Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Biochem Pharmacol ; 171: 113714, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31738894

RESUMEN

Brain tumours are among the deadliest tumours being highly resistant to currently available therapies. The proliferative behaviour of gliomas is strongly influenced by ion channel activity. Small-conductance calcium-activated potassium (SK/KCa) channels are a family of ion channels that are associated with cell proliferation and cell survival. A combined treatment of classical anti-cancer agents and pharmacological SK channel modulators has not been addressed yet. We used the gold-derivative auranofin to induce cancer cell death by targeting thioredoxin reductases in combination with CyPPA to activate SK channels in neuro- and glioblastoma cells. Combined treatment with auranofin and CyPPA induced massive mitochondrial damage and potentiated auranofin-induced toxicity in neuroblastoma cells in vitro. In particular, mitochondrial integrity, respiration and associated energy generation were impaired. These findings were recapitulated in patient-derived glioblastoma neurospheres yet not observed in non-cancerous HT22 cells. Taken together, integrating auranofin and SK channel openers to affect mitochondrial health was identified as a promising strategy to increase the effectiveness of anti-cancer agents and potentially overcome resistance.


Asunto(s)
Auranofina/farmacología , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Neuroblastoma/metabolismo , Pirazoles/farmacología , Pirimidinas/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/agonistas , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Auranofina/administración & dosificación , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología , Pirazoles/administración & dosificación , Pirimidinas/administración & dosificación , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo , Esferoides Celulares/efectos de los fármacos , Reductasa de Tiorredoxina-Disulfuro/antagonistas & inhibidores , Reductasa de Tiorredoxina-Disulfuro/metabolismo
2.
Cell Death Dis ; 10(10): 690, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31534165

RESUMEN

Patients with aggressive brain tumors, named glioblastoma multiforme (GBM), have a poor prognoses. Here we explored if the ER stress/unfolded protein response (UPR) is involved in the pathophysiology of GBM and may provide novel therapeutic targets. Immunohistochemical analyses of a tissue microarray containing primary GBM specimens showed strong variability in expression of the UPR markers GRP78/BiP, XBP1, and ATF4. Interestingly, high ATF4 expression was associated with poor overall survival suggesting involvement of PERK signaling in GBM progression. In vitro experiments using patient-derived neurospheres, enriched for GBM stem cells (GSCs), showed high sensitivity for the ER stressor thapsigargin (Tg) mainly via PERK signaling. In contrast, neurospheres-derived differentiated GBM cells were less sensitive likely due to lower UPR activity as indicated by comparative transcriptional profiling. Tg and Tunicamycin strongly reduced neurosphere forming ability of GSCs that was linked with potent PERK-dependent downregulation of SOX2 protein. Interestingly, SOX2 downregulation occurred directly via PERK, not requiring downstream activation of the PERK-UPR pathway. Moreover, PERK inactivation resulted in aberrant serum-induced differentiation of GBM neurospheres accompanied by persistent SOX2 expression, delayed upregulation of GFAP and reduced cell adherence. In conclusion, we provide evidence that PERK signaling contributes to the prognoses of primary GBM patients and identified PERK as a novel regulator of SOX2 expression and GSC differentiation. The role of PERK appeared to be pleiotropic involving UPR-dependent, as well as novel identified noncanonical mechanisms regulating SOX2. ER stress and PERK modulation appear to provide promising therapeutic targets for therapy in GBM.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Estrés del Retículo Endoplásmico , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Factores de Transcripción SOXB1/metabolismo , eIF-2 Quinasa/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Diferenciación Celular/fisiología , Chaperón BiP del Retículo Endoplásmico , Glioblastoma/genética , Glioblastoma/patología , Humanos , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Factores de Transcripción SOXB1/genética , Transducción de Señal , Respuesta de Proteína Desplegada , eIF-2 Quinasa/genética
3.
Biochem Pharmacol ; 118: 1-8, 2016 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-27106078

RESUMEN

Endoplasmic reticulum (ER) stress disrupts among others protein homeostasis in cells leading to the activation of the unfolded protein response (UPR) that is crucial for restoring this balance and cell survival. Hypoxia, reactive oxygen species and nutrient deprivation, conditions commonly present in the tumor microenvironment, are well-known triggers of the UPR. Apart from being an adaptive response, recently the UPR has been implicated in oncogenesis. Here we review the current understanding of the UPR in the most life threatening brain tumor in adults, glioblastoma multiforme (GBM). The UPR is controlled by BiP/GRP78 and three different sensors, PERK, IRE1 and ATF6. In orthotopic GBM mouse models IRE1 was reported to control angiogenesis, invasion and mesenchymal differentiation. Furthermore, PERK also was found to stimulate GBM growth. However, a direct role of the UPR in gliomagenesis remains to be demonstrated. Patient samples display chronic activation of the UPR and in vitro standard chemo- and radiotherapy partially act by aggravating ER stress leading to cell death. The UPR has been linked to enhanced sensitivity for apoptosis-inducing agents such as TRAIL and MDA-7. A number of agents such as proteasome inhibitors and several natural products were reported to exert cytotoxicity by enhancing ER stress in GBM cells, and some demonstrated activity in clinical studies. Finally, ER stress was suggested to be implicated in the maintenance of homeostasis in GBM stem cells. Taken together, the UPR appears to play an important role in GBM tumor progression and is a promising target for developing novel therapeutic interventions.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinogénesis , Estrés del Retículo Endoplásmico/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Modelos Biológicos , Terapia Molecular Dirigida , Respuesta de Proteína Desplegada/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Carcinogénesis/efectos de los fármacos , Chaperón BiP del Retículo Endoplásmico , Glioblastoma/metabolismo , Glioblastoma/patología , Glioblastoma/prevención & control , Humanos , Terapia Molecular Dirigida/tendencias , Clasificación del Tumor , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
4.
PLoS One ; 10(12): e0145393, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26700636

RESUMEN

Glioblastoma (GBM) is a highly infiltrative brain tumor in which cells with properties of stem cells, called glioblastoma stem cells (GSCs), have been identified. In general, the dominant view is that GSCs are responsible for the initiation, progression, invasion and recurrence of this tumor. In this study, we addressed the question whether the differentiation status of GBM cells is associated with their invasive capacity. For this, several primary GBM cell lines were used, cultured either as neurospheres known to enrich for GSCs or in medium supplemented with 10% FCS that promotes differentiation. The differentiation state of the cells was confirmed by determining the expression of stem cell and differentiation markers. The migration/invasion potential of these cells was tested using in vitro assays and intracranial mouse models. Interestingly, we found that serum-induced differentiation enhanced the invasive potential of GBM cells, which was associated with enhanced MMP9 expression. Chemical inhibition of MMP9 significantly reduced the invasive potential of differentiated cells in vitro. Furthermore, the serum-differentiated cells could revert back to an undifferentiated/stem cell state that were able to form neurospheres, although with a reduced efficiency as compared to non-differentiated counterparts. We propose a model in which activation of the differentiation program in GBM cells enhances their infiltrative potential and that depending on microenvironmental cues a significant portion of these cells are able to revert back to an undifferentiated state with enhanced tumorigenic potential. Thus, effective therapy should target both GSCs and differentiated offspring and targeting of differentiation-associated pathways may offer therapeutic opportunities to reduce invasive growth of GBM.


Asunto(s)
Diferenciación Celular , Glioblastoma/patología , Metaloproteinasa 9 de la Matriz/fisiología , Invasividad Neoplásica , Animales , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Humanos , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Suero/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...