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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 117(32): 19415-19424, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32719125

RESUMO

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.


Assuntos
Antineoplásicos/farmacologia , Endonucleases Flap/antagonistas & inibidores , Recombinação Homóloga/efeitos dos fármacos , Neoplasias/genética , Animais , Proteína BRCA1/deficiência , Proteína BRCA1/genética , Proteína BRCA2/deficiência , Proteína BRCA2/genética , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Endonucleases Flap/genética , Instabilidade Genômica/genética , Humanos , Camundongos , Neoplasias/tratamento farmacológico , RNA Interferente Pequeno/farmacologia , Saccharomyces cerevisiae , Proteínas de Saccharomyces cerevisiae/genética , Bibliotecas de Moléculas Pequenas/farmacologia , Mutações Sintéticas Letais , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Mol Cell ; 43(4): 663-72, 2011 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-21855804

RESUMO

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.


Assuntos
Dano ao DNA , Regulação da Expressão Gênica , Proteína do Retinoblastoma/fisiologia , Transcrição Gênica , Motivos de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Cromatina/metabolismo , Humanos , Camundongos , Domínios e Motivos de Interação entre Proteínas , Mapeamento de Interação de Proteínas , Proteína do Retinoblastoma/química , Proteína do Retinoblastoma/metabolismo
3.
Yale J Biol Med ; 92(4): 771-779, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31866794

RESUMO

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.


Assuntos
Apoptose , Dano ao DNA , Animais , Pontos de Checagem do Ciclo Celular , Reparo do DNA , Humanos , Necroptose , Neoplasias/genética , Neoplasias/patologia
4.
J Immunol ; 191(2): 837-47, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23772033

RESUMO

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.


Assuntos
Colite/imunologia , Colite/prevenção & controle , Inflamação/imunologia , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose , Proteínas Mutadas de Ataxia Telangiectasia , Células da Medula Óssea/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Colite/metabolismo , Reparo do DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células Dendríticas/metabolismo , Ativação Enzimática , Células Epiteliais/metabolismo , Inflamação/prevenção & controle , Interleucina-6/biossíntese , Interleucina-6/farmacologia , Interleucinas/biossíntese , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Estresse Oxidativo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Interleucina 22
5.
Am J Physiol Cell Physiol ; 307(2): C180-9, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24848114

RESUMO

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.


Assuntos
Aderência Bacteriana/fisiologia , Escherichia coli Enteropatogênica/metabolismo , Células Epiteliais/fisiologia , Proteínas Oncogênicas v-abl/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Linhagem Celular , Escherichia coli Enteropatogênica/citologia , Escherichia coli Enteropatogênica/fisiologia , Infecções por Escherichia coli/microbiologia , Regulação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Oncogênicas v-abl/genética , Inibidores de Proteínas Quinases/farmacologia , Receptores do Fator de Crescimento Derivado de Plaquetas/genética
6.
Cancer Cell ; 9(6): 417-8, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16766259

RESUMO

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.


Assuntos
Apoptose , Dano ao DNA , Reparo do DNA , Exodesoxirribonucleases/metabolismo , Proteína MutS de Ligação de DNA com Erro de Pareamento/fisiologia , Fosfoproteínas/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal , Alquilação , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Pareamento Incorreto de Bases , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA , Humanos , Proteína MutS de Ligação de DNA com Erro de Pareamento/genética , Proteínas Serina-Treonina Quinases/metabolismo
7.
EMBO Rep ; 12(2): 164-71, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21212806

RESUMO

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.


Assuntos
Desenvolvimento Muscular/efeitos dos fármacos , Mutagênicos/toxicidade , Proteína MyoD/metabolismo , Fator Regulador Miogênico 5/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia , Evolução Biológica , Proteínas de Ciclo Celular/metabolismo , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , Reagentes de Ligações Cruzadas/toxicidade , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Etoposídeo/toxicidade , Feminino , Técnicas de Silenciamento de Genes , Metanossulfonato de Metila/toxicidade , Camundongos/embriologia , Mitomicina/toxicidade , Proteína MyoD/genética , Fator Regulador Miogênico 5/genética , Fosforilação , Gravidez , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-abl/fisiologia , Interferência de RNA , Somitos/efeitos dos fármacos , Somitos/metabolismo , Proteínas Supressoras de Tumor/metabolismo
8.
Nat Rev Cancer ; 3(2): 130-8, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12563312

RESUMO

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.


Assuntos
Apoptose/fisiologia , Proteína do Retinoblastoma/fisiologia , Animais , Ciclo Celular , Sobrevivência Celular , Humanos , Camundongos , Camundongos Knockout , Neoplasias/mortalidade , Neoplasias/fisiopatologia , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/genética
9.
Nat Genet ; 32(4): 585-93, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12415271

RESUMO

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.


Assuntos
Dano ao DNA , Mioblastos/metabolismo , Células 3T3 , Animais , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células Cultivadas , Cisplatino/farmacologia , Reparo do DNA , Etoposídeo/farmacologia , Metanossulfonato de Metila/farmacologia , Camundongos , Fibras Musculares Esqueléticas/citologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Mutagênicos/farmacologia , Proteína MyoD/efeitos dos fármacos , Proteína MyoD/genética , Proteína MyoD/metabolismo , Mioblastos/citologia , Mioblastos/efeitos dos fármacos , Miogenina/efeitos dos fármacos , Miogenina/metabolismo , Cadeias Pesadas de Miosina/efeitos dos fármacos , Cadeias Pesadas de Miosina/metabolismo , Fosforilação , Mutação Puntual , Proteínas Proto-Oncogênicas c-abl/metabolismo , Proteínas Proto-Oncogênicas c-abl/fisiologia , Proteínas Proto-Oncogênicas c-jun/genética , Proteínas Proto-Oncogênicas c-jun/metabolismo , Proteínas Proto-Oncogênicas c-jun/fisiologia , Radiação Ionizante , Ativação Transcricional/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/fisiologia , Tirosina/metabolismo
10.
Proc Natl Acad Sci U S A ; 106(37): 15762-7, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19717431

RESUMO

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.


Assuntos
Autoantígenos/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Quebras de DNA de Cadeia Dupla , Animais , Autoantígenos/química , Autoantígenos/genética , Transporte Biológico Ativo , Linhagem Celular , Centrômero/metabolismo , Proteína Centromérica A , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/genética , Dano ao DNA/fisiologia , Reparo do DNA/fisiologia , Desoxirribonucleases de Sítio Específico do Tipo II/metabolismo , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Histonas/metabolismo , Humanos , Cinética , Camundongos , Modelos Biológicos , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
11.
Nat Cell Biol ; 6(1): 3-7, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14704671

RESUMO

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.


Assuntos
Retroalimentação Fisiológica/fisiologia , Proteínas Proto-Oncogênicas c-abl/química , Proteínas Proto-Oncogênicas c-abl/fisiologia , Animais , Camundongos , Estrutura Molecular , Ligação Proteica/fisiologia , Conformação Proteica
12.
Nat Cell Biol ; 4(10): 757-65, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12360286

RESUMO

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.


Assuntos
Antígenos CD/metabolismo , Apoptose/genética , Caspases/metabolismo , Células Eucarióticas/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/genética , Células 3T3 , Alelos , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Caspase 3 , Grupo dos Citocromos c/efeitos dos fármacos , Grupo dos Citocromos c/metabolismo , Células Eucarióticas/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Lipopolissacarídeos , Camundongos , Camundongos Knockout , Mutação/genética , Receptores do Fator de Necrose Tumoral/antagonistas & inibidores , Receptores Tipo I de Fatores de Necrose Tumoral , Receptores Tipo II do Fator de Necrose Tumoral , Retina/crescimento & desenvolvimento , Retina/metabolismo , Retina/efeitos da radiação , Choque Séptico/induzido quimicamente , Choque Séptico/genética , Choque Séptico/metabolismo , Transdução de Sinais/genética , Toxinas Biológicas , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
13.
Proc Natl Acad Sci U S A ; 105(37): 13993-8, 2008 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-18768816

RESUMO

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.


Assuntos
Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Apoptose , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenosina Trifosfatases/deficiência , Animais , Apoptose/efeitos dos fármacos , Arginina/genética , Arginina/metabolismo , Linhagem Celular , Chlorocebus aethiops , Cisplatino/farmacologia , Dano ao DNA/genética , Enzimas Reparadoras do DNA/deficiência , Proteínas de Ligação a DNA/deficiência , Regulação da Expressão Gênica , Glutamina/genética , Glutamina/metabolismo , Humanos , Camundongos , Camundongos Knockout , Endonuclease PMS2 de Reparo de Erro de Pareamento , Proteína 1 Homóloga a MutL , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
14.
Proc Natl Acad Sci U S A ; 105(46): 17967-72, 2008 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-19004799

RESUMO

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.


