Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Am J Pathol ; 2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39168365

RESUMEN

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

2.
Am J Pathol ; 191(1): 66-78, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33039352

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Mucosa Intestinal/metabolismo , Regeneración/fisiología , Animales , Proliferación Celular/fisiología , Ratones , Células Madre/metabolismo
3.
Nucleic Acids Res ; 48(22): 12817-12832, 2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33245766

RESUMEN

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.


Asunto(s)
Carcinogénesis/genética , Proteínas de Ciclo Celular/genética , Proliferación Celular/genética , Ribosomas/genética , Animales , Proteína BRCA2/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Factores Eucarióticos de Iniciación/genética , Fibroblastos , Inestabilidad Genómica/genética , Humanos , Ratones , Células 3T3 NIH , Mapas de Interacción de Proteínas/genética , ARN Ribosómico/genética , ARN Ribosómico 5.8S/genética , Subunidades Ribosómicas Grandes de Eucariotas/genética
4.
Sensors (Basel) ; 22(22)2022 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-36433481

RESUMEN

Many researchers have introduced blockchain into the Internet of Vehicles (IoV) to support trading or other authentication applications between vehicles. However, the traditional blockchain cannot well support the query of transactions that occur in a specified area which is important for vehicle users since they are bound to the geolocations. Therefore, the querying efficiency of the geolocation attribute of transactions is vital for blockchain-based applications. Existing work does not well handle the geolocation of vehicles in the blockchain, and thus the querying efficiency is questionable. In this paper, we design a rapid query method of regional transactions in blockchain for IoV, including data structures and query algorithms. The main idea is to utilize the Geohash code to represent the area and serve as the key for transaction indexing and querying, and the geolocation is marked as one of the attributes of transactions in the blockchain. To further verify and evaluate the proposed design, on the basis of the implementation of Ethereum, which is a well-known blockchain, the results show that the proposed design achieves significantly better-querying speed than Ethereum.


Asunto(s)
Cadena de Bloques , Seguridad Computacional , Internet , Algoritmos
5.
Am J Pathol ; 190(6): 1175-1187, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32201259

RESUMEN

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.


Asunto(s)
Proteína BRCA2/genética , Carcinoma Hepatocelular/genética , Proteínas de Ciclo Celular/genética , Neoplasias Hepáticas/genética , Linfoma de Células B/genética , Animales , Proteína BRCA2/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular/metabolismo , Heterocigoto , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Ratones , Ratones Noqueados , Mosaicismo
6.
Breast Cancer Res ; 19(1): 115, 2017 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-29047390

RESUMEN

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.


Asunto(s)
Proteínas de Unión al Calcio/genética , Carcinogénesis/genética , Proteínas de Ciclo Celular/genética , Glándulas Mamarias Humanas/patología , Proteínas Nucleares/genética , Animales , Proteína BRCA2/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Humanos , Glándulas Mamarias Humanas/metabolismo , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/patología , Ratones , Neoplasias de la Mama Triple Negativas , Proteína p53 Supresora de Tumor/genética , Proteína 1 de Unión al Supresor Tumoral P53/genética
7.
PLoS Genet ; 7(9): e1002291, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21966279

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Inestabilidad Cromosómica/genética , Desarrollo Embrionario/genética , Animales , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Proteínas de Ciclo Celular/genética , Segregación Cromosómica , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN/genética , Reparación del ADN/genética , Replicación del ADN/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Ratones , Ratones Transgénicos , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Recombinación Genética , Intercambio de Cromátides Hermanas
8.
bioRxiv ; 2024 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-38979328

RESUMEN

The mammalian non-homologous end joining (NHEJ) is required for V(D)J recombination as well as coping with exogenously induced DNA double strand breaks (DSBs). Initiated by the binding of KU70/KU80 (KU) dimer to DNA ends and the subsequent recruitment of the DNA- dependent protein kinase catalytic subunit (DNA-PKcs), NHEJ plays a key role in DNA repair. While there has been significant structural understandings of how KU70 participates in NHEJ, the specific function of its highly conserved C-terminal SAP domain remains elusive. In this study, we developed a novel mouse model by deleting the SAP domain but preserving the KU70 nuclear localization and its dimerization ability with KU80. We found that the KU70 SAP deletion did not affect the V(D)J recombination or animal development but significantly impaired the animals and cells in repairing exogenously induced DSBs. We further showed an inability of KU70-ΔSAP cells to retain the DNA Ligase IV (LIG4) and other NHEJ co-factors on chromatin, and a spreading pattern of DSB marker γH2AX in KU70-ΔSAP cells after DNA damage. Our findings suggest that a specific inhibition of the SAP function may offer an opportunity to modulate cell sensitivity to therapeutic DSB-inducing agents without interfering with the developmental function of KU70. KeyPoints: Generation of a novel transgenic mouse line lacking the C-terminal conserved KU70-SAP domainKU70-SAP defends against exogenous DSBs, but unessential for development and V(D)J recombinationKU70-SAP aids in recruiting and retaining NHEJ components, such as LIG4, to DSB sites.

9.
Cell Rep ; 39(6): 110794, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35545041

RESUMEN

The mammalian KU70 is a pleiotropic protein functioning in DNA repair and cytoplasmic suppression of apoptosis. We report a regulatory mechanism by which KU70's cytoplasmic function is enabled due to a methylation at K570 of KU70 by SET-domain-containing protein 4 (SETD4). While SETD4 silencing reduces the level of methylated KU70, over-expression of SETD4 enhances methylation of KU70. Mutations of Y272 and Y284 of SETD4 abrogate methylation of KU70. Although SETD4 is predominantly a nuclear protein, the methylated KU70 is enriched in the cytoplasm. SETD4 knockdown enhances staurosporine (STS)-induced apoptosis and cell killing. Over-expression of the wild-type (WT) SETD4, but not the SETD4-Y272/Y284F mutant, suppresses STS-induced apoptosis. The KU70-K570R (mouse Ku70-K568R) mutation dampens the anti-apoptosis activity of KU70. Our study identifies KU70 as a non-histone substrate of SETD4, discovers a post-translational modification of KU70, and uncovers a role for SETD4 and KU70-K570 methylation in the suppression of apoptosis.


Asunto(s)
Apoptosis , Reparación del ADN , Animales , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Apoptosis/genética , Citoplasma/metabolismo , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Mamíferos/metabolismo , Metilación , Metiltransferasas , Ratones , Procesamiento Proteico-Postraduccional
10.
DNA Repair (Amst) ; 86: 102754, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31794893

RESUMEN

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.


Asunto(s)
Eliminación de Gen , Linfoma/genética , Metiltransferasas/genética , Neoplasias Inducidas por Radiación/genética , Neoplasias del Timo/genética , Animales , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Linfoma/inmunología , Linfoma/mortalidad , Ratones , Neoplasias Inducidas por Radiación/inmunología , Neoplasias Inducidas por Radiación/mortalidad , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Análisis de Secuencia de ADN , Neoplasias del Timo/inmunología , Neoplasias del Timo/mortalidad , Proteína p53 Supresora de Tumor/genética
11.
Int J Radiat Oncol Biol Phys ; 107(4): 779-792, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32259569

RESUMEN

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.


Asunto(s)
Hematopoyesis/genética , Hematopoyesis/efectos de la radiación , Metiltransferasas/deficiencia , Metiltransferasas/genética , Animales , Trasplante de Médula Ósea , Técnicas de Inactivación de Genes , Ratones , Irradiación Corporal Total/efectos adversos
12.
Radiother Oncol ; 90(2): 183-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19046788

RESUMEN

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.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas de Unión al Calcio/análisis , Carcinoma de Células Escamosas/radioterapia , Proteínas de Ciclo Celular/análisis , Neoplasias Laríngeas/radioterapia , Proteínas Nucleares/análisis , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/química , Carcinoma de Células Escamosas/mortalidad , Femenino , Humanos , Neoplasias Laríngeas/química , Neoplasias Laríngeas/mortalidad , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/diagnóstico , Pronóstico , Tasa de Supervivencia , Proteína p53 Supresora de Tumor/análisis
13.
BMC Cancer ; 9: 268, 2009 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-19653894

RESUMEN

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.


Asunto(s)
Astrocitoma/metabolismo , Proteína BRCA2/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Nucleares/metabolismo , Astrocitoma/genética , Astrocitoma/patología , Proteína BRCA2/genética , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/genética , Dosificación de Gen , Humanos , Proteínas Nucleares/genética , Unión Proteica
14.
Nucleic Acids Res ; 35(21): 7160-70, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17947333

RESUMEN

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.


Asunto(s)
Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/fisiología , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/fisiología , Daño del ADN , Proteínas Nucleares/química , Proteínas Nucleares/fisiología , Recombinación Genética , Proteínas de Unión al Calcio/antagonistas & inhibidores , Proteínas de Ciclo Celular/antagonistas & inhibidores , Línea Celular , Roturas del ADN de Doble Cadena , ADN de Cadena Simple/análisis , Regulación hacia Abajo , Marcación de Gen , Humanos , Proteínas Nucleares/antagonistas & inhibidores , Estructura Terciaria de Proteína
15.
Mol Cell Biol ; 25(5): 1949-57, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15713648

RESUMEN

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.


Asunto(s)
Proteína BRCA2/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Recombinación Genética/fisiología , Proteína BRCA2/análisis , Proteínas de Unión al Calcio/análisis , Proteínas de Unión al Calcio/genética , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/genética , Núcleo Celular/química , Núcleo Celular/metabolismo , Células Cultivadas , Daño del ADN , Proteínas de Unión al ADN/análisis , Humanos , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Mapeo de Interacción de Proteínas , Isoformas de Proteínas/análisis , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Interferencia de ARN , ARN Interferente Pequeño/genética , Recombinasa Rad51
16.
Cancer Res ; 65(5): 1647-54, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15753358

RESUMEN

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.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Proteínas de Microtúbulos/metabolismo , Fosfoproteínas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Reguladoras de la Apoptosis , Neoplasias Colorrectales/patología , Inhibidores Enzimáticos/farmacología , Humanos , Peróxido de Hidrógeno/metabolismo , Peróxido de Hidrógeno/farmacología , Proteínas de Microtúbulos/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Compuestos Onio/farmacología , Fragmentos de Péptidos , Peroxidasas/metabolismo , Peroxirredoxinas , Fosfoproteínas/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Estatmina , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/deficiencia , Proteína X Asociada a bcl-2
17.
Mol Cancer Res ; 3(1): 21-31, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15671246

RESUMEN

Cancer cells frequently possess defects in the genetic and biochemical pathways of apoptosis. Members of the Bcl-2 family play pivotal roles in regulating apoptosis and possess at least one of four Bcl-2 homology (BH) domains, designated BH1 to BH4. The BH3 domain is the only one conserved in proapoptotic BH3-only proteins and plays an important role in protein-protein interactions in apoptosis by regulating homodimerization and heterodimerization of the Bcl-2 family members. To date, 10 BH3-only proapoptotic proteins have been identified and characterized in the human genome. The completion of the Human Genome Project and the availability of various public databases and sequence analysis algorithms allowed us to use the bioinformatic database-mining approach to identify one novel BH3-only protein, apolipoprotein L6 (ApoL6). The full-length cDNA of ApoL6 was identified, cloned, and functionally expressed in p53-null colorectal cancer cells (DLD-1). We found that overexpression of wild-type ApoL6 induced mitochondria-mediated apoptosis in DLD-1 cells characterized by release of cytochrome c and Smac/DIABLO from mitochondria and activation of caspase-9, whereas ApoL6 BH3 domain deletion allele did not. In addition, overexpression of ApoL6 also induced activation of caspase-8. Furthermore, we showed that adenovirus harboring the full-length cDNA of ApoL6 induced marked apoptosis in a variety of cancer cell types, and ApoL6 recruited and interacted with lipid/fatty acid components during the induction of apoptosis. To our knowledge, this is the first example that intracellular overproduction of an apolipoprotein induces marked apoptosis.


Asunto(s)
Apolipoproteínas/fisiología , Apoptosis , Mitocondrias/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Alelos , Secuencia de Aminoácidos , Apolipoproteínas/metabolismo , Apolipoproteínas L , Caspasa 8 , Caspasa 9 , Caspasas/metabolismo , Muerte Celular , Línea Celular , Línea Celular Tumoral , Citocromos c/metabolismo , ADN Complementario/metabolismo , Bases de Datos como Asunto , Ácidos Grasos/metabolismo , Citometría de Flujo , Eliminación de Gen , Genoma Humano , Humanos , Immunoblotting , Inmunoprecipitación , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutagénesis , Péptidos/química , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-bcl-2/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Factores de Tiempo , Transfección , Proteína p53 Supresora de Tumor/metabolismo
18.
Oncol Rep ; 10(2): 375-9, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12579275

RESUMEN

The present study was designed to investigate the protective effect of a dietary water-soluble extract from cultured medium of Ganoderma lucidum (Rei-shi or Mannentake) mycelia (designated as MAK) on the induction and development of azoxymethane (AOM)-induced colon tumors in male F344/Du Crj rats. A total of 80 animals were divided into five groups at six weeks of age, groups 2, 3 and 4 being given weekly subcutaneous injections of AOM (15 mg/kg body weight) for the initial 3 weeks to induce colon tumors. Rats in group 1 and 5 were injected with the vehicle, 0.9% (w/v) saline, following the same schedule. Rats in groups 1, 2, 3, 4 and 5 were fed MF, MF, 1.25% MAK, 2.5% MAK and 2.5% MAK diets, respectively, starting 1 week before AOM treatment and throughout the six-month experimental period. There were no significant differences in number of ACF, total AC and AC per site among groups 2 to 4, but the tumor incidence was significantly lower, and tumor size was smaller in group 4 (AOM + 2.5% MAK) than in group 2 (AOM + MF). Additionally, beta-catenin positive tumor cell nuclei were significantly decreased in the MAK-fed rats (groups 3 and 4), which also demonstrated lowering of the PCNA labeling index and a shortened germinal region in the colon. The present results thus indicate that dietary MAK could act as a potent chemopreventive agent for colon carcinogenesis.


Asunto(s)
Adenoma/prevención & control , Anticarcinógenos/farmacología , Neoplasias del Colon/prevención & control , Extractos Vegetales/farmacología , Reishi , Adenoma/inducido químicamente , Adenoma/metabolismo , Adenoma/patología , Animales , Azoximetano/toxicidad , Biomarcadores de Tumor/metabolismo , División Celular/efectos de los fármacos , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Medios de Cultivo , Técnicas para Inmunoenzimas , Masculino , Micelio , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Endogámicas F344 , Células Tumorales Cultivadas
19.
Oncol Rep ; 9(1): 69-73, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11748458

RESUMEN

The present study was designed to investigate the effects of fermented miso in the diet on the induction of aberrant crypt foci (ACF) by azoxymethane (AOM) in male F344 rats. A total of 50 rats, 8 weeks of age, were divided into 5 groups and given weekly subcutaneous injections of AOM (15 mg/kg body wt) for 3 weeks. Rats were fed a normal control MF solid diet, or solid diet containing 10% long-term fermented (aged), medium- or short-term fermented miso, or 2.2% NaCl for 5 weeks, starting one week before the first AOM dosing. It was found that, compared to the control (MF) diet, the long-term fermented diet significantly decreased (by 22.2%) ACF/colon, but increased (by 18.2%) the number of aberrant crypts (Acs)/focus. The latter was also increased by the medium-term fermented diet (by 25.3%). The PCNA labeling index was only affected by the short-term fermented diet (36.9% increase) and by 2.2% NaCl diet (27.2% increased). The present results indicate that aged or completely fermented miso supplemented into the diet, could act as a chemopreventive agent for colon carcinogenesis.


Asunto(s)
Neoplasias del Colon/dietoterapia , Proteínas en la Dieta/administración & dosificación , Lesiones Precancerosas/dietoterapia , Proteínas de Soja/administración & dosificación , Animales , Azoximetano/toxicidad , Peso Corporal , Carcinógenos/toxicidad , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/metabolismo , Fermentación , Técnicas para Inmunoenzimas , Masculino , Tamaño de los Órganos , Lesiones Precancerosas/inducido químicamente , Lesiones Precancerosas/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ratas , Ratas Endogámicas F344
20.
Oncol Rep ; 9(3): 613-6, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-11956637

RESUMEN

The present study was designed to investigate the effects of fermented miso in the diet on the induction of gastric tumors by N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) in male CD (SD) rats. A total of 120 animals, 6 weeks of age, were divided into 6 groups and given MNNG (100 ppm) in the drinking water for 16 weeks. Starting 1 week before the carcinogen treatment the rats were fed a normal control MF solid diet, or the same diet containing 10% long-term fermented, medium- or short-term fermented miso, or 1% NaCl until the end of the MNNG exposure period. They were then maintained on the MF control diet and normal tap water until the autopsy time point at 52 weeks. The long-term fermented miso significantly reduced the size of the gastric tumors as compared with the other groups. The present results thus indicate that dietary supplementation with long-term fermented miso could act as a chemopreventive agent for gastric carcinogenesis.


Asunto(s)
Carcinógenos , Metilnitronitrosoguanidina , Extractos Vegetales/uso terapéutico , Neoplasias Gástricas/inducido químicamente , Neoplasias Gástricas/prevención & control , Animales , Avena/metabolismo , Peso Corporal/efectos de los fármacos , Fermentación , Masculino , Tamaño de los Órganos/efectos de los fármacos , Oryza/metabolismo , Ratas , Glycine max/metabolismo , Factores de Tiempo , Triticum/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda