Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cells ; 12(9)2023 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-37174691

RESUMEN

Lysosomotropic agent chloroquine was shown to sensitize non-stem glioblastoma cells to radiation in vitro with p53-dependent apoptosis implicated as one of the underlying mechanisms. The in vivo outcomes of chloroquine or its effects on glioblastoma stem cells have not been previously addressed. This study undertakes a combinatorial approach encompassing in vitro, in vivo and in silico investigations to address the relationship between chloroquine-mediated radiosensitization and p53 status in glioblastoma stem cells. Our findings reveal that chloroquine elicits antagonistic impacts on signaling pathways involved in the regulation of cell fate via both transcription-dependent and transcription-independent mechanisms. Evidence is provided that transcriptional impacts of chloroquine are primarily determined by p53 with chloroquine-mediated activation of pro-survival mevalonate and p21-DREAM pathways being the dominant response in the background of wild type p53. Non-transcriptional effects of chloroquine are conserved and converge on key cell fate regulators ATM, HIPK2 and AKT in glioblastoma stem cells irrespective of their p53 status. Our findings indicate that pro-survival responses elicited by chloroquine predominate in the context of wild type p53 and are diminished in cells with transcriptionally impaired p53. We conclude that p53 is an important determinant of the balance between pro-survival and pro-death impacts of chloroquine and propose that p53 functional status should be taken into consideration when evaluating the efficacy of glioblastoma radiosensitization by chloroquine.


Asunto(s)
Glioblastoma , Fármacos Sensibilizantes a Radiaciones , Humanos , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Cloroquina/farmacología , Fármacos Sensibilizantes a Radiaciones/farmacología , Células Madre/metabolismo , Medición de Riesgo , Proteínas Portadoras , Proteínas Serina-Treonina Quinasas/metabolismo
2.
DNA Repair (Amst) ; 98: 103037, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33418482

RESUMEN

Previously we showed that human monocytes isolated from peripheral blood display downregulation of several DNA repair proteins, including XRCC1, ligase III, PARP-1 and DNA-PKCS, resulting in a deficiency of DNA repair, while in macrophages derived from monocytes the repair protein expression and DNA repair is restored. To see whether this is a specific phenomenon of human monocytes and macrophages, we assessed the expression of these repair genes in mice. We also addressed the question at which differentiation step in bone marrow cells downregulation of DNA repair gene expression occurs. The study revealed that mouse monocytes, similar to human, lack the expression of XRCC1, ligase III, PARP-1 and DNA-PKCS. If mice were treated with total body irradiation, they showed significant apoptosis in bone marrow monocytes, but not in peritoneal macrophages. This was also observed after treatment with the methylating anticancer drug temozolomide, resulting in high death rate of monocytes, but not macrophages. Monocytes arise from hematopoietic stem cells. Even the early stem cell fraction (LT-HSC) expressed detectable amounts of XRCC1, which was transiently upregulated, achieving the highest expression level in CMP (common myeloid progenitor) and, during the subsequent differentiation process, downregulated up to a non-detectable level in monocytes. The immediate monocyte precursor GMP also expressed ligase III, PARP-1 and DNA-PKCS. All these repair genes lacking in monocytes were upregulated again in macrophages. The sensitivity of monocytes, macrophages and precursor cells roughly correlated with their XRCC1 expression level. Monocytes, but not macrophages, also displayed strong γH2AX focal staining, indicating the presence of non-repaired DNA double-strand breaks following total body irradiation. Overall, the data revealed that murine monocytes exhibit the same DNA repair-impaired phenotype and high sensitivity compared to macrophages as observed in human. Therefore, the repair deficiency previously described for human monocytes appears to be a general property of this cell type.


Asunto(s)
Daño del ADN , Reparación del ADN , Rayos gamma , Macrófagos/metabolismo , Monocitos/metabolismo , Temozolomida/toxicidad , Animales , Apoptosis , ADN/efectos de los fármacos , ADN/metabolismo , ADN/efectos de la radiación , Roturas del ADN de Doble Cadena , ADN Ligasa (ATP)/genética , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/genética , Femenino , Regulación de la Expresión Génica , Histonas/análisis , Histonas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/fisiología , Macrófagos/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Monocitos/efectos de los fármacos , Monocitos/fisiología , Monocitos/efectos de la radiación , Poli(ADP-Ribosa) Polimerasa-1/genética , Proteínas de Unión a Poli-ADP-Ribosa/genética , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Células Madre/efectos de la radiación , Temozolomida/farmacología , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X/genética
3.
Cancer Res ; 79(1): 99-113, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30361254

RESUMEN

The DNA-methylating drug temozolomide, which induces cell death through apoptosis, is used for the treatment of malignant glioma. Here, we investigate the mechanisms underlying the ability of temozolomide to induce senescence in glioblastoma cells. Temozolomide-induced senescence was triggered by the specific DNA lesion O6-methylguanine (O6MeG) and characterized by arrest of cells in the G2-M phase. Inhibitor experiments revealed that temozolomide-induced senescence was initiated by damage recognition through the MRN complex, activation of the ATR/CHK1 axis of the DNA damage response pathway, and mediated by degradation of CDC25c. Temozolomide-induced senescence required functional p53 and was dependent on sustained p21 induction. p53-deficient cells, not expressing p21, failed to induce senescence, but were still able to induce a G2-M arrest. p14 and p16, targets of p53, were silenced in our cell system and did not seem to play a role in temozolomide-induced senescence. In addition to p21, the NF-κB pathway was required for senescence, which was accompanied by induction of the senescence-associated secretory phenotype. Upon temozolomide exposure, we found a strong repression of the mismatch repair proteins MSH2, MSH6, and EXO1 as well as the homologous recombination protein RAD51, which was downregulated by disruption of the E2F1/DP1 complex. Repression of these repair factors was not observed in G2-M arrested p53-deficient cells and, therefore, it seems to represent a specific trait of temozolomide-induced senescence. SIGNIFICANCE: These findings reveal a mechanism by which the anticancer drug temozolomide induces senescence and downregulation of DNA repair pathways in glioma cells.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Reparación del ADN/efectos de los fármacos , Glioblastoma/patología , FN-kappa B/metabolismo , Temozolomida/farmacología , Animales , Antineoplásicos Alquilantes/farmacología , Apoptosis , Proteínas de la Ataxia Telangiectasia Mutada/genética , Ciclo Celular , Proliferación Celular , Senescencia Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Metilación de ADN , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , FN-kappa B/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Oncotarget ; 8(17): 27754-27771, 2017 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-27487141

RESUMEN

Integrins have been suggested as possible targets in anticancer therapy. Here we show that knockdown of integrins αVß3, αVß5, α3ß1 and α4ß1 and pharmacological inhibition using a cyclo-RGD integrin αVß3/αVß5 antagonist sensitized multiple high-grade glioma cell lines to temozolomide (TMZ)-induced cytotoxicity. The greatest effect was observed in LN229 cells upon integrin ß3 silencing, which led to inhibition of the FAK/Src/Akt/NFκB signaling pathway and increased formation of γH2AX foci. The integrin ß3 knockdown led to the proteasomal degradation of Rad51, reduction of Rad51 foci and reduced repair of TMZ-induced DNA double-strand breaks by impairing homologous recombination efficiency. The down-regulation of ß3 in Rad51 knockdown (LN229-Rad51kd) cells neither further sensitized them to TMZ nor increased the number of γH2AX foci, confirming causality between ß3 silencing and Rad51 reduction. RIP1 was found cleaved and IκBα significantly less degraded in ß3-silenced/TMZ-exposed cells, indicating inactivation of NFκB signaling. The anti-apoptotic proteins Bcl-xL, survivin and XIAP were proteasomally degraded and caspase-3/-2 cleaved. Increased H2AX phosphorylation, caspase-3 cleavage, reduced Rad51 and RIP1 expression, as well as sustained IκBα expression were also observed in mouse glioma xenografts treated with the cyclo-RGD inhibitor and TMZ, confirming the molecular mechanism in vivo. Our data indicates that ß3 silencing in glioma cells represents a promising strategy to sensitize high-grade gliomas to TMZ therapy.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Glioblastoma/tratamiento farmacológico , Integrina alfaVbeta3/metabolismo , Reparación del ADN por Recombinación , Animales , Antineoplásicos Alquilantes/uso terapéutico , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Dacarbazina/uso terapéutico , Regulación hacia Abajo , Citometría de Flujo , Técnicas de Silenciamiento del Gen , Glioblastoma/genética , Glioblastoma/patología , Humanos , Integrina alfaVbeta3/antagonistas & inhibidores , Integrina alfaVbeta3/genética , Integrina beta3/genética , Integrina beta3/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Inhibidor NF-kappaB alfa/metabolismo , Clasificación del Tumor , Péptidos Cíclicos/farmacología , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Receptores de Vitronectina/antagonistas & inhibidores , Receptores de Vitronectina/genética , Receptores de Vitronectina/metabolismo , Transducción de Señal , Temozolomida , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Oncotarget ; 7(41): 67235-67250, 2016 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-27626497

RESUMEN

Glioblastoma multiforme (GBM), a malignant brain tumor with a dismal prognosis, shows a high level of chemo- and radioresistance and, therefore, attempts to sensitize glioma cells are highly desired. Here, we addressed the question of whether artesunate (ART), a drug currently used in the treatment of malaria, enhances the killing response of glioblastoma cells to temozolomide (TMZ), which is the first-line therapeutic for GBM. We measured apoptosis, necrosis, autophagy and senescence, and the extent of DNA damage in glioblastoma cells. Further, we determined the tumor growth in nude mice. We show that ART enhances the killing effect of TMZ in glioblastoma cell lines and in glioblastoma stem-like cells. The DNA double-strand break level induced by TMZ was not clearly enhanced in the combined treatment regime. Also, we did not observe an attenuation of TMZ-induced autophagy, which is considered a survival mechanism. However, we observed a significant effect of ART on homologous recombination (HR) with downregulation of RAD51 protein expression and HR activity. Further, we found that ART is able to inhibit senescence induced by TMZ. Since HR and senescence are pro-survival mechanisms, its inhibition by ART appears to be a key node in enhancing the TMZ-induced killing response. Enhancement of the antitumor effect of TMZ by co-administration of ART was also observed in a mouse tumor model. In conclusion, the amelioration of TMZ-induced cell death upon ART co-treatment provides a rational basis for a combination regime of TMZ and ART in glioblastoma therapy.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias Encefálicas/patología , Senescencia Celular/efectos de los fármacos , Glioma/patología , Recombinación Homóloga/efectos de los fármacos , Animales , Artemisininas/farmacología , Artesunato , Muerte Celular/efectos de los fármacos , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Humanos , Ratones , Ratones Desnudos , Temozolomida , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Cancer Ther ; 15(11): 2665-2678, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27474153

RESUMEN

Malignant gliomas exhibit a high level of intrinsic and acquired drug resistance and have a dismal prognosis. First- and second-line therapeutics for glioblastomas are alkylating agents, including the chloroethylating nitrosoureas (CNU) lomustine, nimustine, fotemustine, and carmustine. These agents target the tumor DNA, forming O6-chloroethylguanine adducts and secondary DNA interstrand cross-links (ICL). These cross-links are supposed to be converted into DNA double-strand breaks, which trigger cell death pathways. Here, we show that lomustine (CCNU) with moderately toxic doses induces ICLs in glioblastoma cells, inhibits DNA replication fork movement, and provokes the formation of DSBs and chromosomal aberrations. Since homologous recombination (HR) is involved in the repair of DSBs formed in response to CNUs, we elucidated whether pharmacologic inhibitors of HR might have impact on these endpoints and enhance the killing effect. We show that the Rad51 inhibitors RI-1 and B02 greatly ameliorate DSBs, chromosomal changes, and the level of apoptosis and necrosis. We also show that an inhibitor of MRE11, mirin, which blocks the formation of the MRN complex and thus the recognition of DSBs, has a sensitizing effect on these endpoints as well. In a glioma xenograft model, the Rad51 inhibitor RI-1 clearly enhanced the effect of CCNU on tumor growth. The data suggest that pharmacologic inhibition of HR, for example by RI-1, is a reasonable strategy for enhancing the anticancer effect of CNUs. Mol Cancer Ther; 15(11); 2665-78. ©2016 AACR.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Recombinación Homóloga/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Aberraciones Cromosómicas/efectos de los fármacos , Daño del ADN , Metilasas de Modificación del ADN/metabolismo , Reparación del ADN , Enzimas Reparadoras del ADN/metabolismo , Replicación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Lomustina/farmacología , Proteína Homóloga de MRE11 , Ratones , Recombinasa Rad51/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Oncotarget ; 6(32): 33755-68, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26418950

RESUMEN

The outcome of cancer therapy strongly depends on the complex network of cell signaling pathways, including transcription factor activation following drug exposure. Here we assessed whether and how the MAP kinase (MAPK) cascade and its downstream target, the transcription factor AP-1, influence the sensitivity of malignant glioma cells to the anticancer drugs temozolomide (TMZ) and nimustine (ACNU). Both drugs induce apoptosis in glioma cells at late times following treatment. Activation of the MAPK cascade precedes apoptosis, as shown by phosphorylation of Jun kinase (JNK) and c-Jun, a main component of AP-1. Pharmacological inhibition and siRNA mediated knockdown of JNK and c-Jun reduced the level of apoptosis in LN-229 glioma cells treated with TMZ or ACNU. Analyzing the underlying molecular mechanism, we identified the pro-apoptotic gene BIM as a critical target of AP-1, which is upregulated following TMZ and ACNU. Importantly, shRNA mediated downregulation of BIM in the malignant glioma cell lines LN-229 and U87MG led to an attenuated cleavage of caspase-9 and, consequently, reduced the level of apoptosis following TMZ and ACNU treatment. Overall, we identified JNK/c-Jun activation and BIM induction as a late pro-apoptotic response of glioma cells treated with alkylating anticancer drugs.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Dacarbazina/análogos & derivados , Glioblastoma/patología , MAP Quinasa Quinasa 4/metabolismo , Proteínas de la Membrana/metabolismo , Nimustina/farmacología , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Antineoplásicos Alquilantes/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proteína 11 Similar a Bcl2 , Línea Celular Tumoral/efectos de los fármacos , Núcleo Celular/metabolismo , Ensayo Cometa , Dacarbazina/farmacología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Fosforilación , Estructura Terciaria de Proteína , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Temozolomida , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA