(s)
Ciclo Celular , Electroforesis/instrumentación , Electroforesis/métodos , División Celular , Línea Celular Tumoral , Separación Celular , Técnicas Citológicas , Electroquímica/métodos , Diseño de Equipo , Citometría de Flujo , Humanos , Cinética , Técnicas Analíticas Microfluídicas , Modelos Teóricos
16.
Mol Biol Cell ; 18(10): 4143-54, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17686996

RESUMEN

The nonreceptor Abl tyrosine kinase stimulates F-actin microspikes and membrane ruffles in response to adhesion and growth factor signals. We show here that induced dimerization of Abl-FKBP, but not the kinase-defective AblKD-FKBP, inhibits cell spreading on fibronectin. Conversely, knockdown of cellular Abl by shRNA stimulates cell spreading. The Abl kinase inhibitor, imatinib, also stimulates cell spreading and its effect is overridden by the imatinib-resistant AblT315I. Expression of Abl but not AbkKD in Abl/Arg-deficient cells again inhibits spreading. Furthermore, Abl inhibits spreading of cells that express the activated Rac, RacV12, correlating with RacV12 localization to dorsal membrane protrusions. Ectopic expression of CrkII, a Rac activator that is inactivated by Abl-mediated tyrosine phosphorylation, antagonizes Abl-mediated dorsal membrane localization of RacV12. Ectopic expression of a dynamin-2 mutant, previously shown to induce Rac-GTP localization to the dorsal membrane, abolishes the stimulatory effect of imatinib on cell spreading. These results suggest that Abl tyrosine kinase, through CrkII phosphorylation and in collaboration with dynamin-2 can regulate the partitioning of Rac-GTP to favor dorsal ruffles during cell spreading. The Abl-dependent dorsal membrane localization of activated Rac explains its positive role in ruffling and negative role in cell spreading and migration.


Asunto(s)
Fibronectinas/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Seudópodos/enzimología , Animales , Benzamidas , Movimiento Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Forma de la Célula , Dimerización , Dinaminas/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Guanosina Trifosfato/metabolismo , Mesilato de Imatinib , Ratones , Proteínas Mutantes/metabolismo , Células 3T3 NIH , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Proteínas Tirosina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-abl/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-crk/metabolismo , Seudópodos/efectos de los fármacos , Seudópodos/metabolismo , Pirimidinas/farmacología , Proteínas Recombinantes de Fusión/metabolismo , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/metabolismo , Proteínas de Unión al GTP rac/metabolismo
17.
BMC Biol ; 7: 35, 2009 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-19558638

RESUMEN

BACKGROUND: The p53 tumor suppressor and its related protein, p73, share a homologous DNA binding domain, and mouse genetics studies have suggested that they have overlapping as well as distinct biological functions. Both p53 and p73 are activated by genotoxic stress to regulate an array of cellular responses. Previous studies have suggested that p53 and p73 independently activate the cellular apoptotic program in response to cytotoxic drugs. The goal of this study was to compare the promoter-binding activity of p53 and p73 at steady state and after genotoxic stress induced by hydroxyurea. RESULTS: We employed chromatin immunoprecipitation, the NimbleGen promoter arrays and a model-based algorithm for promoter arrays to identify promoter sequences enriched in anti-p53 or anti-p73 immunoprecipitates, either before or after treatment with hydroxyurea, which increased the expression of both p53 and p73 in the human colon cancer cell line HCT116-3(6). We calculated a model-based algorithm for promoter array score for each promoter and found a significant correlation between the promoter occupancy profiles of p53 and p73. We also found that after hydroxyurea treatment, the p53-bound promoters were still bound by p73, but p73 became associated with additional promoters that that did not bind p53. In particular, we showed that hydroxyurea induces the binding of p73 but not p53 to the promoter of MLH3, which encodes a mismatch repair protein, and causes an up-regulation of the MLH3 mRNA. CONCLUSION: These results suggest that hydroxyurea exerts differential effects on the promoter-binding functions of p53 and p73 and illustrate the power of model-based algorithm for promoter array in the analyses of promoter occupancy profiles of highly homologous transcription factors.


Asunto(s)
Daño del ADN , Proteínas de Unión al ADN/metabolismo , Hidroxiurea/toxicidad , Proteínas Nucleares/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/toxicidad , Regiones Promotoras Genéticas , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Algoritmos , Proteínas Portadoras/genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Técnicas de Transferencia de Gen , Células HCT116 , Humanos , Hidroxiurea/metabolismo , Proteínas de la Membrana/metabolismo , Modelos Biológicos , Proteínas MutL , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Inhibidores de la Síntesis del Ácido Nucleico/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Terminación de Péptidos/genética , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño , Proteína Tumoral p73 , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
18.
Biol Reprod ; 81(4): 749-58, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19553600

RESUMEN

Leiomyomata uteri (i.e., uterine fibroids) are benign tumors arising from the abnormal growth of uterine smooth muscle cells (SMCs). We show here that the expression of platelet-derived growth factor C (PDGFC) is higher in approximately 80% of uterine fibroids than in adjacent myometrial tissues examined. Increased expression of PDGFC is also observed in fibroid-derived SMCs (fSMCs) relative to myometrial-derived SMCs (mSMCs). Recombinant bioactive PDGFCC homodimer stimulates the growth of fSMCs and mSMCs in ex vivo cultures and prolongs the survival of fSMCs in Matrigel plugs implemented subcutaneously in immunocompromised mice. The knockdown of PDGF receptor-alpha (PDGFRA) through lentiviral-mediated RNA interference reduces the growth of fSMCs and mSMCs in ex vivo cultures and in Matrigel implants. Furthermore, two small molecule inhibitors of the PDGFR tyrosine kinase (i.e., imatinib and dasatinib) exerted negative effects on fSMC and mSMC growth in ex vivo cultures, albeit at concentrations that cannot be achieved in vivo. These results suggest that the PDGFCC/PDGFRA signaling module plays an important role in fSMC and mSMC growth, and that the upregulation of PDGFC expression may contribute to the clonal expansion of fSMCs in the development of uterine fibroids.


Asunto(s)
Leiomioma/metabolismo , Linfocinas/metabolismo , Miometrio/metabolismo , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Neoplasias Uterinas/metabolismo , Adulto , Animales , Proliferación Celular , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Noqueados , Persona de Mediana Edad , Miocitos del Músculo Liso/fisiología , Inhibidores de Proteínas Quinasas , ARN/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes , Regulación hacia Arriba
19.
J Cell Biol ; 156(5): 879-92, 2002 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-11864995

RESUMEN

The nonreceptor tyrosine kinase encoded by the c-Abl gene has the unique feature of an F-actin binding domain (FABD). Purified c-Abl tyrosine kinase is inhibited by F-actin, and this inhibition can be relieved through mutation of its FABD. The c-Abl kinase is activated by physiological signals that also regulate the actin cytoskeleton. We show here that c-Abl stimulated the formation of actin microspikes in fibroblasts spreading on fibronectin. This function of c-Abl is dependent on kinase activity and is not shared by c-Src tyrosine kinase. The Abl-dependent F-actin microspikes occurred under conditions where the Rho-family GTPases were inhibited. The FABD-mutated c-Abl, which is active in detached fibroblasts, stimulated F-actin microspikes independent of cell attachment. Moreover, FABD-mutated c-Abl stimulated the formation of F-actin branches in neurites of rat embryonic cortical neurons. The reciprocal regulation between F-actin and the c-Abl tyrosine kinase may provide a self-limiting mechanism in the control of actin cytoskeleton dynamics.


Asunto(s)
Actinas/metabolismo , Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Corteza Cerebral/embriología , Citoesqueleto/enzimología , Neuritas/metabolismo , Proteínas Proto-Oncogénicas c-abl/deficiencia , Animales , Benzamidas , Adhesión Celular/fisiología , Tamaño de la Célula/fisiología , Corteza Cerebral/citología , Corteza Cerebral/enzimología , Citoesqueleto/ultraestructura , Matriz Extracelular/metabolismo , Fibronectinas/farmacología , Técnica del Anticuerpo Fluorescente , Mesilato de Imatinib , Ratones , Neuritas/enzimología , Neuritas/ultraestructura , Piperazinas , Proteínas Proto-Oncogénicas c-abl/genética , Pirimidinas/farmacología , Ratas , Proteínas de Unión al GTP rho/genética , Proteínas de Unión al GTP rho/metabolismo
20.
J Cell Biol ; 165(4): 493-503, 2004 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-15148308

RESUMEN

Filopodia are dynamic F-actin structures that cells use to explore their environment. c-Abl tyrosine kinase promotes filopodia during cell spreading through an unknown mechanism that does not require Cdc42 activity. Using an unbiased approach, we identified Dok1 as a specific c-Abl substrate in spreading fibroblasts. When activated by cell adhesion, c-Abl phosphorylates Y361 of Dok1, promoting its association with the Src homology 2 domain (SH2)/SH3 adaptor protein Nck. Each signaling component was critical for filopodia formation during cell spreading, as evidenced by the finding that mouse fibroblasts lacking c-Abl, Dok1, or Nck had fewer filopodia than cells reexpressing the product of the disrupted gene. Dok1 and c-Abl stimulated filopodia in a mutually interdependent manner, indicating that they function in the same signaling pathway. Dok1 and c-Abl were both detected in filopodia of spreading cells, and therefore may act locally to modulate actin. Our data suggest a novel pathway by which c-Abl transduces signals to the actin cytoskeleton through phosphorylating Dok1 Y361 and recruiting Nck.


Asunto(s)
Movimiento Celular/fisiología , Proteínas de Unión al ADN/fisiología , Fosfoproteínas/fisiología , Proteínas Proto-Oncogénicas c-abl/fisiología , Seudópodos/fisiología , Proteínas de Unión al ARN/fisiología , Actinas/biosíntesis , Proteínas Adaptadoras Transductoras de Señales , Animales , Adhesión Celular/genética , Línea Celular Transformada , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , Proteínas Oncogénicas/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación , Estructura Terciaria de Proteína/genética , Proteínas Proto-Oncogénicas c-abl/genética , Proteínas Proto-Oncogénicas c-abl/metabolismo , Seudópodos/enzimología , Seudópodos/ultraestructura , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/genética , Dominios Homologos src/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA