Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Am J Pathol ; 2024 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-39168365

RESUMO

Germline mutations of homologous-recombination (HR) genes are among the top contributors to medulloblastomas. A significant portion of human medulloblastomas exhibit genomic signatures of HR defects. We queried whether ablation of Brca2 and Palb2, and their related Brca1 and Bccip genes, in the mouse brain can differentially initiate medulloblastomas. Conditional knockout mouse models of these HR genes and a conditional knockdown of Bccip (shBccip-KD) were established. Deletion of any of these genes led to microcephaly and neurologic defects, with Brca1- and Bccip- producing the worst. Trp53 co-deletion significantly rescued the microcephaly with Brca1, Palb2, and Brca2 deficiency but exhibited limited impact on Bccip- mice. For the first time, inactivation of either Brca1 or Palb2 with Trp53 was found to induce medulloblastomas. Despite shBccip-CKD being highly penetrative, Bccip/Trp53 deletions failed to induce medulloblastomas. The tumors displayed diverse immunohistochemical features and chromosome copy number variation. Although there were widespread up-regulations of cell proliferative pathways, most of the tumors expressed biomarkers of the sonic hedgehog subgroup. The medulloblastomas developed from Brca1-, Palb2-, and Brca2- mice were highly sensitive to a poly (ADP-ribose) polymerase inhibitor but not the ones from shBccip-CKD mice. These models recapitulate the spontaneous medulloblastoma development with high penetrance and a narrow time window, providing ideal platforms to test therapeutic agents with the ability to differentiate HR-defective and HR-proficient tumors.

2.
Am J Pathol ; 191(1): 66-78, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33039352

RESUMO

BCCIP was originally identified as a BRCA2 and CDKN1A/p21 interaction protein. Although a partial loss of BCCIP function is sufficient to trigger genomic instability and tumorigenesis, complete deletion of BCCIP is lethal to cells. Using Rosa26-CreERT2 mouse models, we found that induced Bccip deletion in adult mice caused an acute intestinal epithelial denudation that cannot be relieved by co-deletion of Trp53. The critical role of Bccip in intestine epithelial renewal was verified with a Villin-CreERT2 mouse model. The epithelium degeneration was associated with a rapid loss of the proliferative capability of the crypt progenitor cells in vivo, lack of crypt base columnar stem cell markers, and a failure of in vitro crypt organoid growth. RNA-Seq analysis of freshly isolated intestinal crypt cells showed that Bccip deletion caused an overwhelming down-regulation of genes involved in mitotic cell division but an up-regulation of genes involved in apoptosis and stress response to microbiomes. Our data not only indicate that intestinal epithelium is the most sensitive tissue to whole-body deletion of Bccip but also point to Bccip as a novel and critical factor for the proliferation of the intestinal progenitors. These findings have significant implications for understanding why a hypomorphic loss of BCCIP functions is more relevant to tumorigenesis.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Mucosa Intestinal/metabolismo , Regeneração/fisiologia , Animais , Proliferação de Células/fisiologia , Camundongos , Células-Tronco/metabolismo
3.
Nucleic Acids Res ; 48(22): 12817-12832, 2020 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-33245766

RESUMO

Ribosome biogenesis is a fundamental process required for cell proliferation. Although evolutionally conserved, the mammalian ribosome assembly system is more complex than in yeasts. BCCIP was originally identified as a BRCA2 and p21 interacting protein. A partial loss of BCCIP function was sufficient to trigger genomic instability and tumorigenesis. However, a complete deletion of BCCIP arrested cell growth and was lethal in mice. Here, we report that a fraction of mammalian BCCIP localizes in the nucleolus and regulates 60S ribosome biogenesis. Both abrogation of BCCIP nucleolar localization and impaired BCCIP-eIF6 interaction can compromise eIF6 recruitment to the nucleolus and 60S ribosome biogenesis. BCCIP is vital for a pre-rRNA processing step that produces 12S pre-rRNA, a precursor to the 5.8S rRNA. However, a heterozygous Bccip loss was insufficient to impair 60S biogenesis in mouse embryo fibroblasts, but a profound reduction of BCCIP was required to abrogate its function in 60S biogenesis. These results suggest that BCCIP is a critical factor for mammalian pre-rRNA processing and 60S generation and offer an explanation as to why a subtle dysfunction of BCCIP can be tumorigenic but a complete depletion of BCCIP is lethal.


Assuntos
Carcinogênese/genética , Proteínas de Ciclo Celular/genética , Proliferação de Células/genética , Ribossomos/genética , Animais , Proteína BRCA2/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Fatores de Iniciação em Eucariotos/genética , Fibroblastos , Instabilidade Genômica/genética , Humanos , Camundongos , Células NIH 3T3 , Mapas de Interação de Proteínas/genética , RNA Ribossômico/genética , RNA Ribossômico 5,8S/genética , Subunidades Ribossômicas Maiores de Eucariotos/genética
4.
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
5.
Am J Pathol ; 190(6): 1175-1187, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32201259

RESUMO

Hepatocellular carcinoma (HCC) is the most common form of liver tumors. Although HCC is associated with chronic viral infections, alcoholic cirrhosis, and nonalcoholic fatty liver disease, genetic factors that contribute to the HCC risk remain unknown. The BRCA2 DNA repair associated (BRCA2) and cyclin-dependent kinase inhibitor 1A (CDKN1A) interacting protein, known as BCCIP, are essential for cell viability and maintenance of genomic stability. In this study, we established a new genetically engineered mouse model with Bccip deficiency. Mosaic or heterozygous Bccip deletion conferred an increased risk of spontaneous liver tumorigenesis and B-cell lymphoma development at old age. These abnormalities are accompanied with chronic inflammation, histologic features of nonalcoholic steatohepatitis, keratin and ubiquitin aggregates within cytoplasmic Mallory-Denk bodies, and changes of the intracellular distribution of high-mobility group box 1 protein. Our study suggests BCCIP dysregulation as a risk factor for HCC and offers a novel mouse model for future investigations of nonviral or nonalcoholic causes of HCC development.


Assuntos
Proteína BRCA2/genética , Carcinoma Hepatocelular/genética , Proteínas de Ciclo Celular/genética , Neoplasias Hepáticas/genética , Linfoma de Células B/genética , Animais , Proteína BRCA2/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proteínas de Ciclo Celular/metabolismo , Heterozigoto , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Linfoma de Células B/metabolismo , Linfoma de Células B/patologia , Camundongos , Camundongos Knockout , Mosaicismo
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.
Breast Cancer Res ; 19(1): 115, 2017 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-29047390

RESUMO

BACKGROUND: Dysregulated DNA repair and cell proliferation controls are essential driving forces in mammary tumorigenesis. BCCIP was originally identified as a BRCA2 and CDKN1A interacting protein that has been implicated in maintenance of genomic stability, cell cycle regulation, and microtubule dynamics. The aims of this study were to determine whether BCCIP deficiency contributes to mammary tumorigenesis, especially for a subset of breast cancers with 53BP1 abnormality, and to reveal the mechanistic implications of BCCIP in breast cancer interventions. METHODS: We analyzed the BCCIP protein level in 470 cases of human breast cancer to determine the associations between BCCIP and 53BP1, p53, and subtypes of breast cancer. We further constructed a unique BCCIP knockdown mouse model to determine whether a partial BCCIP deficiency leads to spontaneous breast cancer formation. RESULTS: We found that the BCCIP protein level is downregulated in 49% of triple-negative breast cancer and 25% of nontriple-negative breast cancer. The downregulation of BCCIP is mutually exclusive with p53 mutations but concurrent with 53BP1 loss in triple-negative breast cancer. In a K14-Cre-mediated conditional BCCIP knockdown mouse model, we found that BCCIP downregulation causes a formation of benign modules in the mammary glands, resembling the epidermal inclusion cyst of the breast. However, the majority of these benign lesions remain indolent, and only ~ 10% of them evolve into malignant tumors after a long latency. This tumor progression is associated with a loss of 53BP1 and p16 expression. BCCIP knockdown did not alter the latency of mammary tumor formation induced by conditional Trp53 deletion. CONCLUSIONS: Our data suggest a confounding role of BCCIP deficiency in modulating breast cancer development by enhancing tumor initiation but hindering progression. Furthermore, secondary genetic alternations may overcome the progression suppression imposed by BCCIP deficiency through a synthetic viability mechanism.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Carcinogênese/genética , Proteínas de Ciclo Celular/genética , Glândulas Mamárias Humanas/patologia , Proteínas Nucleares/genética , Animais , Proteína BRCA2/genética , Inibidor de Quinase Dependente de Ciclina p21/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Humanos , Glândulas Mamárias Humanas/metabolismo , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Camundongos , Neoplasias de Mama Triplo Negativas , Proteína Supressora de Tumor p53/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética
8.
Cancer Res ; 73(23): 7122-33, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24145349

RESUMO

Dysfunctions of genome caretaker genes contribute to genomic instability and tumor initiation. Because many of the caretaker genes are also essential for cell viability, permanent loss of function of these genes would prohibit further tumor progression. How essential caretaker genes contribute to tumorigenesis is not fully understood. Here, we report a "hit-and-run" mode of action for an essential caretaker gene in tumorigenesis. Using a BRCA2-interacting protein BCCIP as the platform, we found that a conditional BCCIP knockdown and concomitant p53 deletion caused rapid development of medulloblastomas, which bear a wide spectrum of alterations involving the Sonic Hedgehog (Shh) pathway, consistent with a caretaker responsibility of BCCIP on genomic integrity. Surprisingly, the progressed tumors have spontaneously lost the transgenic BCCIP knockdown cassette and restored BCCIP expression. Thus, a transient downregulation of BCCIP, but not necessarily a permanent mutation, is sufficient to initiate tumorigenesis. After the malignant transformation has been accomplished and autonomous cancer growth has been established, BCCIP reverses its role from a tumor-initiation suppressor to become a requisite for progression. This exemplifies a new type of tumor suppressor, which is distinct from the classical tumor suppressors that are often permanently abrogated during tumorigenesis. It has major implications on how a nonmutagenic or transient regulation of essential caretaker gene contributes to tumorigenesis. We further suggest that BCCIP represents a paradoxical class of modulators for tumorigenesis as a suppressor for initiation but a requisite for progression (SIRP).


Assuntos
Proteínas de Ligação ao Cálcio/fisiologia , Carcinogênese/genética , Proteínas de Ciclo Celular/fisiologia , Neoplasias/genética , Neoplasias/patologia , Proteínas Nucleares/fisiologia , Animais , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/patologia , Progressão da Doença , Genes Supressores de Tumor/fisiologia , Proteínas Hedgehog/fisiologia , Masculino , Meduloblastoma/genética , Meduloblastoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurogênese/genética , Transdução de Sinais/fisiologia
9.
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
10.
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
11.
PLoS One ; 7(1): e30638, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22292003

RESUMO

Multiple DNA repair pathways are involved in the orderly development of neural systems at distinct stages. The homologous recombination (HR) pathway is required to resolve stalled replication forks and critical for the proliferation of progenitor cells during neural development. BCCIP is a BRCA2 and CDKN1A interacting protein implicated in HR and inhibition of DNA replication stress. In this study, we determined the role of BCCIP in neural development using a conditional BCCIP knock-down mouse model. BCCIP deficiency impaired embryonic and postnatal neural development, causing severe ataxia, cerebral and cerebellar defects, and microcephaly. These development defects are associated with spontaneous DNA damage and subsequent cell death in the proliferative cell populations of the neural system during embryogenesis. With in vitro neural spheroid cultures, BCCIP deficiency impaired neural progenitor's self-renewal capability, and spontaneously activated p53. These data suggest that BCCIP and its anti-replication stress functions are essential for normal neural development by maintaining an orderly proliferation of neural progenitors.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Proliferação de Células , Células-Tronco Neurais/fisiologia , Neurogênese/genética , Neurônios/fisiologia , Animais , Ataxia/complicações , Ataxia/congênito , Ataxia/genética , Ataxia/patologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Embrião de Mamíferos , Proteína Glial Fibrilar Ácida/genética , Transtornos do Crescimento/complicações , Transtornos do Crescimento/congênito , Transtornos do Crescimento/genética , Transtornos do Crescimento/patologia , Integrases/genética , Integrases/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Células-Tronco Neurais/metabolismo , Neurônios/metabolismo , Especificidade de Órgãos/genética , Equilíbrio Postural/genética , Regiões Promotoras Genéticas/genética , Transtornos de Sensação/complicações , Transtornos de Sensação/congênito , Transtornos de Sensação/genética , Transtornos de Sensação/patologia , Células-Tronco/metabolismo , Células-Tronco/fisiologia
12.
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
13.
PLoS Genet ; 7(9): e1002291, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21966279

RESUMO

BCCIP is a BRCA2- and CDKN1A(p21)-interacting protein that has been implicated in the maintenance of genomic integrity. To understand the in vivo functions of BCCIP, we generated a conditional BCCIP knockdown transgenic mouse model using Cre-LoxP mediated RNA interference. The BCCIP knockdown embryos displayed impaired cellular proliferation and apoptosis at day E7.5. Consistent with these results, the in vitro proliferation of blastocysts and mouse embryonic fibroblasts (MEFs) of BCCIP knockdown mice were impaired considerably. The BCCIP deficient mouse embryos die before E11.5 day. Deletion of the p53 gene could not rescue the embryonic lethality due to BCCIP deficiency, but partially rescues the growth delay of mouse embryonic fibroblasts in vitro. To further understand the cause of development and proliferation defects in BCCIP-deficient mice, MEFs were subjected to chromosome stability analysis. The BCCIP-deficient MEFs displayed significant spontaneous chromosome structural alterations associated with replication stress, including a 3.5-fold induction of chromatid breaks. Remarkably, the BCCIP-deficient MEFs had a ∼20-fold increase in sister chromatid union (SCU), yet the induction of sister chromatid exchanges (SCE) was modestly at 1.5 fold. SCU is a unique type of chromatid aberration that may give rise to chromatin bridges between daughter nuclei in anaphase. In addition, the BCCIP-deficient MEFs have reduced repair of irradiation-induced DNA damage and reductions of Rad51 protein and nuclear foci. Our data suggest a unique function of BCCIP, not only in repair of DNA damage, but also in resolving stalled replication forks and prevention of replication stress. In addition, BCCIP deficiency causes excessive spontaneous chromatin bridges via the formation of SCU, which can subsequently impair chromosome segregations in mitosis and cell division.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Instabilidade Cromossômica/genética , Desenvolvimento Embrionário/genética , Animais , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Proteínas de Ciclo Celular/genética , Segregação de Cromossomos , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA/genética , Reparo do DNA/genética , Replicação do DNA/genética , Fibroblastos/citologia , Fibroblastos/metabolismo , Camundongos , Camundongos Transgênicos , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , Recombinação Genética , Troca de Cromátide Irmã
14.
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
15.
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
16.
BMC Cancer ; 9: 268, 2009 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-19653894

RESUMO

BACKGROUND: Loss of heterozygosity of chromosome 10q26 has been shown to be associated with the aggressiveness of astrocytic tumors (or astrocytomas), but the responsible gene(s) residing in this region has not been fully identified. The BCCIP gene is located at chromosome 10q26. It encodes a BRCA2 and CDKN1A (p21) interacting protein. Previous studies have shown that down-regulation of BCCIP impairs recombinational DNA repair, G1/S cell cycle checkpoint, p53 trans-activation activity, cytokinesis, and chromosome stability, suggesting a potential role of BCCIP in cancer etiology. In this study, we investigated whether BCCIP is altered in astrocytomas. METHODS: Genomic DNA from 45 cases of grade IV astrocytic tumor (glioblastoma) tissues and 12 cases of normal tissues were analyzed by quantitative PCR. The BCCIP protein expression in 96 cases of grade II-IV astrocytic tumors was detected by immunohistochemistry (IHC). IHC staining of glial fibrillary acid protein (GFAP), a marker for astrocytic cells, was used to identify cells of the astrocytic lineage. RESULTS: We found that BCCIP protein is expressed in normal cells with positive staining of GFAP. However, BCCIP protein expression was not detectable in approximately 45% of all astrocytic tumors, and in > 60% in the grade IV glioblastoma. About 45% glioblastoma have significant (p < 0.01) reduction of BCCIP gene copy number when compared to normal DNA. Furthermore, the frequency of lacking BCCIP expression is associated with the aggressiveness of astrocytic tumors. CONCLUSION: Our data implicate a role of BCCIP in astrocytic tumorigenesis, and lack of BCCIP may be used as a marker for astrocytomas.


Assuntos
Astrocitoma/metabolismo , Proteína BRCA2/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Nucleares/metabolismo , Astrocitoma/genética , Astrocitoma/patologia , Proteína BRCA2/genética , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ciclo Celular/genética , Dosagem de Genes , Humanos , Proteínas Nucleares/genética , Ligação Proteica
17.
Radiother Oncol ; 90(2): 183-8, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19046788

RESUMO

BACKGROUND: Recent studies have shown that BCCIP (BRCA2 and CDKN1A interacting protein) is essential for maintaining the transactivation activity of wild type p53. We analyzed the expression of BCCIP and p53 in a cohort of laryngeal cancer treated with radiotherapy and assessed whether BCCIP and p53, alone or in combination, would correlate with local control and overall survival. METHODS: One hundred twenty-three patients treated between 1975 and 2000 for early stage (stages I and II) squamous cell carcinoma of the larynx were included in the study. Treatment consisted of radiation therapy (RT) with standard fields and fractionation to a median dose of 66Gy. Tissue was collected from pre-RT biopsies and constructed in a tissue microarray, and BCCIP expression and p53 expression were determined using immunohistochemistry. RESULTS: Loss of expression of BCCIP in combination with normal p53 (negative p53 staining) was associated with local recurrence (RR 2.04; 95% CI 0.99-4.56, p=0.05) and poor overall survival (RR 2.09; 95% CI 1.21-4.00, p=0.008) compared to patients who did express BCCIP. Expression of BCCIP or p53 alone was not found to be independently associated with benefits in local control or overall survival. CONCLUSIONS: This study provides clinical evidence that BCCIP contributes to outcomes in patients with laryngeal cancer treated with RT. This benefit may be a result of increased radiosensitivity in patients who have functional BCCIP and p53. These data may be used to identify sub-groups of laryngeal cancer patients who are more likely to be cured with radiotherapy.


Assuntos
Biomarcadores Tumorais/análise , Proteínas de Ligação ao Cálcio/análise , Carcinoma de Células Escamosas/radioterapia , Proteínas de Ciclo Celular/análise , Neoplasias Laríngeas/radioterapia , Proteínas Nucleares/análise , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/química , Carcinoma de Células Escamosas/mortalidade , Feminino , Humanos , Neoplasias Laríngeas/química , Neoplasias Laríngeas/mortalidade , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/diagnóstico , Prognóstico , Taxa de Sobrevida , Proteína Supressora de Tumor p53/análise
18.
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
19.
Cancer Res ; 65(5): 1647-54, 2005 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-15753358

RESUMO

Increased amounts of reactive oxygen species (ROS) induce apoptosis in mammalian cells. PUMA (P53 up-regulated modulator of apoptosis), a mitochondrial proapoptotic BH3-only protein, induces rapid apoptosis through a Bax- and mitochondria-dependent pathway. However, the molecular basis of PUMA-induced apoptosis is largely not understood. Using a combination of biophysical and biochemical methods and PUMA-inducible colorectal cells, DLD-1.PUMA, we showed that (a) PUMA-induced apoptosis is dose and time dependent; (b) PUMA-induced apoptosis is directly associated with ROS generation; (c) diphenyleneiodonium chloride, a ROS blocker, or BAX-inhibiting peptide, a suppressor of BAX translocation, decreased ROS generation and apoptosis in DLD-1.PUMA cells; (d) overexpression of PUMA induced up-regulation (>1.34-fold) of peroxiredoxin 1 and down-regulation (by 25%) of stathmin through proteasome-mediated degradation; and (e) hydrogen peroxide down-regulated stathmin and disrupted the cellular microtubule network. Our findings indicate that PUMA induces apoptosis, in part, through the BAX-dependent generation of superoxide and hydrogen peroxide. ROS overproduction and oxidative stress induce proteome-wise alterations, such as stathmin degradation and disorganization of the cell microtubule network, in apoptotic cells.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Proteínas dos Microtúbulos/metabolismo , Fosfoproteínas/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Reguladoras de Apoptose , Neoplasias Colorretais/patologia , Inibidores Enzimáticos/farmacologia , Humanos , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Proteínas dos Microtúbulos/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Óxido Nítrico Sintase/antagonistas & inibidores , Oniocompostos/farmacologia , Fragmentos de Peptídeos , Peroxidases/metabolismo , Peroxirredoxinas , Fosfoproteínas/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Estatmina , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/deficiência , Proteína X Associada a bcl-2
20.
Mol Cell Biol ; 25(5): 1949-57, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15713648

RESUMO

Homologous recombinational repair (HRR) of DNA damage is critical for maintaining genome stability and tumor suppression. RAD51 and BRCA2 colocalization in nuclear foci is a hallmark of HRR. BRCA2 has important roles in RAD51 focus formation and HRR of DNA double-strand breaks (DSBs). We previously reported that BCCIPalpha interacts with BRCA2. We show that a second isoform, BCCIPbeta, also interacts with BRCA2 and that this interaction occurs in a region shared by BCCIPalpha and BCCIPbeta. We further show that chromatin-bound BRCA2 colocalizes with BCCIP nuclear foci and that most radiation-induced RAD51 foci colocalize with BCCIP. Reducing BCCIPalpha by 90% or BCCIPbeta by 50% by RNA interference markedly reduces RAD51 and BRCA2 foci and reduces HRR of DSBs by 20- to 100-fold. Similarly, reducing BRCA2 by 50% reduces RAD51 and BCCIP foci. These data indicate that BCCIP is critical for BRCA2- and RAD51-dependent responses to DNA damage and HRR.


Assuntos
Proteína BRCA2/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Proteínas Nucleares/metabolismo , Recombinação Genética/fisiologia , Proteína BRCA2/análise , Proteínas de Ligação ao Cálcio/análise , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ciclo Celular/análise , Proteínas de Ciclo Celular/genética , Núcleo Celular/química , Núcleo Celular/metabolismo , Células Cultivadas , Dano ao DNA , Proteínas de Ligação a DNA/análise , Humanos , Proteínas Nucleares/análise , Proteínas Nucleares/genética , Mapeamento de Interação de Proteínas , Isoformas de Proteínas/análise , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética , Rad51 Recombinase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA