Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Oncol Lett ; 25(5): 207, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37123020

RESUMEN

Cancer stem cells (CSCs) are known to be a major cause of metastasis, resistance and recurrence. Spheroid formation is one of the methods used to recruit CSCs utilizing an anchorage-independent environment in vitro. It was aimed to investigate the availability of spheroid formation culture methods in the research field of CSCs and resistance using 5-fluorouracil (5-FU)-resistant colorectal cancer cells. The wild type SNU-C5 and 5-FU-resistant SNU-C5 (SNU-C5/5-FUR) cells were cultured as usual (monolayer), and in 3-dimensional non-adhesive environments supplemented with fetal bovine serum (FBS) or growth factors, respectively. The characteristics of the spheroids were evaluated by morphometry, cell viability assay, western blotting, immunocytochemistry and enzyme-linked immunosorbent assay. Spheroid formation was induced in an environment supplemented with FBS, while SNU-C5/5-FUR cells only formed spheres in media supplemented with GFs. Sphere-formed cells showed slower cell proliferation than cells from monolayer, which coincided with an increased level of p21 and a decreased level of ß-catenin. Markers for CSCs and drug resistance were not significantly changed after spheroid formation. Sphere-formed cells showed significantly increased levels of soluble E-cadherin, particularly in the environment supplemented with FBS. These results suggested that spheroid formation may be related to soluble E-cadherin, but is not related to CSCs or resistance markers.

2.
Cell Death Dis ; 13(6): 528, 2022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35668076

RESUMEN

The cell signaling factors EGFR, EphA2, and Ephexin1 are associated with lung and colorectal cancer and play an important role in tumorigenesis. Although the respective functional roles of EGFR and EphA2 are well known, interactions between these proteins and a functional role for the complex is not understood. Here, we showed that Ephexin1, EphA2, and EGFR are each expressed at higher levels in lung and colorectal cancer patient tissues, and binding of EGFR to EphA2 was associated with both increased tumor grade and metastatic cases in both cancer types. Treatment with Epidermal Growth Factor (EGF) induced binding of the RR domain of EGFR to the kinase domain of EphA2, and this binding was promoted by Ephexin1. Additionally, the AKT-mediated phosphorylation of EphA2 (at Ser897) promoted interactions with EGFR, pointing to the importance of this pathway. Two mutations in EGFR, L858R and T790M, that are frequently observed in lung cancer patients, promoted binding to EphA2, and this binding was dependent on Ephexin1. Our results indicate that the formation of a complex between EGFR, EphA2, and Ephexin1 plays an important role in lung and colorectal cancers, and that inhibition of this complex may be an effective target for cancer therapy.


Asunto(s)
Neoplasias Colorrectales , Neoplasias Pulmonares , Receptor EphA2 , Carcinogénesis/genética , Línea Celular Tumoral , Transformación Celular Neoplásica , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Receptores ErbB/metabolismo , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Receptor EphA2/genética , Receptor EphA2/metabolismo
3.
Cell Death Dis ; 13(4): 309, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35387978

RESUMEN

The Hsp70-binding protein 1 (HspBP1) belongs to a family of co-chaperones that regulate Hsp70 activity and whose biological significance is not well understood. In the present study, we show that when HspBP1 is either knocked down or overexpressed in BRCA1-proficient breast cancer cells, there were profound changes in tumorigenesis, including anchorage-independent cell growth in vitro and in tumor formation in xenograft models. However, HspBP1 did not affect tumorigenic properties in BRCA1-deficient breast cancer cells. The mechanisms underlying HspBP1-induced tumor suppression were found to include interactions with BRCA1 and promotion of BRCA1-mediated homologous recombination DNA repair, suggesting that HspBP1 contributes to the suppression of breast cancer by regulating BRCA1 function and thereby maintaining genomic stability. Interestingly, independent of BRCA1 status, HspBP1 facilitates cell survival in response to ionizing radiation (IR) by interfering with the association of Hsp70 and apoptotic protease-activating factor-1. These findings suggest that decreased HspBP1 expression, a common occurrence in high-grade and metastatic breast cancers, leads to genomic instability and enables resistance to IR treatment.


Asunto(s)
Neoplasias de la Mama , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Apoptosis/genética , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Reparación del ADN , Femenino , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Reparación del ADN por Recombinación
4.
Nucleic Acids Res ; 50(3): 1501-1516, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35061896

RESUMEN

Homologous recombination (HR) is critical for error-free repair of DNA double-strand breaks. Chromatin loading of RAD51, a key protein that mediates the recombination, is a crucial step in the execution of the HR repair. Here, we present evidence that SUMOylation of RAD51 is crucial for the RAD51 recruitment to chromatin and HR repair. We found that topoisomerase 1-binding arginine/serine-rich protein (TOPORS) induces the SUMOylation of RAD51 at lysine residues 57 and 70 in response to DNA damaging agents. The SUMOylation was facilitated by an ATM-induced phosphorylation of TOPORS at threonine 515 upon DNA damage. Knockdown of TOPORS or expression of SUMOylation-deficient RAD51 mutants caused reduction in supporting normal RAD51 functions during the HR repair, suggesting the physiological importance of the modification. We found that the SUMOylation-deficient RAD51 reduces the association with its crucial binding partner BRCA2, explaining its deficiency in supporting the HR repair. These findings altogether demonstrate a crucial role for TOPORS-mediated RAD51 SUMOylation in promoting HR repair and genomic maintenance.


Asunto(s)
Recombinasa Rad51 , Reparación del ADN por Recombinación , Cromatina , ADN/metabolismo , Daño del ADN , Reparación del ADN/genética , Recombinación Homóloga , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Sumoilación
5.
Cell Death Dis ; 12(11): 1013, 2021 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-34711817

RESUMEN

ABSTRCT: Ephexin1 was reported to be highly upregulated by oncogenic Ras, but the functional consequences of this remain poorly understood. Here, we show that Ephexin1 is highly expressed in colorectal cancer (CRC) and lung cancer (LC) patient tissues. Knockdown of Ephexin1 markedly inhibited the cell growth of CRC and LC cells with oncogenic Ras mutations. Ephexin1 contributes to the positive regulation of Ras-mediated downstream target genes and promotes Ras-induced skin tumorigenesis. Mechanically, Akt phosphorylates Ephexin1 at Ser16 and Ser18 (pSer16/18) and pSer16/18 Ephexin1 then interacts with oncogenic K-Ras to promote downstream MAPK signaling, facilitating tumorigenesis. Furthermore, pSer16/18 Ephexin1 is associated with both an increased tumor grade and metastatic cases of CRC and LC, and those that highly express pSer16/18 exhibit poor overall survival rates. These data indicate that Ephexin1 plays a critical role in the Ras-mediated CRC and LC and pSer16/18 Ephexin1 might be an effective therapeutic target for CRC and LC.


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Oncogenes , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas ras/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/química , Factores de Intercambio de Guanina Nucleótido/genética , Células HEK293 , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Modelos Biológicos , Fosforilación , Fosfoserina/metabolismo , Pronóstico , Unión Proteica , Dominios Proteicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba
6.
Korean J Physiol Pharmacol ; 24(3): 267-276, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32392918

RESUMEN

In the present study, we investigated the effect of oncogenic H-Ras on rat mdr1b expression in NIH3T3 cells. The constitutive expression of H-RasV12 was found to downregulate the mdr1b promoter activity and mdr1b mRNA expression. The doxorubicin-induced mdr1b promoter activity of the H-RasV12 expressing NIH3T3 cells was markedly lower than that of control NIH3T3 cells. Additionally, there is a positive correlation between the level of H-RasV12 expression and a sensitivity to doxorubicin toxicity. To examine the detailed mechanism of H-RasV12-mediated down-regulation of mdr1b expression, antioxidant N-acetylcysteine (NAC) and NADPH oxidase inhibitor diphenylene iodonium (DPI) were used. Pretreating cells with either NAC or DPI significantly enhanced the oncogenic H-Ras-mediated down-regulation of mdr1b expression and markedly prevented doxorubicin-induced cell death. Moreover, NAC and DPI treatment led to a decrease in ERK activity, and the ERK inhibitors PD98059 or U0126 enhanced the mdr1b-Luc activity of H-RasV12-NIH3T3 and reduced doxorubicin-induced apoptosis. These data suggest that RasV12 expression could downregulate mdr1b expression through intracellular reactive oxygen species (ROS) production, and ERK activation induced by ROS, is at least in part, contributed to the downregulation of mdr1b expression.

7.
Int J Mol Sci ; 21(7)2020 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-32290222

RESUMEN

Mediator of DNA damage checkpoint protein 1 (MDC1) plays a vital role in DNA damage response (DDR) by coordinating the repair of double strand breaks (DSBs). Here, we identified a novel interaction between MDC1 and karyopherin α-2 (KPNA2), a nucleocytoplasmic transport adaptor, and showed that KPNA2 is necessary for MDC1 nuclear import. Thereafter, we identified a functional nuclear localization signal (NLS) between amino acid residues 1989-1994 of the two Breast Cancer 1 (BRCA1) carboxyl-terminal (tBRCT) domain of MDC1 and demonstrated disruption of this NLS impaired interaction between MDC1 and KPNA2 and reduced nuclear localization of MDC1. In KPNA2-depleted cells, the recruitment of MDC1, along with the downstream signaling p roteins Ring Finger Protein 8 (RNF8), 53BP1-binding protein 1 (53BP1), BRCA1, and Ring Finger Protein 168 (RNF168), to DNA damage sites was abolished. Additionally, KPNA2-depleted cells had a decreased rate of homologous recombination (HR) repair. Our data suggest that KPNA2-mediated MDC1 nuclear import is important for DDR signaling and DSB repair.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Señales de Localización Nuclear , Dominios y Motivos de Interacción de Proteínas , alfa Carioferinas/metabolismo , Transporte Activo de Núcleo Celular , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas de Ciclo Celular/química , Línea Celular Tumoral , Daño del ADN , Técnicas de Silenciamiento del Gen , Humanos , Unión Proteica , Reparación del ADN por Recombinación , alfa Carioferinas/genética
8.
Nat Commun ; 8(1): 903, 2017 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-29026069

RESUMEN

MDC1 plays a critical role in the DNA damage response (DDR) by interacting directly with several factors including γ-H2AX. However, the mechanism by which MDC1 is recruited to damaged sites remains elusive. Here, we show that MDC1 interacts with a helix-loop-helix (HLH)-containing protein called inhibitor of DNA-binding 3 (ID3). In response to double-strand breaks (DSBs) in the genome, ATM phosphorylates ID3 at serine 65 within the HLH motif, and this modification allows a direct interaction with MDC1. Moreover, depletion of ID3 results in impaired formation of ionizing radiation (IR)-induced MDC1 foci, suppression of γ-H2AX-bound MDC1, impaired DSB repair, cellular hypersensitivity to IR, and genomic instability. Disruption of the MDC1-ID3 interaction prevents accumulation of MDC1 at sites of DSBs and suppresses DSB repair. Thus, our study uncovers an ID3-dependent mechanism of recruitment of MDC1 to DNA damage sites and suggests that the ID3-MDC1 interaction is crucial for DDR.MDC1 is a key component of the DNA damage response and interacts with several factors such as γ-H2AX. Here the authors show that MDC1 interacts with ID3, facilitating MDC1 recruitment to sites of damage and repair of breaks.


Asunto(s)
Daño del ADN , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Bovinos , Proteínas de Ciclo Celular , Roturas del ADN de Doble Cadena , Inestabilidad Genómica , Células HEK293 , Células HeLa , Secuencias Hélice-Asa-Hélice , Histonas/metabolismo , Humanos , Proteínas Inhibidoras de la Diferenciación , Ratones , Proteínas de Neoplasias , Radiación Ionizante , Ratas
9.
Cancer Res ; 75(7): 1298-310, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25627978

RESUMEN

MDC1 is critical component of the DNA damage response (DDR) machinery and orchestrates the ensuring assembly of the DDR protein at the DNA damage sites, and therefore loss of MDC1 results in genomic instability and tumorigenicity. However, the molecular mechanisms controlling MDC1 expression are currently unknown. Here, we show that miR-22 inhibits MDC1 translation via direct binding to its 3' untranslated region, leading to impaired DNA damage repair and genomic instability. We demonstrated that activated Akt1 and senescence hinder DDR function of MDC1 by upregulating endogenous miR-22. After overexpression of constitutively active Akt1, homologous recombination was inhibited by miR-22-mediated MDC1 repression. In addition, during replicative senescence and stress-induced premature senescence, MDC1 was downregulated by upregulating miR-22 and thereby accumulating DNA damage. Our results demonstrate a central role of miR-22 in the physiologic regulation of MDC1-dependent DDR and suggest a molecular mechanism for how aberrant Akt1 activation and senescence lead to increased genomic instability, fostering an environment that promotes tumorigenesis.


Asunto(s)
Reparación del ADN , Inestabilidad Genómica , MicroARNs/fisiología , Proteínas Nucleares/genética , Transactivadores/genética , Proteínas Adaptadoras Transductoras de Señales , Adolescente , Anciano , Animales , Proteínas de Ciclo Celular , Senescencia Celular , Daño del ADN , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HEK293 , Humanos , Ratones , Persona de Mediana Edad , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , Transactivadores/metabolismo , Adulto Joven
10.
Redox Rep ; 18(5): 165-73, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23883737

RESUMEN

Mutations in mismatch repair (MMR) genes are commonly associated with the development of colorectal cancer. Additionally, base excision repair, which involves apurinic/apyrimidinic endonuclease 1 (APE1), recognizes and eliminates oxidative DNA damage. Here, we investigated the possible roles of APE1 in dextran sulfate sodium (DSS)-induced acute colitis using the young rat model. Four-week-old Sprague-Dawley rats were administered 2% DSS in drinking water for 1 week. MMR and APE1 expression levels were assessed by western blotting and immunohistochemistry. Following DSS treatment, growth of young rats failed and the animals had loose stools. Together with the histological changes associated with acute colitis, APE1 and MSH2 levels increased significantly at 3 and 5 days after DSS treatment, respectively. The difference between APE1 and MSH2 expression was significant. DSS-induced DNA damage and subsequent repair activity were evaluated by staining for 8-hydroxy-deoxyguanosine (8-OHdG) and APE1, respectively; 8-OHdG immunoreactivity increased throughout the colonic mucosa, while APE1 levels in the surface epithelium increased at an earlier timepoint. Taken together, our data suggest that changes in APE1 expression after DSS treatment occurred earlier and were more widespread than changes in MMR expression, suggesting that APE1 is more sensitive for prediction of DNA deterioration in DSS-induced colitis.


Asunto(s)
Colitis/inducido químicamente , Colitis/metabolismo , Sulfato de Dextran/toxicidad , Animales , Colitis/genética , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa , Ratas , Ratas Sprague-Dawley
11.
J Clin Invest ; 2013 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-23863623

RESUMEN

Aberrant expression of apurinic-apyrimidinic endonuclease-1 (APEX1) has been reported in numerous human solid tumors and is positively correlated with cancer progression; however, the role of APEX1 in tumor progression is poorly defined. Here, we show that APEX1 contributes to aggressive colon cancer behavior and functions as an upstream activator in the Jagged1/Notch signaling pathway. APEX1 overexpression or knockdown in human colon cancer cell lines induced profound changes in malignant properties such as cell proliferation, anchorage-independent growth, migration, invasion, and angiogenesis in vitro and in tumor formation and metastasis in mouse xenograft models. These oncogenic effects of APEX1 were mediated by the upregulation of Jagged1, a major Notch ligand. Furthermore, APEX1 expression was associated with Jagged1 in various colon cancer cell lines and in tissues from colon cancer patients. This finding identifies APEX1 as a positive regulator of Jagged1/Notch activity and suggests that it is a potential therapeutic target in colon cancers that exhibit high levels of Jagged1/Notch signaling.