Assuntos
Transformação Celular Neoplásica/patologia , Proteínas de Fusão bcr-abl/metabolismo , Células Progenitoras de Granulócitos e Macrófagos/patologia , Células Progenitoras Mieloides/patologia , Células-Tronco Neoplásicas/patologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , beta Catenina/metabolismo
15.
Proc Natl Acad Sci U S A ; 104(52): 20708-12, 2007 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18093921

RESUMO

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.


Assuntos
Ciclo Celular , Eletroforese/instrumentação , Eletroforese/métodos , Divisão Celular , Linhagem Celular Tumoral , Separação Celular , Técnicas Citológicas , Eletroquímica/métodos , Desenho de Equipamento , Citometria de Fluxo , Humanos , Cinética , Técnicas Analíticas Microfluídicas , Modelos Teóricos
16.
Mol Biol Cell ; 18(10): 4143-54, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17686996

RESUMO

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.


Assuntos
Fibronectinas/metabolismo , Proteínas Proto-Oncogênicas c-abl/metabolismo , Pseudópodes/enzimologia , Animais , Benzamidas , Movimento Celular/efeitos dos fármacos , Polaridade Celular/efeitos dos fármacos , Forma Celular , Dimerização , Dinaminas/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Guanosina Trifosfato/metabolismo , Mesilato de Imatinib , Camundongos , Proteínas Mutantes/metabolismo , Células NIH 3T3 , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-crk/metabolismo , Pseudópodes/efeitos dos fármacos , Pseudópodes/metabolismo , Pirimidinas/farmacologia , Proteínas Recombinantes de Fusão/metabolismo , Proteínas de Ligação a Tacrolimo/química , Proteínas de Ligação a Tacrolimo/metabolismo , Proteínas rac de Ligação ao GTP/metabolismo
17.
BMC Biol ; 7: 35, 2009 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-19558638

RESUMO

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.


Assuntos
Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Hidroxiureia/toxicidade , Proteínas Nucleares/metabolismo , Inibidores da Síntese de Ácido Nucleico/toxicidade , Regiões Promotoras Genéticas , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Algoritmos , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Técnicas de Transferência de Genes , Células HCT116 , Humanos , Hidroxiureia/metabolismo , Proteínas de Membrana/metabolismo , Modelos Biológicos , Proteínas MutL , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Inibidores da Síntese de Ácido Nucleico/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fatores de Terminação de Peptídeos/genética , Ligação Proteica/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Proteína Tumoral p73 , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética
18.
Biol Reprod ; 81(4): 749-58, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19553600

RESUMO

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.


Assuntos
Leiomioma/metabolismo , Linfocinas/metabolismo , Miométrio/metabolismo , Fator de Crescimento Derivado de Plaquetas/metabolismo , Neoplasias Uterinas/metabolismo , Adulto , Animais , Proliferação de Células , Células Cultivadas , Feminino , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Miócitos de Músculo Liso/fisiologia , Inibidores de Proteínas Quinases , RNA/metabolismo , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Proteínas Recombinantes , Regulação para Cima
19.
J Cell Biol ; 156(5): 879-92, 2002 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-11864995

RESUMO

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.


Assuntos
Actinas/metabolismo , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Córtex Cerebral/embriologia , Citoesqueleto/enzimologia , Neuritos/metabolismo , Proteínas Proto-Oncogênicas c-abl/deficiência , Animais , Benzamidas , Adesão Celular/fisiologia , Tamanho Celular/fisiologia , Córtex Cerebral/citologia , Córtex Cerebral/enzimologia , Citoesqueleto/ultraestrutura , Matriz Extracelular/metabolismo , Fibronectinas/farmacologia , Imunofluorescência , Mesilato de Imatinib , Camundongos , Neuritos/enzimologia , Neuritos/ultraestrutura , Piperazinas , Proteínas Proto-Oncogênicas c-abl/genética , Pirimidinas/farmacologia , Ratos , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo
20.
J Cell Biol ; 165(4): 493-503, 2004 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-15148308

RESUMO

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.


Assuntos
Movimento Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Fosfoproteínas/fisiologia , Proteínas Proto-Oncogênicas c-abl/fisiologia , Pseudópodes/fisiologia , Proteínas de Ligação a RNA/fisiologia , Actinas/biossíntese , Proteínas Adaptadoras de Transdução de Sinal , Animais , Adesão Celular/genética , Linhagem Celular Transformada , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Camundongos , Proteínas Oncogênicas/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilação , Estrutura Terciária de Proteína/genética , Proteínas Proto-Oncogênicas c-abl/genética , Proteínas Proto-Oncogênicas c-abl/metabolismo , Pseudópodes/enzimologia , Pseudópodes/ultraestrutura , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais/genética , Domínios de Homologia de src/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA