Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 668: 42-48, 2023 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-37244033

RESUMEN

Camptothecin (CPT) exhibits strong cytotoxicity by inducing DNA double-strand breaks (DSBs) through DNA replication. Unlike radiation-induced DSBs, which have two DNA ends, CPT-induced DSBs are considered to have only one DNA end. However, the differences in cellular responses to one-ended and two-ended DSBs are not well understood. Our previous study showed that proteasome inhibitor treatment suppressed CPT-induced activation of DNA-PK, a factor required for non-homologous end-joining in DSB repair, suggesting that the ubiquitin-proteasome pathway is involved in DNA-PK activation in response to one-ended DSBs. To identify the ubiquitination factors required for DNA-PK activation, we screened an siRNA library against E2 ubiquitin-conjugating enzymes and identified UbcH5c. Knockdown of UbcH5c suppressed DNA-PK activation caused by CPT, but not by the radio-mimetic drug neocarzinostatin. UbcH5c-dependent DNA-PK activation occurred independent of DNA end resection. Furthermore, loss of UbcH5c reduced DNA-PK-dependent chromosomal aberrations and suppressed the activation of cell cycle checkpoint in response to CPT. These results suggest that UbcH5c regulates DNA-PK activation in response to one-ended DSBs caused by replication fork collapse. To our knowledge, this is the first report of a DSB repair-related factor that is differentially involved in the response to one- and two-ended DSBs.


Asunto(s)
Roturas del ADN de Doble Cadena , Proteína Quinasa Activada por ADN , Proteína Quinasa Activada por ADN/metabolismo , Replicación del ADN , ADN/metabolismo , Reparación del ADN por Unión de Extremidades , Reparación del ADN
2.
DNA Repair (Amst) ; 113: 103318, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35325630

RESUMEN

DNA-damaging anti-cancer drugs are used clinically to induce cell death by causing DNA strand breaks or DNA replication stress. Camptothecin (CPT) and cisplatin are commonly used anti-cancer drugs, and their combined use enhances the anti-tumour effects. However, the mechanism underlying this enhanced effect has not been well studied. In this study, we analysed the combined effect of CPT and cisplatin or ultraviolet (UV) and found that CPT suppresses transcription recovery after UV damage and induces the disappearance of the Cockayne syndrome group B (CSB) protein, a transcription-coupled nucleotide excision repair (TC-NER) factor. This CPT-induced disappearance of CSB expression was suppressed by proteasome and transcription inhibitors. Moreover, CSB ubiquitination was detected after CPT treatment in a transcription-dependent manner, suggesting that the transcription stress caused by CPT induces CSB ubiquitination, resulting in CSB undetectability. However, Cockayne syndrome group A (CSA) and CUL4A were not involved in the CPT-induced CSB undetectability, suggesting that CSB ubiquitination caused by CPT is regulated differently from the UV response. However, cisplatin or UV sensitivity was enhanced by CPT even in CSB- or CSA-knockout cells. Furthermore, the excessive CSB expression, which suppressed CSB ubiquitination, did not cancel the combined effect of CPT. These results suggest that CPT blocks the repair of cisplatin or UV-induced DNA damage regardless of TC-NER status. CPT possibly compromised the alternative repair pathways other than TC-NER, leading to the suppression of transcription recovery and enhancement of cell killing.

3.
Mech Ageing Dev ; 196: 111492, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33862037

RESUMEN

Tropomyosin (Tpm) 1 and 2 are important in the epithelial mesenchymal transition of lens epithelial cells; however, the effect of Tpm1 depletion during aging remains obscure. We analyzed the age-related changes in the crystalline lens of Tpm1- conditional knockout mice (Tpm1-CKO). Floxed alleles of Tpm1 were conditionally deleted in the lens, using Pax6-cre transgenic mice. Lenses of embryonic day (ED) 14, postnatal 1-, 11-, and 48-week-old Tpm1-CKO and wild type mice were dissected to prepare paraffin sections, which subsequently underwent histological and immunohistochemical analysis. Tpm1 and α smooth muscle actin (αSMA) mRNA expression were assessed using RT-PCR. The homozygous Tpm1-CKO (Tpm1-/-) lenses displayed a dramatic reduction in Tpm1 transcript, with no change to αSMA mRNA expression. Tpm1-/- mice had small lenses with disorganized, vesiculated fiber cells, and loss of epithelial cells. The lenses of Tpm1-/- mice had abnormal and disordered lens fiber cells with cortical and peri-nuclear liquefaction. Expression of filamentous-actin was reduced in the equator region of lenses derived from ED14, 1-, 11-, and 48-week-old Tpm1-/- mice. Therefore, Tpm1 plays an integral role in mediating the integrity and fate of lens fiber differentiation and lens homeostasis during aging. Age-related Tpm1 dysregulation or deficiency may induce cataract formation.


Asunto(s)
Actinas/metabolismo , Envejecimiento/fisiología , Catarata , Senescencia Celular/fisiología , Tropomiosina/genética , Animales , Catarata/metabolismo , Catarata/patología , Catarata/fisiopatología , Diferenciación Celular , Transición Epitelial-Mesenquimal/fisiología , Perfilación de la Expresión Génica , Inmunohistoquímica , Cristalino/metabolismo , Cristalino/patología , Ratones , Ratones Noqueados , ARN Mensajero
4.
Genes Cells ; 26(5): 313-327, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33662167

RESUMEN

As the sentinels of innate and adaptive immune system, dendritic cells (DCs) have been considered to hold a great promise for medical application. Among the diverse types of DCs, monocyte-derived DCs (mo-DCs) generated in vitro have been most commonly employed. We have been improving the culture protocol and devised a protocol to produce mature interferon-α-induced DCs (IFN-DCs), hereinafter called (mat)IFN-DCs. While exploring the relationship between the expression of CD56 and the cytotoxic activity of (mat)IFN-DCs, we unexpectedly found that sorting of (mat)IFN-DCs with CD56 antibody-coated microbeads (MB) resulted in fractionating cells with tumoricidal activity into the flow-through (FT) but not MB-bound fraction. We uncovered that the FT fraction contains cells expressing low but substantial level of CD56. Moreover, those cells express granzyme B (GrB), perforin (PFN), and serpin B9 at high levels. By employing a specific inhibitor of PFN, we confirmed that direct tumoricidal activity relies on the GrB/PFN pathway. We designated subpopulation in FT fraction as CD56dim and that in CD56 positively sorted fraction as CD56bright , respectively. This is the first time, to our knowledge, to identify subpopulations of CD56-positive IFN-DCs with distinct tumoricidal activity which is ascribed to high expression of the components of GrB/PFN pathway.


Asunto(s)
Antígeno CD56/metabolismo , Células Dendríticas/metabolismo , Granzimas/metabolismo , Interferón-alfa/farmacología , Perforina/metabolismo , Serpinas/metabolismo , Biomarcadores/metabolismo , Muerte Celular/efectos de los fármacos , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Humanos , Receptores de Lipopolisacáridos/metabolismo , Macrólidos/farmacología , Monocitos/metabolismo
5.
Sci Rep ; 10(1): 22206, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33335142

RESUMEN

Collagen type VI (COL6) deposition occurs in various glomerular diseases, causing serious pathological damage like nodular lesions. However, the mechanisms underlying the deposition of COL6 remain unclear. In renal biopsy samples, immunohistochemical analyses revealed that COL6 and phosphorylated histone H2AX (γ-H2AX), a DNA damage marker, were detected mainly in diabetic nodular glomerulosclerosis, in which the γ-H2AX-positive area was identified as the independent factor significantly associated with the COL6-positive area (ß: 0.539, t = 2.668). In in vitro studies, COL6 secretion from human renal glomerular endothelial cells (HRGECs) was assessed by measuring the decrease in the cytoplasmic COL6-positive cells and an increase in the amount of COL6 in the culture medium. Mitomycin C (MMc) treatment of HRGECs increased the number of γ-H2AX-positive cells and COL6 secretion, which were suppressed by a specific inhibitor of ataxia telangiectasia and Rad3-related (ATR). MMc-induced COL6 secretion was also suppressed by Annexin A2 (ANXA2) siRNA transfection. Moreover, the inhibition of ATR activity did not induce any extra suppression in the MMc-induced COL6 secretion by ANXA2 siRNA transfected cells. These results confirm that nodular glomerulosclerosis partially results from DNA damage in the glomerulus and that DNA damage-induced COL6 secretion from HRGECs occurs through an ATR and ANXA2-mediated pathway.


Asunto(s)
Anexina A2/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Daño del ADN , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/metabolismo , Células Endoteliales/metabolismo , Glomérulos Renales/metabolismo , Glomérulos Renales/patología , Adulto , Anciano , Biomarcadores , Biopsia , Colágeno Tipo VI/metabolismo , Nefropatías Diabéticas/patología , Susceptibilidad a Enfermedades , Femenino , Técnica del Anticuerpo Fluorescente , Histonas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Unión Proteica
6.
Oncogenesis ; 9(6): 60, 2020 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-32541651

RESUMEN

The nucleus of mammalian cells is compartmentalized by nuclear bodies such as nuclear speckles, however, involvement of nuclear bodies, especially nuclear speckles, in DNA repair has not been actively investigated. Here, our focused screen for nuclear speckle factors involved in homologous recombination (HR), which is a faithful DNA double-strand break (DSB) repair mechanism, identified transcription-related nuclear speckle factors as potential HR regulators. Among the top hits, we provide evidence showing that USP42, which is a hitherto unidentified nuclear speckles protein, promotes HR by facilitating BRCA1 recruitment to DSB sites and DNA-end resection. We further showed that USP42 localization to nuclear speckles is required for efficient HR. Furthermore, we established that USP42 interacts with DHX9, which possesses DNA-RNA helicase activity, and is required for efficient resolution of DSB-induced R-loop. In conclusion, our data propose a model in which USP42 facilitates BRCA1 loading to DSB sites, resolution of DSB-induced R-loop and preferential DSB repair by HR, indicating the importance of nuclear speckle-mediated regulation of DSB repair.

7.
Appl Radiat Isot ; 154: 108890, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31525597

RESUMEN

DNA double-strand breaks (DSBs) of peripheral blood lymphocyte were prospectively assessed in 9 patients who were injected with 201Tl-chloride and 123I-beta-methyl-p-iodophenyl-pentadecanoic acid in dual-isotope imaging. Phosphorylated H2AX (γH2AX) was used as a biomarker for detecting DSBs, and the mean number of γH2AX foci per cell was measured microscopically. Mean γH2AX foci before administration of radiopharmaceuticals and at 3, 6, and 24 h following administration were 0.22 ±â€¯0.34, 0.10 ±â€¯0.14, 0.59 ±â€¯0.46, and 0.52 ±â€¯0.40, respectively (p = n.s. for all combinations).


Asunto(s)
Daño del ADN , Linfocitos/metabolismo , Linfocitos/efectos de la radiación , Imagen de Perfusión Miocárdica/efectos adversos , Tomografía Computarizada de Emisión de Fotón Único/efectos adversos , Anciano , Biomarcadores/sangre , Roturas del ADN de Doble Cadena , Ácidos Grasos , Femenino , Histonas/sangre , Humanos , Radioisótopos de Yodo/efectos adversos , Yodobencenos , Masculino , Persona de Mediana Edad , Imagen de Perfusión Miocárdica/métodos , Estudios Prospectivos , Radiofármacos/efectos adversos , Radioisótopos de Talio/efectos adversos , Tomografía Computarizada de Emisión de Fotón Único/métodos
8.
Exp Cell Res ; 362(2): 450-460, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29233683

RESUMEN

X-ray repair cross-complementing group 4 (XRCC4), a repair protein for DNA double-strand breaks, is cleaved by caspases during apoptosis. In this study, we examined the role of XRCC4 in apoptosis. Cell lines, derived from XRCC4-deficient M10 mouse lymphoma cells and stably expressing wild-type XRCC4 or caspase-resistant XRCC4, were established and treated with staurosporine (STS) to induce apoptosis. In STS-induced apoptosis, expression of wild-type, but not caspase-resistant, XRCC4 in XRCC4-deficient cells enhanced oligonucleosomal DNA fragmentation and the appearance of TUNEL-positive cells by promoting nuclear translocation of caspase-activated DNase (CAD), a major nuclease for oligonucleosomal DNA fragmentation. CAD activity is reportedly regulated by the ratio of two inhibitor of CAD (ICAD) splice variants, ICAD-L and ICAD-S mRNA, which, respectively, produce proteins with and without the ability to transport CAD into the nucleus. The XRCC4-dependent promotion of nuclear import of CAD in STS-treated cells was associated with reduction of ICAD-S mRNA and protein, and enhancement of phosphorylation and nuclear import of serine/arginine-rich splicing factor (SRSF) 1. These XRCC4-dependent, apoptosis-enhancing effects were canceled by depletion of SRSF1 or SR protein kinase (SRPK) 1. In addition, overexpression of SRSF1 in XRCC4-deficient cells restored the normal level of apoptosis, suggesting that SRSF1 functions downstream of XRCC4 in activating CAD. This XRCC4-dependent, SRPK1/SRSF1-mediated regulatory mechanism was conserved in apoptosis in Jurkat human leukemia cells triggered by STS, and by two widely used anti-cancer agents, Paclitaxel and Vincristine. These data imply that the level of XRCC4 expression could be used to predict the effects of apoptosis-inducing drugs in cancer treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas de Unión al ADN/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinasas/genética , Factores de Empalme Serina-Arginina/genética , Animales , Núcleo Celular/genética , Fragmentación del ADN/efectos de los fármacos , Reparación del ADN/genética , Desoxirribonucleasas/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Células Jurkat , Ratones , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Paclitaxel/farmacología , Transducción de Señal/efectos de los fármacos , Estaurosporina/farmacología , Vincristina/farmacología
9.
Sci Rep ; 7(1): 13808, 2017 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-29061988

RESUMEN

Accumulating evidence indicates that transcription is closely related to DNA damage formation and that the loss of RNA biogenesis factors causes genome instability. However, whether such factors are involved in DNA damage responses remains unclear. We focus here on the RNA helicase Aquarius (AQR), a known R-loop processing factor, and show that its depletion in human cells results in the accumulation of DNA damage during S phase, mediated by R-loop formation. We investigated the involvement of Aquarius in DNA damage responses and found that AQR knockdown decreased DNA damage-induced foci formation of Rad51 and replication protein A, suggesting that Aquarius contributes to homologous recombination (HR)-mediated repair of DNA double-strand breaks (DSBs). Interestingly, the protein level of CtIP, a DSB processing factor, was decreased in AQR-knockdown cells. Exogenous expression of Aquarius partially restored CtIP protein level; however, CtIP overproduction did not rescue defective HR in AQR-knockdown cells. In accordance with these data, Aquarius depletion sensitized cells to genotoxic agents. We propose that Aquarius contributes to the maintenance of genomic stability via regulation of HR by CtIP-dependent and -independent pathways.


Asunto(s)
Proteínas Portadoras/metabolismo , Roturas del ADN de Doble Cadena , Inestabilidad Genómica , Neoplasias/genética , Proteínas Nucleares/metabolismo , ARN Helicasas/metabolismo , Reparación del ADN por Recombinación , Proteínas Portadoras/genética , Endodesoxirribonucleasas , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/genética , ARN Helicasas/antagonistas & inhibidores , ARN Helicasas/genética , Células Tumorales Cultivadas
10.
Int J Biol Sci ; 12(2): 172-83, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26884715

RESUMEN

UNLABELLED: The ubiquitously expressed ß2-spectrin (ß2SP, SPTBN1) is the most common non-erythrocytic member of the ß-spectrin gene family. Loss of ß2-spectrin leads to defects in liver development, and its haploinsufficiency spontaneously leads to chronic liver disease and the eventual development of hepatocellular cancer. However, the specific role of ß2-spectrin in liver homeostasis remains to be elucidated. Here, we reported that ß2-spectrin was cleaved by caspase-3/7 upon treatment with acetaminophen which is the main cause of acute liver injury. Blockage of ß2-spectrin cleavage robustly attenuated ß2-spectrin-specific functions, including regulation of the cell cycle, apoptosis, and transcription. Cleaved fragments of ß2-spectrin were physiologically active, and the N- and C-terminal fragments retained discrete interaction partners and activity in transcriptional regulation and apoptosis, respectively. Cleavage of ß2-spectrin facilitated the redistribution of the resulting fragments under conditions of liver damage induced by acetaminophen. In contrast, downregulation of ß2-spectrin led to resistance to acetaminophen-induced cytotoxicity, and its insufficiency in the liver promoted suppression of acetaminophen-induced liver damage and enhancement of liver regeneration. CONCLUSIONS: ß2-Spectrin, a TGF-ß mediator and signaling molecule, is cleaved and activated by caspase-3/7, consequently enhancing apoptosis and transcriptional control to determine cell fate upon liver damage. These findings have extended our knowledge on the spectrum of ß2-spectrin functions from a scaffolding protein to a target and transmitter of TGF-ß in liver damage.


Asunto(s)
Acetaminofén/toxicidad , Caspasa 3/fisiología , Caspasa 7/fisiología , Enfermedad Hepática Inducida por Sustancias y Drogas , Espectrina/fisiología , Animales , Células COS , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular , Chlorocebus aethiops , Regulación hacia Abajo , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Hígado/efectos de los fármacos , Hígado/patología , Hígado/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/metabolismo , Transducción de Señal , Espectrina/genética , Espectrina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
11.
Clin Exp Nephrol ; 20(3): 479-88, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26440363

RESUMEN

BACKGROUNDS: The relationship between DNA damage and glomerular fibrosis in renal allografts remains unclear. METHODS: We examined renal allograft specimens from 35 patients in which DNA double-strand breaks (DSBs) and glomerular fibrosis were detected by phospho-histone H2A.X (γ-H2AX) expression and collagen (COL) types III, IV, and VI accumulation. We also examined the in vitro relationship between DNA damage and COL accumulation by mitomycin C (MMc)-induced DNA damage in human glomerular endothelial cells (HRGEc). RESULTS: The γ-H2AX and COL type VI, which mainly accumulated in the subendothelial and mesangial regions, were positively correlated with the duration of the post-renal transplant (RT) period. In multiple regression analysis, the duration of the post-RT period and cg in the Banff '07 classification were identified as a significant predictor of COL type VI accumulation and γ-H2AX expression in the glomerular capillaries. In addition, the γ-H2AX-positive area was also identified as a predictor of glomerular accumulation of COL type VI. COL type VI was detected in the cytoplasm of the HRGEc, which was secreted into the supernatant after MMc stimulation with γ-H2AX expression. The number of γ-H2AX (-)/COL type VI (+) cells was inversely associated with the number of γ-H2AX (+)/COL type VI (-) cells during 24-h MMc treatment. CONCLUSIONS: Our findings suggest that the long-term RT induces DSBs and HRGEc-secreted COL type VI accumulation in the glomerular capillaries, which might progress to intractable glomerular fibrosis.


Asunto(s)
Roturas del ADN de Doble Cadena , Enfermedades Renales/genética , Glomérulos Renales/metabolismo , Trasplante de Riñón/efectos adversos , Adulto , Anciano , Aloinjertos , Capilares/efectos de los fármacos , Capilares/metabolismo , Capilares/patología , Células Cultivadas , Colágeno/metabolismo , Femenino , Fibrosis , Histonas/metabolismo , Humanos , Inmunohistoquímica , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Masculino , Persona de Mediana Edad , Mitomicina/toxicidad , Factores de Tiempo , Resultado del Tratamiento
12.
Genes Genet Syst ; 90(4): 187-94, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26616758

RESUMEN

Camptothecin (CPT) inhibits DNA topoisomerase I (Top1) through a non-catalytic mechanism that stabilizes the Top1-DNA cleavage complex (Top1cc) and blocks the DNA re-ligation step, resulting in the accumulation in the genome of DNA single-strand breaks (SSBs), which are converted to secondary strand breaks when they collide with the DNA replication and RNA transcription machinery. DNA strand breaks mediated by replication, which have one DNA end, are distinct in repair from the DNA double-strand breaks (DSBs) that have two ends and are caused by ionizing radiation and other agents. In contrast to two-ended DSBs, such one-ended DSBs are preferentially repaired through the homologous recombination pathway. Conversely, the repair of one-ended DSBs by the non-homologous end-joining pathway is harmful for cells and leads to cell death. The choice of repair pathway has a crucial impact on cell fate and influences the efficacy of anticancer drugs such as CPT derivatives. In addition to replication-mediated one-ended DSBs, transcription also generates DNA strand breaks upon collision with the Top1cc. Some reports suggest that transcription-mediated DNA strand breaks correlate with neurodegenerative diseases. However, the details of the repair mechanisms of, and cellular responses to, transcription-mediated DNA strand breaks still remain unclear. In this review, combining our recent results and those of previous reports, we introduce and discuss the responses to CPT-induced DNA damage mediated by DNA replication and RNA transcription.


Asunto(s)
Camptotecina/farmacología , Roturas del ADN de Cadena Simple , Reparación del ADN , Replicación del ADN/efectos de los fármacos , ADN-Topoisomerasas de Tipo I/metabolismo , Humanos , Transcripción Genética/efectos de los fármacos
13.
PLoS Pathog ; 10(3): e1004012, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24651521

RESUMEN

RIG-I is a DExD/H-box RNA helicase and functions as a critical cytoplasmic sensor for RNA viruses to initiate antiviral interferon (IFN) responses. Here we demonstrate that another DExD/H-box RNA helicase DHX36 is a key molecule for RIG-I signaling by regulating double-stranded RNA (dsRNA)-dependent protein kinase (PKR) activation, which has been shown to be essential for the formation of antiviral stress granule (avSG). We found that DHX36 and PKR form a complex in a dsRNA-dependent manner. By forming this complex, DHX36 facilitates dsRNA binding and phosphorylation of PKR through its ATPase/helicase activity. Using DHX36 KO-inducible MEF cells, we demonstrated that DHX36 deficient cells showed defect in IFN production and higher susceptibility in RNA virus infection, indicating the physiological importance of this complex in host defense. In summary, we identify a novel function of DHX36 as a critical regulator of PKR-dependent avSG to facilitate viral RNA recognition by RIG-I-like receptor (RLR).


Asunto(s)
ARN Helicasas DEAD-box/inmunología , Infecciones por Virus ARN/inmunología , Transducción de Señal/inmunología , eIF-2 Quinasa/inmunología , Gránulos Citoplasmáticos/inmunología , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/metabolismo , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Técnicas de Inactivación de Genes , Células HEK293 , Células HeLa , Humanos , Inmunoprecipitación , Virus ARN/inmunología , ARN Bicatenario/inmunología , ARN Interferente Pequeño/genética , ARN Viral/inmunología , Receptores Inmunológicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Fisiológico , Transfección
14.
Histochem Cell Biol ; 141(1): 101-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23949737

RESUMEN

RBM8A (Y14) is carrying RNA-binding motif and forms the tight heterodimer with MAGOH. The heterodimer is known to be a member of exon junction complex on exporting mRNA and is required for mRNA metabolisms such as splicing, mRNA export and nonsense-mediated mRNA decay. Almost all RBM8A-MAGOH complexes localize in nucleoplasm and shuttle between nuclei and cytoplasm for RNA metabolism. Recently, the abnormality of G2/M transition and aberrant centrosome regulation in RBM8A- or MAGOH-deficient cells has been reported. These results prompt us to the reevaluation of the localization of RBM8A-MAGOH in human cells. Interestingly, our immunostaining experiments showed the localization of these proteins in centrosome in addition to nuclei. Furthermore, the transiently expressed eYFP-tagged RBM8A and Flag-tagged MAGOH also co-localized with centrosome signals. In addition, the proximity ligation in situ assay was performed to detect the complex formation in centrosome. Our experiments clearly showed that Myc-tagged RBM8A and Flag-tagged MAGOH formed a complex in centrosome. GFP-tagged PLK1 also co-localized with Myc-RBM8A. Our results show that RBM8A-MAGOH complex is required for M-phase progression via direct localization to centrosome rather than indirect effect.


Asunto(s)
Centrosoma/metabolismo , Proteínas Nucleares/farmacocinética , Proteínas de Unión al ARN/farmacocinética , Transporte Activo de Núcleo Celular/genética , División Celular/genética , Línea Celular , Núcleo Celular/genética , Proteínas Fluorescentes Verdes/genética , Humanos , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , ARN Mensajero/genética , Proteínas de Unión al ARN/biosíntesis , Proteínas de Unión al ARN/genética
15.
Cancer Lett ; 345(1): 115-23, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24333722

RESUMEN

The ataxia telangiectasia mutated (ATM) signaling pathway responds rapidly to DNA double-strand breaks (DSBs) and it is characterized by recruitment of sensor, mediator, transducer and repair proteins to sites of DNA damage. Data suggest that CK2 is implicated in the early cellular response to DSBs. We demonstrate that CK2 binds constitutively the adaptor protein 53BP1 through the tandem Tudor domains and that the interaction is disrupted upon induction of DNA damage. Down-regulation of CK2 results in significant reduction of (i) 53BP1 foci formation, (ii) binding to dimethylated histone H4 and (iii) ATM autophosphorylation. Our data suggest that CK2 is required for 53BP1 accumulation at sites of DSBs which is a prerequisite for efficient activation of the ATM-mediated signaling pathway.


Asunto(s)
Antineoplásicos/farmacología , Quinasa de la Caseína II/metabolismo , Roturas del ADN de Doble Cadena , Glioblastoma/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Quinasa de la Caseína II/deficiencia , Quinasa de la Caseína II/genética , Línea Celular Tumoral , Reparación del ADN por Unión de Extremidades , ADN de Neoplasias/efectos de los fármacos , ADN de Neoplasias/genética , Glioblastoma/tratamiento farmacológico , Glioblastoma/enzimología , Glioblastoma/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Fosforilación , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Dosis de Radiación , Efectos de la Radiación , Transducción de Señal , Transfección , Proteína 1 de Unión al Supresor Tumoral P53
16.
PLoS One ; 8(8): e72253, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23967291

RESUMEN

Nonhomologous end-joining (NHEJ) and homologous recombination (HR) are two major pathways for repairing DNA double-strand breaks (DSBs); however, their respective roles in human somatic cells remain to be elucidated. Here we show using a series of human gene-knockout cell lines that NHEJ repairs nearly all of the topoisomerase II- and low-dose radiation-induced DNA damage, while it negatively affects survival of cells harbouring replication-associated DSBs. Intriguingly, we find that loss of DNA ligase IV, a critical NHEJ ligase, and Artemis, an NHEJ factor with endonuclease activity, independently contribute to increased resistance to replication-associated DSBs. We also show that loss of Artemis alleviates hypersensitivity of DNA ligase IV-null cells to low-dose radiation- and topoisomerase II-induced DSBs. Finally, we demonstrate that Artemis-null human cells display increased gene-targeting efficiencies, particularly in the absence of DNA ligase IV. Collectively, these data suggest that DNA ligase IV and Artemis act cooperatively to promote NHEJ, thereby suppressing HR. Our results point to the possibility that HR can only operate on accidental DSBs when NHEJ is missing or abortive, and Artemis may be involved in pathway switching from incomplete NHEJ to HR.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , ADN Ligasas/metabolismo , Recombinación Homóloga , Proteínas Nucleares/metabolismo , Línea Celular , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Reparación del ADN por Unión de Extremidades/efectos de la radiación , ADN Ligasa (ATP) , ADN Ligasas/deficiencia , Replicación del ADN/efectos de los fármacos , Replicación del ADN/efectos de la radiación , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN , Endonucleasas , Técnicas de Inactivación de Genes , Marcación de Gen , Recombinación Homóloga/efectos de los fármacos , Recombinación Homóloga/efectos de la radiación , Humanos , Mutación , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Inhibidores de Topoisomerasa II/farmacología
17.
Exp Biol Med (Maywood) ; 238(8): 889-97, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23970407

RESUMEN

RBM8A (Y14) contains an RNA-binding motif and forms a tight heterodimer with Magoh. The heterodimer is known to be a member of the exon junction complex that forms on mRNA before export and it is required for mRNA metabolism processes such as splicing, mRNA export and nonsense-mediated mRNA decay. Recently, deficient cellular proliferation has been observed in RBM8A- or Magoh-depleted cells. These results prompted us to study the role of RBM8A in cell cycle progression of human tumour cells. The depletion of RBM8A in A549 cells resulted in poor cell survival and the accumulation of mitotic cells. After release from G1/S arrest induced by a double thymidine block, the RBM8A-silenced cells could not proceed to the next G1 phase beyond G2/M phase. Finally, the sub-G1 population increased and the apoptosis markers caspases 3/7 were activated. Silenced cells exhibited an increased frequency of multipolar or monopolar centrosomes, which may have caused the observed deficiency in cell cycle progression. Finally, silencing of either RBM8A or Magoh resulted in mutual downregulation of the other protein. These results illustrate that the RBM8A-Magoh mRNA binding complex is required for M phase progression and both proteins may be novel targets for anticancer therapy.


Asunto(s)
Adenocarcinoma/patología , Apoptosis/fisiología , Ciclo Celular/fisiología , Células HeLa/patología , Neoplasias Pulmonares/patología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/fisiología , Apoptosis/genética , Caspasas/genética , Caspasas/fisiología , Ciclo Celular/genética , División Celular/genética , División Celular/fisiología , Línea Celular Tumoral , Células Cultivadas , Centrosoma/fisiología , Regulación hacia Abajo/efectos de los fármacos , Femenino , Fase G1/genética , Fase G1/fisiología , Silenciador del Gen/efectos de los fármacos , Humanos , Proteínas Nucleares/efectos de los fármacos , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , ARN Interferente Pequeño/farmacología , Proteínas de Unión al ARN/efectos de los fármacos
18.
Ann Nucl Med ; 27(3): 233-8, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23264066

RESUMEN

PURPOSE: The purpose of our study was to evaluate the degree of radiotoxicity to lymphocytes in thyroid cancer after iodine-131(I-131) therapy using γ-H2AX foci immunodetection. METHODS: This study focused on 15 patients who underwent I-131 therapy for differentiated thyroid cancer after surgery. All patients received 3.7 GBq of I-131. Venous blood samples were collected from each patient before therapy and 4 days thereafter. Lymphocytes were isolated from the blood samples and subjected to γ-H2AX immunofluorescence staining. RESULTS: The number (mean ± SD) of foci per lymphocyte nucleus was 0.41 ± 0.51 before and 6.19 ± 1.80 after radioiodine therapy, and this difference was statistically significant (P = 0.001 < 0.05). Absorbed doses estimated for the 15 patients were 0.77 ± 0.31 Gy applying standard line in vitro external radiation doses. CONCLUSION: γ-H2AX foci immunodetection in lymphocytes may detect radiation-induced DNA damage associated with I-131 therapy for thyroid cancer, and may facilitate estimation of the radiation doses absorbed with this therapy.


Asunto(s)
Daño del ADN , Histonas/genética , Linfocitos/metabolismo , Linfocitos/efectos de la radiación , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/radioterapia , Adulto , Femenino , Humanos , Radioisótopos de Yodo/efectos adversos , Radioisótopos de Yodo/uso terapéutico , Masculino , Persona de Mediana Edad , Dosis de Radiación
19.
Biochem Pharmacol ; 84(12): 1592-603, 2012 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-23022228

RESUMEN

Store-operated calcium (Ca(2+)) entry (SOCE) is important for cellular activities such as gene transcription, cell cycle progression and proliferation in most non-excitable cells. Stromal interaction molecule 1 (STIM1), a newly identified Ca(2+)-sensing protein, monitors the depletion of endoplasmic reticulum (ER) Ca(2+) stores and activates store-operated Ca(2+) channels at the plasma membrane to induce SOCE. To investigate the possible roles of STIM1 in tumor growth in relation to SOCE, we established STIM1 knockdown (KD) clones of human epidermoid carcinoma A431 cells by RNA interference. Thapsigargin, an inhibitor of ER Ca(2+)-ATPase, -induced and phospholipase C-coupled receptor agonist-induced SOCEs were reduced in two STIM1 KD clones compared to a negative control clone. Re-expression of a KD-resistant full-length STIM1, but not a Ca(2+) release-activated Ca(2+) channel activation domain (CAD)-deleted STIM1 mutant, in the KD clone restored the amplitude of SOCE, suggesting the specificity of the STIM1 knockdown. The cell growth of the STIM1 KD clones was slower than that of the negative control clone. DNA synthesis assessed by BrdU incorporation, as well as EGF-stimulated EGF receptor activation, decreased in the STIM1 KD clones. Xenograft growth of the STIM1 KD clones was significantly retarded compared with that of the negative control. Cell migration was attenuated in the STIM1 KD clone and the STIM1 silencing effect was reversed by transient re-expression of the full-length STIM1 but not CAD-deletion mutant. These results indicate that STIM1 plays an important role in SOCE, cell-growth and tumorigenicity in human epidermoid carcinoma A431cells, suggesting the potential use of STIM1-targeting agents for treating epidermoid carcinoma.


Asunto(s)
Calcio/metabolismo , Carcinoma de Células Escamosas/patología , Proliferación Celular , Transformación Celular Neoplásica , Técnicas de Silenciamiento del Gen , Proteínas de la Membrana/fisiología , Proteínas de Neoplasias/fisiología , Secuencia de Bases , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Cartilla de ADN , Humanos , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Molécula de Interacción Estromal 1
20.
Nucleic Acids Res ; 37(7): 2176-93, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19228710

RESUMEN

Recruitment of RAD18 to stalled replication forks facilitates monoubiquitination of PCNA during S-phase, promoting translesion synthesis at sites of UV irradiation-induced DNA damage. In this study, we show that RAD18 is also recruited to ionizing radiation (IR)-induced sites of DNA double-strand breaks (DSBs) forming foci which are co-localized with 53BP1, NBS1, phosphorylated ATM, BRCA1 and gamma-H2AX. RAD18 associates with 53BP1 and is recruited to DSB sites in a 53BP1-dependent manner specifically during G1-phase, RAD18 monoubiquitinates KBD domain of 53BP1 at lysine 1268 in vitro. A monoubiquitination-resistant 53BP1 mutant harboring a substitution at lysine 1268 is not retained efficiently at the chromatin in the vicinity of DSBs. In Rad18-null cells, retention of 53BP1 foci, efficiency of DSB repair and post-irradiation viability are impaired compared with wild-type cells. Taken together, these results suggest that RAD18 promotes 53BP1-directed DSB repair by enhancing retention of 53BP1, possibly through an interaction between RAD18 and 53BP1 and the modification of 53BP1.


Asunto(s)
Cromatina/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Fase G1/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Animales , Línea Celular , Núcleo Celular/metabolismo , Núcleo Celular/efectos de la radiación , Proteínas Cromosómicas no Histona , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Epigénesis Genética , Fase G1/efectos de la radiación , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Proteína 1 de Unión al Supresor Tumoral P53 , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas , Ubiquitinación , Dedos de Zinc
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...