12.
Biochim Biophys Acta ; 1823(12): 2099-108, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22982065

RESUMEN

The catalytic subunit of DNA-dependent protein kinase (DNA-PKcs) plays an essential role in double-strand break repair by initially recognizing and binding to DNA breaks. Here, we show that DNA-PKcs interacts with the regulatory γ1 subunit of AMP-activated protein kinase (AMPK), a heterotrimeric enzyme that has been proposed to function as a "fuel gauge" to monitor changes in the energy status of cells and is controlled by the upstream kinases LKB1 and Ca²âº/calmodulin-dependent kinase kinase (CaMKK). In co-immunoprecipitation analyses, DNA-PKcs and AMPKγ1 interacted physically in DNA-PKcs-proficient M059K cells but not in DNA-PKcs-deficient M059J cells. Glucose deprivation-stimulated phosphorylation of AMPKα on Thr172 and of acetyl-CoA carboxylase (ACC), a downstream target of AMPK, is substantially reduced in M059J cells compared with M059K cells. The inhibition or down-regulation of DNA-PKcs by the DNA-PKcs inhibitors, wortmannin and Nu7441, or by DNA-PKcs siRNA caused a marked reduction in AMPK phosphorylation, AMPK activity, and ACC phosphorylation in response to glucose depletion in M059K, WI38, and IMR90 cells. In addition, DNA-DNA-PKcs(-/-) mouse embryonic fibroblasts (MEFs) exhibited decreased AMPK activation in response to glucose-free conditions. Furthermore, the knockdown of DNA-PKcs led to the suppression of AMPK (Thr172) phosphorylation in LKB1-deficient HeLa cells under glucose deprivation. Taken together, these findings support the positive regulation of AMPK activation by DNA-PKcs under glucose-deprived conditions in mammalian cells.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Proteína Quinasa Activada por ADN/metabolismo , Glioma/metabolismo , Glucosa/deficiencia , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Western Blotting , Células Cultivadas , Reparación del ADN/genética , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteína Quinasa Activada por ADN/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Inhibidores Enzimáticos/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Glioma/genética , Glioma/patología , Células HeLa , Humanos , Inmunoprecipitación , Ratones , Ratones Noqueados , Fosforilación , Proteínas Serina-Treonina Quinasas/deficiencia , ARN Interferente Pequeño/genética , Técnicas del Sistema de Dos Híbridos
13.
Cell Cycle ; 11(17): 3237-49, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22895183

RESUMEN

The p53R2 protein, a newly identified member of the ribonucleotide reductase family that provides nucleotides for DNA damage repair, is directly regulated by p53. We show that p53R2 is also regulated by a MEK2 (ERK kinase 2/MAP kinase kinase 2)-dependent pathway. Increased MEK1/2 phosphorylation by serum stimulation coincided with an increase in the RNR activity in U2OS and H1299 cells. The inhibition of MEK2 activity, either by treatment with a MEK inhibitor or by transfection with MEK2 siRNA, dramatically decreased the serum-stimulated RNR activity. Moreover, p53R2 siRNA, but not R2 siRNA, significantly inhibits serum-stimulated RNR activity, indicating that p53R2 is specifically regulated by a MEK2-dependent pathway. Co-immunoprecipitation analyses revealed that the MEK2 segment comprising amino acids 65-171 is critical for p53R2-MEK2 interaction, and the binding domain of MEK2 is required for MEK2-mediated increased RNR activity. Phosphorylation of MEK1/2 was greatly augmented by ionizing radiation, and RNR activity was concurrently increased. Ionizing radiation-induced RNR activity was markedly attenuated by transfection of MEK2 or p53R2 siRNA, but not R2 siRNA. These data show that MEK2 is an endogenous regulator of p53R2 and suggest that MEK2 may associate with p53R2 and upregulate its activity.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Reparación del ADN/genética , Regulación Enzimológica de la Expresión Génica/fisiología , MAP Quinasa Quinasa 2/metabolismo , Ribonucleótido Reductasas/metabolismo , Anticuerpos Monoclonales , Western Blotting , Línea Celular Tumoral , Rayos gamma , Vectores Genéticos/genética , Humanos , Inmunoprecipitación , MAP Quinasa Quinasa 2/fisiología , Fosforilación , Interferencia de ARN , Conteo por Cintilación
14.
Cell Physiol Biochem ; 30(2): 359-71, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22739356

RESUMEN

Interstitial cells of Cajal (ICCs) are pacemaker cells that activate the periodic spontaneous depolarization (pacemaker potentials) responsible for the production of slow waves in gastrointestinal smooth muscle. Under current clamping, ICCs had a mean resting membrane potential of -58 ± 3 mV and externally applied ET produced membrane depolarization in a dosedependent manner. These effects were reduced by intracellular GDP beta S. A comparison of the concentration-dependent membrane depolarizations on pacemaker potentials to ET-1, ET-2 and ET-3 showed a rank order of potency ET-1≥ET-2≥ET-3 in cultured murine small intestinal ICCs. The pretreatment with Ca(2+)-free solution and thapsigargin, a Ca(2+)-ATPase inhibitor in endoplasmic reticulum, abolished the generation of pacemaker potentials and suppressed the ET-1 induced membrane depolarizations. Chelerythrine and calphostin C, protein kinase C inhibitors or naproxen, an inhibitor of cyclooxygenase, did not block the ET-1 induced effects on pacemaker potentials. Pretreatment with BQ-123 (ET(A )receptor antagonist) or BQ-788 (ET(B )receptor antagonist) blocked the ET-1 induced effects on pacemaker potentials in cultured murine small intestinal ICCs. However, pretreatment with BQ-788 selectively did not block the ET-1 induced effects on pacemaker potentials in cultured murine large intestinal ICCs. Also, only externally applied selective ET(B )receptor agonist, IRL 1620 did not show any influence on pacemaker potentials in cultured murine large intestine ICCs. RT-PCR results indicated the presence of the ET(A )and ET(B )receptor in ICCs. These results suggested that ET-1 modulates pacemaker potentials through ET(A )and ET(B )receptor activation in murine small intestinal ICCs and ET(A )receptor activation in murine large intestinal ICCs by external Ca(2+) influx and internal Ca(2+) release via protein kinase C or cyclooxygenase-independent mechanism. Therefore, the ICCs are targets for ET and their interaction can affect intestinal motility.


Asunto(s)
Células Intersticiales de Cajal/metabolismo , Intestino Grueso/citología , Intestino Delgado/citología , Receptores de Endotelina/metabolismo , Animales , Benzofenantridinas/farmacología , Calcio/metabolismo , ATPasas Transportadoras de Calcio/antagonistas & inhibidores , ATPasas Transportadoras de Calcio/metabolismo , Membrana Celular/fisiología , Células Cultivadas , Endotelina-1/farmacología , Endotelina-2/farmacología , Endotelina-3/farmacología , Células Intersticiales de Cajal/citología , Células Intersticiales de Cajal/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Ratones , Ratones Endogámicos BALB C , Naproxeno/farmacología , Oligopéptidos/farmacología , Técnicas de Placa-Clamp , Péptidos Cíclicos/farmacología , Piperidinas/farmacología , Prostaglandina-Endoperóxido Sintasas/química , Prostaglandina-Endoperóxido Sintasas/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Receptores de Endotelina/agonistas , Tapsigargina/farmacología
15.
Acta Histochem ; 114(7): 647-52, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22172708

RESUMEN

Apurinic/apyrimidinic endonuclease (APE) acts as a regulator of p53 or vice versa in the cellular response to oxidative stress. Since oxidative stress-induced apoptosis is suggested in the pathophysiology of diabetic nephropathy, we proposed that APE may have a feasible role in the progression of diabetic complications. We investigated the interrelationship between APE and p53 in streptozotocin-induced diabetic rat kidneys. Variable parameters on kidneys were checked 12 weeks after streptozotocin administration with or without chitosan oligosaccharide (COS) treatment. Streptozotocin administration caused changes as seen in early diabetic nephropathy with increased kidney size, increased p53, decreased APE, and increased cleaved caspase-3. COS was not suspected as being detrimental to renal measurements, and caused the augmentation of APE after streptozotocin administration. The augmented APE, in association with increased p53, suppressed cleaved caspase-3. 8-OHdG was mainly immunolocalized in the distal tubules, but also in the proximal tubules after streptozotocin administration without COS treatment, while APE was observed in proximal tubules in all groups. These results suggested that p53-dependent apoptosis resulting in suppressed APE might be an underlying mechanism of streptozotocin-induced nephropathy.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Diabetes Mellitus Experimental/enzimología , Nefropatías Diabéticas/enzimología , Animales , Nitrógeno de la Urea Sanguínea , Caspasa 3/metabolismo , Quitosano/farmacología , Quitosano/uso terapéutico , Creatinina/sangre , Daño del ADN , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/tratamiento farmacológico , Nefropatías Diabéticas/sangre , Nefropatías Diabéticas/tratamiento farmacológico , Activadores de Enzimas/farmacología , Activadores de Enzimas/uso terapéutico , Masculino , Nefronas/efectos de los fármacos , Nefronas/enzimología , Nefronas/patología , Tamaño de los Órganos/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Estreptozocina , Proteína p53 Supresora de Tumor/metabolismo
16.
Free Radic Res ; 45(6): 728-34, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21473738

RESUMEN

p53 plays a major role in apoptosis through activation of pro-apoptotic gene Bax. It also regulates apurinic/apyrimidinic endonuclease (APE) expression in the base excision repair pathway against oxidative DNA damages. This study investigated whether p53-dependent apoptosis is correlated with APE using an experimental rat model of hydronephrosis. Hydronephrosis was induced by partial ligation of the right ureter. Animals were sacrificed on scheduled time after unilateral ureteral obstruction and the expression of 8-OHdG, γ-H2AX, apoptotic proteins and APE was determined. The accumulated p53 activated Bax and caspase-3 7 days after hydronephrosis induction and the resulting high levels of p53-dependent apoptotic proteins and γ-H2AX tended to decrease APE. The intensities of 8-OHdG and caspase-3 immunolocalization significantly increased in obstructed kidneys than in sham-operated kidneys, although APE immunoreactivity increased after hydronephrosis induction. These results suggest that oxidative DNA damages in obstructed kidneys may trigger p53-dependent apoptosis through repression of APE.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/biosíntesis , Hidronefrosis/patología , Riñón/patología , Proteína p53 Supresora de Tumor/metabolismo , 8-Hidroxi-2'-Desoxicoguanosina , Animales , Apoptosis , Proteínas Reguladoras de la Apoptosis/metabolismo , Caspasa 3/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Represión Enzimática , Hidronefrosis/metabolismo , Riñón/metabolismo , Masculino , Oxidación-Reducción , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley
17.
J Chem Neuroanat ; 40(3): 239-42, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20609383

RESUMEN

Unlike steroidogenic acute regulatory protein (StAR), one of the cholesterol transport protein, little attention is given to StarD6 which belongs to a family of StAR-related lipid transfer domain proteins. Although we undertook previous works with StarD6 in the nervous system, the characteristics are in controversy to date. Therefore, we attempted to investigate the morphological characteristics of StarD6 in the nervous system are the same as StAR in vitro and in vivo. The number of immunoreactive cells was significantly different by StAR or StarD6 in the cultured glioblastoma cell lines and dopaminergic neuronal cell lines. StarD6 immunoreactivity was changed by the presence of DNA-dependent protein kinase, while the dependency was not observed in StAR immunoreactivity. Besides, StarD6 was mainly observed in the stratum pyramidale and StAR in the other strata of normal rat hippocampus proper. Increased immunolocalization of StAR and StarD6 was seen in the stratum pyramidale and the strata lacunosum-moleculare, respectively, 3h after pilocarpine-induced epilepsy. Taken together, morphological aspects of StarD6 were significantly different from those of StAR in cultured glial and neuronal cells, as well as the distribution in the normal and epileptic rat hippocampus. These results suggested that StarD6 did not mark the same as StAR in vitro and in vivo.


Asunto(s)
Proteínas Portadoras/metabolismo , Hipocampo/metabolismo , Neuroglía/metabolismo , Neuronas/metabolismo , Fosfoproteínas/metabolismo , Animales , Células Cultivadas , Inmunohistoquímica , Ratas
18.
Urol Int ; 85(2): 216-20, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20530960

RESUMEN

AIM: Since DNA damage-related apoptosis is raising concerns regarding abnormal spermatogenesis, we investigated the changes in γ-H2AX during testicular germ cell apoptotic responses in the varicocele model. MATERIALS AND METHODS: Varicocele was induced by partial ligation of the left renal vein and animals were sacrificed at 1, 3, and 4 weeks after varicocele creation. The levels of activated p53 and γ-H2AX formation were determined by Western blot analysis and immunohistochemistry. RESULTS: γ-H2AX formation was augmented after varicocele creation, while a significant increase in p53 phosphorylation was detected in a time course-dependent manner. Varicocele-dependent nuclear γ-H2AX staining in the primary spermatocytes was prominent as degenerative foci, while little differences could be detected in spermatogonia. CONCLUSIONS: These results show that experimental varicocele may induce p53-dependent apoptosis through activation of γ-H2AX in the primary spermatocytes, and suggest that γ-H2AX may be related to apoptotic signal transduction in experimental varicocele.


Asunto(s)
Apoptosis , Histonas/metabolismo , Transducción de Señal , Espermatocitos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Varicocele/metabolismo , Animales , Western Blotting , Modelos Animales de Enfermedad , Inmunohistoquímica , Masculino , Fosforilación , Ratas , Ratas Sprague-Dawley , Espermatocitos/patología , Espermatogonias/metabolismo , Espermatogonias/patología , Factores de Tiempo , Varicocele/patología
19.
Mol Cell Biol ; 29(8): 2264-77, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19188437

RESUMEN

Apurinic/apyrimidinic endonuclease 1 (Ape1/Ref-1) dysregulation has been identified in several human tumors and in patients with a variety of neurodegenerative diseases. However, the function of Ape1/Ref-1 is unclear. We show here that Ape1/Ref-1 increases the expression of glial cell-derived neurotropic factor (GDNF) receptor alpha1 (GFRalpha1), a key receptor for GDNF. Expression of Ape1/Ref-1 led to an increase in the GDNF responsiveness in human fibroblast. Ape1/Ref-1 induced GFRalpha1 transcription through enhanced binding of NF-kappaB complexes to the GFRalpha1 promoter. GFRalpha1 levels correlate proportionally with Ape1/Ref-1 in cancer cells. The knockdown of endogenous Ape1/Ref-1 in pancreatic cancer cells markedly suppressed GFRalpha1 expression and invasion in response to GNDF, while overexpression of GFRalpha1 restored invasion. In neuronal cells, the Ape1/Ref-1-mediated increase in GDNF responsiveness not only stimulated neurite outgrowth but also protected the cells from beta-amyloid peptide and oxidative stress. Our results show that Ape1/Ref-1 is a novel physiological regulator of GDNF responsiveness, and they also suggest that Ape1/Ref-1-induced GFRalpha1 expression may play important roles in pancreatic cancer progression and neuronal cell survival.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/fisiología , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Factor Neurotrófico Derivado de la Línea Celular Glial/fisiología , Regulación hacia Arriba/genética , Animales , Línea Celular Tumoral , Humanos , Ratones , FN-kappa B/metabolismo , Invasividad Neoplásica , Neuritas , Neuronas/citología , Estrés Oxidativo , Neoplasias Pancreáticas/patología , Regiones Promotoras Genéticas
20.
J Biol Chem ; 284(15): 9845-53, 2009 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-19176521

RESUMEN

53BP1 (p53-binding protein 1) is a conserved nuclear protein that is phosphorylated in response to DNA damage and rapidly recruited to the site of DNA double strand breaks, demonstrating its role in the early events to DNA damage and repair of damaged DNA. In this study, we used the yeast two-hybrid system to identify proteins that interact with 53BP1. Identification and characterization of 53BP1 protein interactions may help to further elucidate the function and regulation of 53BP1. We identified protein phosphatase 5 (PP5), a serine/threonine phosphatase that has been implicated in multiple cellular function, as a 53BP1-binding protein. This interaction further confirmed that 53BP1 interacts with PP5 in PP5-overexpressing U2OS cells, after radiomimetic agent neocarzinostatin (NCS) treatment. 53BP1 dephosphorylation at Ser-25 and Ser-1778 was accelerated in PP5-overexpressing U2OS cells following NCS treatment, and its dephosphorylation was correlated with reduced phospho-53BP1 foci formation. In contrast, the overexpression of PP5 had no effect on NCS-activated BRCA1-Ser-1524 phosphorylation. Additionally, PP5 down-regulation inhibited the dephosphorylation of 53BP1 on Ser-1778 and the disappearance of phospho-53BP1 foci following NCS treatment. Moreover, non-homologous end-joining activity was reduced in PP5-overexpressing U2OS cells. These findings indicate that PP5 plays an important role in the regulation of 53BP1 phosphorylation and activity in vivo.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Línea Celular , Línea Celular Tumoral , ADN/metabolismo , Daño del ADN , Reparación del ADN , Regulación hacia Abajo , Humanos , Microscopía Fluorescente/métodos , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Fosforilación , Unión Proteica , Factores de Tiempo , Proteína 1 de Unión al Supresor Tumoral P53 , Cinostatina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA