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










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38766081

RESUMO

Precision nuclear run-on (PRO) sequencing (PRO-seq) is a powerful technique for mapping polymerase active sites with nucleotide resolution and measuring newly synthesized transcripts at both promoters and enhancer elements. The current PRO-seq protocol is time-intensive, technically challenging, and requires a large amount of starting material. To overcome these limitations, we developed rapid PRO-seq (rPRO-seq) which utilizes pre-adenylated single-stranded DNAs (AppDNA), a dimer blocking oligonucleotide (DBO), on-bead 5' RNA end repair, and column-based purification. These modifications enabled efficient transcriptome mapping within a single day (∼12 hours) increasing ligation efficiency, abolished adapter dimers, and reduced sample loss and RNA degradation. We demonstrate the reproducibility of rPRO-seq in measuring polymerases at promoters, gene bodies, and enhancers as compared to original PRO-seq protocols. Additionally, rPRO-seq is scalable, allowing for transcriptome mapping with as little as 25,000 cells. We apply rPRO-seq to study the role of Integrator in mouse hematopoietic stem and progenitor cell (mHSPC) homeostasis, identifying Ints11 as an essential component of transcriptional regulation and RNA processing in mHSPC homeostasis. Overall, rPRO-seq represents a significant advance in the field of nascent transcript analyses and will be a valuable tool for generating patient-specific genome-wide transcription profiles with minimal sample requirements.

2.
Sci Adv ; 7(45): eabe3393, 2021 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-34730992

RESUMO

Integrator regulates the 3'-end processing and termination of multiple classes of noncoding RNAs. Depletion of INTS11, the catalytic subunit of Integrator, or ectopic expression of its catalytic dead enzyme impairs the 3'-end processing and termination of a set of protein-coding transcripts termed Integrator-regulated termination (IRT) genes. This defect is manifested by increased RNA polymerase II (RNAPII) readthrough and occupancy of serine-2 phosphorylated RNAPII, de novo trimethylation of lysine-36 on histone H3, and a compensatory elevation of the cleavage and polyadenylation (CPA) complex beyond the canonical polyadenylation sites. 3' RNA sequencing reveals that proximal polyadenylation site usage relies on the endonuclease activity of INTS11. The DNA sequence encompassing the transcription end sites of IRT genes features downstream polyadenylation motifs and an enrichment of GC content that permits the formation of secondary structures within the 3'UTR. Together, this study identifies a subset of protein-coding transcripts whose 3' end processing requires the Integrator complex.

3.
Cell Rep ; 32(3): 107917, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32697989

RESUMO

Transcription by RNA polymerase II (RNAPII) is pervasive in the human genome. However, the mechanisms controlling transcription at promoters and enhancers remain enigmatic. Here, we demonstrate that Integrator subunit 11 (INTS11), the catalytic subunit of the Integrator complex, regulates transcription at these loci through its endonuclease activity. Promoters of genes require INTS11 to cleave nascent transcripts associated with paused RNAPII and induce their premature termination in the proximity of the +1 nucleosome. The turnover of RNAPII permits the subsequent recruitment of an elongation-competent RNAPII complex, leading to productive elongation. In contrast, enhancers require INTS11 catalysis not to evict paused RNAPII but rather to terminate enhancer RNA transcription beyond the +1 nucleosome. These findings are supported by the differential occupancy of negative elongation factor (NELF), SPT5, and tyrosine-1-phosphorylated RNAPII. This study elucidates the role of Integrator in mediating transcriptional elongation at human promoters through the endonucleolytic cleavage of nascent transcripts and the dynamic turnover of RNAPII.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Endonucleases/metabolismo , Elongação da Transcrição Genética , Biocatálise , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica , Células HeLa , Humanos , Nucleossomos/metabolismo , Fosforilação , Regiões Promotoras Genéticas , RNA/metabolismo , RNA Polimerase II/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Terminação da Transcrição Genética
4.
Genes Dev ; 34(9-10): 637-649, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32241802

RESUMO

The emergence of drug resistance is a major obstacle for the success of targeted therapy in melanoma. Additionally, conventional chemotherapy has not been effective as drug-resistant cells escape lethal DNA damage effects by inducing growth arrest commonly referred to as cellular dormancy. We present a therapeutic strategy termed "targeted chemotherapy" by depleting protein phosphatase 2A (PP2A) or its inhibition using a small molecule inhibitor (1,10-phenanthroline-5,6-dione [phendione]) in drug-resistant melanoma. Targeted chemotherapy induces the DNA damage response without causing DNA breaks or allowing cellular dormancy. Phendione treatment reduces tumor growth of BRAFV600E-driven melanoma patient-derived xenografts (PDX) and diminishes growth of NRASQ61R-driven melanoma, a cancer with no effective therapy. Remarkably, phendione treatment inhibits the acquisition of resistance to BRAF inhibition in BRAFV600E PDX highlighting its effectiveness in combating the advent of drug resistance.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Melanoma/tratamento farmacológico , Pirazóis/farmacologia , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dano ao DNA/efeitos dos fármacos , Humanos , Melanoma/enzimologia , Melanoma/fisiopatologia , Proteína Fosfatase 2/antagonistas & inibidores
5.
Int J Radiat Oncol Biol Phys ; 107(4): 779-792, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32259569

RESUMO

PURPOSE: Acquired hematopoietic failure is commonly caused by therapeutic and accidental exposure of the bone marrow (BM) to toxic agents. Efficient recovery from BM failure is dictated not only by the intrinsic sensitivity and proliferation capacity of the hematopoietic stem and progenitor cells but also by the BM environment niche. Identification of genetic factors that improve recovery from hematopoietic failure is essential. Vertebrate SETD4 is a poorly characterized and putatively nonhistone methyltransferase. This study aims to identify the roles of SETD4 in BM recovery. METHODS AND MATERIALS: An inducible SETD4 knockout mouse model (Setd4flox/flox;Rosa26-CreERT2+) was used. Adult sex-matched littermates were treated with tamoxifen to induce Setd4 deletion or oil as the control. Tamoxifen-treated Setd4wt/wt;Rosa26-CreERT2+ mice were included as another control. Those mice were irradiated to induce hematopoietic syndrome and analyzed to identify the roles and mechanisms of Setd4 in of BM recovery. RESULTS: Loss of Setd4 in adult mice improved the survival of whole-body irradiation-induced BM failure. This was associated with improved recoveries of long-term and short-term hematopoietic stem cells (HSCs) and early progenitor cells. BM transplantation analyses surprisingly showed that the improved recovery was not due to radiation resistance of the Setd4-deficient HSCs but that Setd4-deficient HSCs were actually more sensitive to radiation. However, the Setd4-deficient mice were better recipients for allogeneic HSC transplantation. Furthermore, there was enhanced splenic erythropoiesis in Setd4-deficient mice. CONCLUSION: These findings not only revealed a previously unrecognized role of Setd4 as a unique modulator of hematopoiesis but also underscored the critical role of the BM niche in recovery from hematopoietic failure. Our study also implicated Setd4 as a potential target for therapeutic inhibition to improve the conditioning of the BM niche before allogeneic transplantation.


Assuntos
Hematopoese/genética , Hematopoese/efeitos da radiação , Metiltransferases/deficiência , Metiltransferases/genética , Animais , Transplante de Medula Óssea , Técnicas de Inativação de Genes , Camundongos , Irradiação Corporal Total/efeitos adversos
6.
DNA Repair (Amst) ; 86: 102754, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31794893

RESUMO

Radiation-induced lymphomagenesis results from a clonogenic lymphoid cell proliferation due to genetic alterations and immunological dysregulation. Mouse models had been successfully used to identify risk and protective factors for radiation-induced DNA damage and carcinogenesis. The mammalian SETD4 is a poorly understood putative methyl-transferase. Here, we report that conditional Setd4 deletion in adult mice significantly extended the survival of radiation-induced T-lymphoma. However, in Tp53 deficient mice, Setd4 deletion did not delay the radiation-induced lymphomagenesis although it accelerated the spontaneous T-lymphomagenesis in non-irradiated mice. The T-lymphomas were largely clonogenic in both Setd4flox/flox and Setd4Δ/Δ mice based on sequencing analysis of the T-cell antigen ß receptors. However, the Setd4Δ/Δ T-lymphomas were CD4+/CD8+ double positive, while the littermate Setd4flox/floxtumor were largely CD8+ single positive. A genomic sequencing analysis on chromosome deletion, inversion, duplication, and translocation, revealed a larger contribution of inversion but a less contribution of deletion to the overall chromosome rearrangements in the in Setd4Δ/Δ tumors than the Setd4flox/flox tumors. In addition, the Setd4flox/flox mice died more often from the large sizes of primary thymus lymphoma at earlier time, but there was a slight increase of lymphoma dissemination among peripheral organs in Setd4Δ/Δ at later times. These results suggest that Setd4 has a critical role in modulating lymphomagenesis and may be targeted to suppress radiation-induced carcinogenesis.


Assuntos
Deleção de Genes , Linfoma/genética , Metiltransferases/genética , Neoplasias Induzidas por Radiação/genética , Neoplasias do Timo/genética , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Modelos Animais de Doenças , Linfoma/imunologia , Linfoma/mortalidade , Camundongos , Neoplasias Induzidas por Radiação/imunologia , Neoplasias Induzidas por Radiação/mortalidade , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Análise de Sequência de DNA , Neoplasias do Timo/imunologia , Neoplasias do Timo/mortalidade , Proteína Supressora de Tumor p53/genética
7.
Nat Commun ; 10(1): 4925, 2019 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-31664040

RESUMO

AML1-ETO (AE) is a fusion transcription factor, generated by the t(8;21) translocation, that functions as a leukemia promoting oncogene. Here, we demonstrate that TATA-Box Binding Protein Associated Factor 1 (TAF1) associates with K43 acetylated AE and this association plays a pivotal role in the proliferation of AE-expressing acute myeloid leukemia (AML) cells. ChIP-sequencing indicates significant overlap of the TAF1 and AE binding sites. Knockdown of TAF1 alters the association of AE with chromatin, affecting of the expression of genes that are activated or repressed by AE. Furthermore, TAF1 is required for leukemic cell self-renewal and its reduction promotes the differentiation and apoptosis of AE+ AML cells, thereby impairing AE driven leukemogenesis. Together, our findings reveal a role of TAF1 in leukemogenesis and identify TAF1 as a potential therapeutic target for AE-expressing leukemia.


Assuntos
Carcinogênese/patologia , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Histona Acetiltransferases/metabolismo , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Proteínas de Fusão Oncogênica/metabolismo , Proteína 1 Parceira de Translocação de RUNX1/metabolismo , Fatores Associados à Proteína de Ligação a TATA/metabolismo , Fator de Transcrição TFIID/metabolismo , Acetilação , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Autorrenovação Celular , Cromatina/metabolismo , Regulação Leucêmica da Expressão Gênica , Histona Acetiltransferases/química , Humanos , Lisina/metabolismo , Camundongos Endogâmicos C57BL , Células Mieloides/patologia , Ligação Proteica , Domínios Proteicos , Fatores Associados à Proteína de Ligação a TATA/química , Fator de Transcrição TFIID/química
8.
Genes Dev ; 31(17): 1809-1820, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28982763

RESUMO

Activating mutations in the mitogen-activated protein kinase (MAPK) cascade, also known as the RAS-MEK-extracellular signal-related kinase (ERK1/2) pathway, are an underlying cause of >70% of human cancers. While great strides have been made toward elucidating the cytoplasmic components of MAPK signaling, the key downstream coactivators that coordinate the transcriptional response have not been fully illustrated. Here, we demonstrate that the MAPK transcriptional response in human cells is funneled through Integrator, an RNA polymerase II-associated complex. Integrator depletion diminishes ERK1/2 transcriptional responsiveness and cellular growth in human cancers harboring activating mutations in MAPK signaling. Pharmacological inhibition of the MAPK pathway abrogates the stimulus-dependent recruitment of Integrator at immediate early genes and their enhancers. Following epidermal growth factor (EGF) stimulation, activated ERK1/2 is recruited to immediate early genes and phosphorylates INTS11, the catalytic subunit of Integrator. Importantly, in contrast to the broad effects of Integrator knockdown on MAPK responsiveness, depletion of a number of critical subunits of the coactivator complex Mediator alters only a few MAPK-responsive genes. Finally, human cancers with activating mutations in the MAPK cascade, rendered resistant to targeted therapies, display diminished growth following depletion of Integrator. We propose Integrator as a crucial transcriptional coactivator in MAPK signaling, which could serve as a downstream therapeutic target for cancer treatment.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Transdução de Sinais , Células A549 , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Proliferação de Células , Cromatina/metabolismo , Endorribonucleases , Ativação Enzimática , Técnicas de Silenciamento de Genes , Humanos , Indazóis/farmacologia , Sistema de Sinalização das MAP Quinases/genética , Neoplasias/enzimologia , Neoplasias/fisiopatologia , Fosforilação , Piperazinas/farmacologia , Regiões Promotoras Genéticas/genética , Transporte Proteico/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Proteínas ras/genética , Proteínas ras/metabolismo
9.
Cancer Cell ; 25(5): 697-710, 2014 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-24746704

RESUMO

MEK inhibitors are clinically active in BRAF(V600E) melanomas but only marginally so in KRAS mutant tumors. Here, we found that MEK inhibitors suppress ERK signaling more potently in BRAF(V600E), than in KRAS mutant tumors. To understand this, we performed an RNAi screen in a KRAS mutant model and found that CRAF knockdown enhanced MEK inhibition. MEK activated by CRAF was less susceptible to MEK inhibitors than when activated by BRAF(V600E). MEK inhibitors induced RAF-MEK complexes in KRAS mutant models, and disrupting such complexes enhanced inhibition of CRAF-dependent ERK signaling. Newer MEK inhibitors target MEK catalytic activity and also impair its reactivation by CRAF, either by disrupting RAF-MEK complexes or by interacting with Ser 222 to prevent MEK phosphorylation by RAF.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , MAP Quinase Quinase 1/antagonistas & inibidores , Melanoma/enzimologia , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas/genética , Fator 3 Associado a Receptor de TNF/genética , Proteínas ras/genética , Animais , Benzamidas/farmacologia , Linhagem Celular , Cumarínicos/farmacologia , Difenilamina/análogos & derivados , Difenilamina/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Células HEK293 , Humanos , Indóis/farmacologia , MAP Quinase Quinase 1/química , MAP Quinase Quinase 1/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Melanoma/tratamento farmacológico , Melanoma/genética , Camundongos , Camundongos Nus , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras) , Piridonas/farmacologia , Pirimidinonas/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Sulfonamidas/farmacologia , Ressonância de Plasmônio de Superfície , Fator 3 Associado a Receptor de TNF/metabolismo , Vemurafenib , Quinases raf/metabolismo
10.
PLoS One ; 8(1): e54389, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23372718

RESUMO

Dubowitz Syndrome is an autosomal recessive disorder with a unique set of clinical features including microcephaly and susceptibility to tumor formation. Although more than 140 cases of Dubowitz syndrome have been reported since 1965, the genetic defects of this disease has not been identified. In this study, we systematically analyzed the DNA damage response and repair capability of fibroblasts established from a Dubowitz Syndrome patient. Dubowitz syndrome fibroblasts are hypersensitive to ionizing radiation, bleomycin, and doxorubicin. However, they have relatively normal sensitivities to mitomycin-C, cisplatin, and camptothecin. Dubowitz syndrome fibroblasts also have normal DNA damage signaling and cell cycle checkpoint activations after DNA damage. These data implicate a defect in repair of DNA double strand break (DSB) likely due to defective non-homologous end joining (NHEJ). We further sequenced several genes involved in NHEJ, and identified a pair of novel compound mutations in the DNA Ligase IV gene. Furthermore, expression of wild type DNA ligase IV completely complement the DNA repair defects in Dubowitz syndrome fibroblasts, suggesting that the DNA ligase IV mutation is solely responsible for the DNA repair defects. These data suggests that at least subset of Dubowitz syndrome can be attributed to DNA ligase IV mutations.


Assuntos
Quebras de DNA de Cadeia Dupla , DNA Ligases/genética , Reparo do DNA , Eczema/genética , Transtornos do Crescimento/genética , Deficiência Intelectual/genética , Microcefalia/genética , Mutação , Adulto , Antineoplásicos/farmacologia , Dano ao DNA , DNA Ligase Dependente de ATP , Eczema/patologia , Eczema/radioterapia , Fácies , Evolução Fatal , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibroblastos/efeitos da radiação , Raios gama , Transtornos do Crescimento/patologia , Transtornos do Crescimento/radioterapia , Humanos , Deficiência Intelectual/patologia , Deficiência Intelectual/radioterapia , Microcefalia/patologia , Microcefalia/radioterapia , Tolerância a Radiação
11.
Cell Biosci ; 3(1): 7, 2013 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-23388158

RESUMO

Filamin-A (FLNA), also called actin-binding protein 280 (ABP-280), was originally identified as a non-muscle actin binding protein, which organizes filamentous actin into orthogonal networks and stress fibers. Filamin-A also anchors various transmembrane proteins to the actin cytoskeleton and provides a scaffold for a wide range of cytoplasmic and nuclear signaling proteins. Intriguingly, several studies have revealed that filamin-A associates with multiple non-cytoskeletal proteins of diverse function and is involved in several unrelated pathways. Mutations and aberrant expression of filamin-A have been reported in human genetic diseases and several types of cancer. In this review, we discuss the implications of filamin-A in cancer progression, including metastasis and DNA damage response.

12.
Int J Biol Sci ; 9(1): 67-77, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23289018

RESUMO

Filamin-A cross-links actin filaments into dynamic orthogonal networks, and interacts with an array of proteins of diverse cellular functions. Because several filamin-A interaction partners are implicated in signaling of cell mobility regulation, we tested the hypothesis that filamin-A plays a role in cancer metastasis. Using four pairs of filamin-A proficient and deficient isogenic cell lines, we found that filamin-A deficiency in cancer cells significantly reduces their migration and invasion. Using a xenograft tumor model with subcutaneous and intracardiac injections of tumor cells, we found that the filamin-A deficiency causes significant reduction of lung, splenic and systemic metastasis in nude mice. We evaluated the expression of filamin-A in breast cancer tissues by immunohistochemical staining, and found that low levels of filamin-A expression in cancer cells of the tumor tissues are associated with a better distant metastasis-free survival than those with normal levels of filamin-A. These data not only validate filamin-A as a prognostic marker for cancer metastasis, but also suggest that inhibition of filamin-A in cancer cells may reduce metastasis and that filamin-A can be used as a therapeutic target for filamin-A positive cancer.


Assuntos
Neoplasias da Mama/complicações , Neoplasias da Mama/metabolismo , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proteínas Contráteis/genética , Feminino , Filaminas , Regulação Neoplásica da Expressão Gênica , Imuno-Histoquímica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos , Proteínas dos Microfilamentos/genética , Metástase Neoplásica/genética
13.
Int J Biol Sci ; 8(4): 442-50, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22419889

RESUMO

Filamin-A, also called Actin Binding Protein-280, is not only an essential component of the cytoskeleton networks, but also serves as the scaffold in various signaling networks. It has been shown that filamin-A facilitates DNA repair and filamin-A proficient cells are more resistant to ionizing radiation, bleomycin, and cisplatin. In this study, we assessed the role of filamin-A in modulating cancer cell sensitivity to Topo II poisons, including etoposide and doxorubicin. Intriguingly, we found that cells with filamin-A expression are more sensitive to Topo II poisons than those with defective filamin-A, and filamin-A proficient xenograft melanomas have better response to etoposide treatment than the filamin-A deficient tumors. This is associated with more potent induction of DNA double strand breaks (DSBs) by Topo II poisons in filamin-A proficient cells than the deficient cells. Although the expression of filamin-A enables cells a slightly stronger capability to repair DSB, the net outcome is that filamin-A proficient cells bear more DSBs due to the significantly enhanced DSB induction by Topo II poisons in these cells. We further found that filamin-A proficient cells have increased drug influx and decreased drug efflux, suggesting that filamin-A modulates the intra-cellular drug kinetics of Topo II poisons to facilitate the generation of DSB after Topo II poison exposure. These data suggest a novel function of filamin-A in regulating the pharmacokinetics of Topo II poisons, and that the status of filamin-A may be used as a prognostic marker for Topo II poisons based cancer treatments.


Assuntos
Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Inibidores da Topoisomerase II/uso terapêutico , Animais , Bleomicina/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Cromatografia Líquida de Alta Pressão , Cisplatino/uso terapêutico , Proteínas Contráteis/genética , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Feminino , Filaminas , Citometria de Fluxo , Humanos , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Camundongos , Camundongos Nus , Proteínas dos Microfilamentos/genética , Microscopia de Fluorescência , Radiação Ionizante , Ensaios Antitumorais Modelo de Xenoenxerto
14.
DNA Repair (Amst) ; 11(2): 192-200, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22051193

RESUMO

Filamin-A, also called actin binding protein 280 (ABP-280), cross-links the actin filaments into dynamic orthogonal network to serve as scaffolds in multiple signaling pathways. It has been reported that filamin-A interacts with DNA damage response proteins BRCA1 and BRCA2. Defects of filamin-A impair the repair of DNA double strand breaks (DSBs), resulting in sensitization of cells to ionizing radiation. In this study, we sought to test the hypothesis that filamin-A can be used as a target for cancer chemotherapy and as a biomarker to predict cancer response to therapeutic DNA damage. We found that reduction of filamin-A sensitizes cancer cells to chemotherapy reagents bleomycin and cisplatin, delays the repair of not only DSBs but also single strand breaks (SSBs) and interstrand crosslinks (ICLs), and increases chromosome breaks after the drug treatment. By treating a panel of human melanoma cell lines with variable filamin-A expression, we observed a correlation between expression level of filamin-A protein and drug IC(50). We further inhibited the expression of filamin-A in melanoma cells, and found that this confers an increased sensitivity to bleomycin and cisplatin treatment in a mouse xenograft tumor model. These results suggest that filamin-A plays a role in repair of a variety of DNA damage, that lack of filamin-A is a prognostic marker for a better outcome after DNA damage based treatment, and filamin-A can be inhibited to sensitize filamin-A positive cancer cells to therapeutic DNA damage. Thus filamin-A can be used as a biomarker and a target for DNA damage based cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Biomarcadores Tumorais/metabolismo , Proteínas Contráteis/metabolismo , Dano ao DNA , Melanoma/patologia , Proteínas dos Microfilamentos/metabolismo , Terapia de Alvo Molecular/métodos , Animais , Antineoplásicos/uso terapêutico , Biomarcadores Tumorais/deficiência , Bleomicina/farmacologia , Bleomicina/uso terapêutico , Linhagem Celular Tumoral , Instabilidade Cromossômica/efeitos dos fármacos , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Proteínas Contráteis/deficiência , Quebras de DNA de Cadeia Simples/efeitos dos fármacos , Reparo do DNA/efeitos dos fármacos , Filaminas , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/metabolismo , Camundongos , Proteínas dos Microfilamentos/deficiência , Prognóstico , Ensaios Antitumorais Modelo de Xenoenxerto
15.
PLoS One ; 5(8): e12055, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20700465

RESUMO

BACKGROUND: Caveolin-1 (Cav-1), the major component of caveolae, is a 21-24 kDa integral membrane protein that interacts with a number of signaling molecules. By acting as a scaffolding protein, Cav-1 plays crucial roles in the regulation of various physiologic and patho-physiologic processes including oncogenic transformation and tumorigenesis, and tumor invasion and metastasis. METHODOLOGY/PRINCIPAL FINDINGS: In the present study we sought to explore the role of Cav-1 in response to DNA damage and the mechanism involved. We found that the level of Cav-1 was up-regulated rapidly in cells treated with ionizing radiation. The up-regulation of Cav-1 following DNA damage occurred only in cells expressing endogenous Cav-1, and was associated with the activation of DNA damage response pathways. Furthermore, we demonstrated that the expression of Cav-1 protected cells against DNA damage through modulating the activities of both the homologous recombination (HR) and non-homologous end joining (NHEJ) repair systems, as evidenced by the inhibitory effects of the Cav-1-targeted siRNA on cell survival, HR frequency, phosphorylation of DNA-dependent protein kinase (DNA-PK), and nuclear translocation of epidermal growth factor receptor (EGFR) following DNA damage, and by the stimulatory effect of the forced expression of Cav-1 on NHEJ frequency. CONCLUSION/SIGNIFICANCE: Our results indicate that Cav-1 may play a critical role in sensing genotoxic stress and in orchestrating the response of cells to DNA damage through regulating the important molecules involved in maintaining genomic integrity.


Assuntos
Caveolina 1/metabolismo , Dano ao DNA , Reparo do DNA , Recombinação Genética , Animais , Caveolina 1/deficiência , Caveolina 1/genética , Linhagem Celular Tumoral , DNA/genética , Quebras de DNA de Cadeia Dupla , Inativação Gênica , Humanos , Transdução de Sinais , Regulação para Cima
16.
Cancer Res ; 69(20): 7978-85, 2009 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-19808958

RESUMO

The human actin-binding protein filamin-A (also known as ABP-280) cross-links actin into a dynamic three-dimensional structure. It interacts with >45 proteins of diverse functions, serving as the scaffold in various signaling networks. BRCA2 is a protein that regulates RAD51-dependent recombinational repair of DNA double strand breaks (DSB). Proximate to the COOH terminus of the BRCA2 protein, a conserved and DNA binding domain (BRCA2-DBD) interacts with filamin-A and BCCIP. In this study, we sought to test the hypothesis that filamin-A influences homologous recombinational repair of DSB and the maintenance of genomic stability. We used three pairs of cell lines with normal and reduced filamin-A expression, including breast cancer and melanoma cells. We found that lack or reduction of filamin-A sensitizes cells to ionizing radiation, slows the removal of DNA damage-induced gammaH2AX nuclear foci, reduces RAD51 nuclear focus formation and recruitment to chromatin in response to irradiation, and results in a 2-fold reduction of homologous recombinational repair of DSB. Furthermore, filamin-A-deficient cells have increased frequencies of micronucleus formation after irradiation. Our data illustrate the importance of the cytoskeleton structure in supporting the homologous recombinational DNA repair machinery and genome integrity, and further implicate a potential of filamin-A as a marker for prognosis in DNA damage-based cancer therapy.


Assuntos
Neoplasias da Mama/genética , Proteínas Contráteis/fisiologia , Reparo do DNA/genética , Melanoma/genética , Proteínas dos Microfilamentos/fisiologia , Recombinação Genética , Apoptose/efeitos da radiação , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Western Blotting , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Núcleo Celular/efeitos da radiação , Proliferação de Células/efeitos da radiação , Cromatina/genética , Aberrações Cromossômicas , Ensaio de Unidades Formadoras de Colônias , Citoesqueleto/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Filaminas , Imunofluorescência , Instabilidade Genômica , Histonas/metabolismo , Humanos , Melanoma/patologia , Testes para Micronúcleos , Rad51 Recombinase/metabolismo , Radiação Ionizante
17.
Nucleic Acids Res ; 35(21): 7160-70, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17947333

RESUMO

Homologous recombination (HR) is critical for maintaining genome stability through precise repair of DNA double-strand breaks (DSBs) and restarting stalled or collapsed DNA replication forks. HR is regulated by many proteins through distinct mechanisms. Some proteins have direct enzymatic roles in HR reactions, while others act as accessory factors that regulate HR enzymatic activity or coordinate HR with other cellular processes such as the cell cycle. The breast cancer susceptibility gene BRCA2 encodes a critical accessory protein that interacts with the RAD51 recombinase and this interaction fluctuates during the cell cycle. We previously showed that a BRCA2- and p21-interacting protein, BCCIP, regulates BRCA2 and RAD51 nuclear focus formation, DSB-induced HR and cell cycle progression. However, it has not been clear whether BCCIP acts exclusively through BRCA2 to regulate HR and whether BCCIP also regulates the alternative DSB repair pathway, non-homologous end joining. In this study, we found that BCCIP fragments that interact with BRCA2 or with p21 each inhibit DSB repair by HR. We further show that transient down-regulation of BCCIP in human cells does not affect non-specific integration of transfected DNA, but significantly inhibits homology-directed gene targeting. Furthermore, human HT1080 cells with constitutive down-regulation of BCCIP display increased levels of spontaneous single-stranded DNA (ssDNA) and DSBs. These data indicate that multiple BCCIP domains are important for HR regulation, that BCCIP is unlikely to regulate non-homologous end joining, and that BCCIP plays a critical role in resolving spontaneous DNA damage.


Assuntos
Proteínas de Ligação ao Cálcio/química , Proteínas de Ligação ao Cálcio/fisiologia , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/fisiologia , Dano ao DNA , Proteínas Nucleares/química , Proteínas Nucleares/fisiologia , Recombinação Genética , Proteínas de Ligação ao Cálcio/antagonistas & inibidores , Proteínas de Ciclo Celular/antagonistas & inibidores , Linhagem Celular , Quebras de DNA de Cadeia Dupla , DNA de Cadeia Simples/análise , Regulação para Baixo , Marcação de Genes , Humanos , Proteínas Nucleares/antagonistas & inibidores , Estrutura Terciária de Proteína
18.
J Biol Chem ; 282(3): 1570-6, 2007 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-17135243

RESUMO

The tumor suppression function of p53 is mostly conferred by its transactivation activity, which is inactivated by p53 mutations in approximately 50% of human cancers. In cancers harboring wild type p53, the p53 transactivation activity may be compromised by other mechanisms. Identifying the mechanisms by which wild type p53 transactivation activity can be abrogated may provide insights into the molecular etiology of cancers harboring wild type p53. In this report, we show that BCCIP, a BRCA2 and CDKN1A-interacting protein, is required for the transactivation activity of wild type p53. In p53 wild type cells, BCCIP knock down by RNA interference diminishes the transactivation activity of p53 without reducing the p53 protein level, inhibits the binding of p53 to the promoters of p53 target genes p21 and HDM2, and reduces the tetrameric formation of p53. These data demonstrate a critical role of BCCIP in maintaining the transactivation activity of wild type p53 and further suggest down-regulation of BCCIP as a novel mechanism to impair the p53 function in cells harboring wild type p53.


Assuntos
Proteínas de Ligação ao Cálcio/biossíntese , Proteínas de Ciclo Celular/biossíntese , Regulação para Baixo , Genes p53/genética , Mutação , Neoplasias/genética , Proteínas Nucleares/biossíntese , Ativação Transcricional , Proteína Supressora de Tumor p53/fisiologia , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Humanos , Ligação Proteica , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteína Supressora de Tumor p53/metabolismo
19.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 24(6): 1295-300, 2007 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-18232480

RESUMO

Two kinds of chitosan of different molecular weight (50 kDa and 400 kDa) were employed to form nanoparticles with 32P-labeled plasmid DNA at different N/P ratios by complex coacervation method. The characteristics of chitosan gene nanoparticles (CGN) were measured. The cellular uptake of DNA nanoparticles was evaluated by A10 and K562 cells. The in vitro cytotoxicity of DNA nanoparticles was determined by the MTT assays. Cellular uptake of the DNA nanoparticles increased with increasing chitosan molecular weight and N/P ratio. It also correlated with the zeta potential of the DNA nanoparticles. Chitosan-DNA nanoparticles were much less cytotoxic when compared with Lipofectamine 2000-DNA nanoparticles.


Assuntos
Biopolímeros/toxicidade , Quitosana/toxicidade , DNA/toxicidade , Nanopartículas/toxicidade , Plasmídeos/toxicidade , Biopolímeros/química , Quitosana/química , Testes Imunológicos de Citotoxicidade , DNA/química , Técnicas de Transferência de Genes , Humanos , Células K562 , Nanopartículas/química , Plasmídeos/química
20.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 27(6): 718-22, 2005 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-16447645

RESUMO

OBJECTIVE: To evaluate the feasibility and stability of chemically conjugating IgM on collagen films. METHODS: IgM was labeled with 125I using the chloramine-T method. Six collagen films were randomly divided into two groups. In chemical coupling group 125I-labeled IgM was chemically coupled with the films through N-succinmiclyl-3- (2-pyridyl-dithio) propionate reaction. In control group 125I-labeled IgM was absorbed onto collagen films. The amount of IgM on the collagen films and the amount of IgM remained on the films after extensive rinsing with phosphate buffered saline were monitored by counting the radioactivity of 125I. RESULTS: The amount of antibodies loaded onto collagen films in the chemical coupling group was 15 times higher than that on the control films, showing significant statistical difference (P < 0.01). And the stability of conjugation antibodies on collagen films was significantly better than the control films. CONCLUSION: Chemical coupling is an effective approach to immobilize antibodies on collagen for further plasmid DNA tethering.


Assuntos
Anticorpos Antinucleares/metabolismo , Materiais Revestidos Biocompatíveis/metabolismo , Colágeno/metabolismo , Vetores Genéticos , Imunoglobulina M/metabolismo , Angioplastia Coronária com Balão/instrumentação , Animais , Bovinos , Materiais Revestidos Biocompatíveis/química , Colágeno/química , Técnicas In Vitro , Camundongos , Ligação Proteica , Stents , Propriedades de Superfície
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...