Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 14161, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34239013

RESUMO

The integrated stress response (ISR) is a central cellular adaptive program that is activated by diverse stressors including ER stress, hypoxia and nutrient deprivation to orchestrate responses via activating transcription factor 4 (ATF4). We hypothesized that ATF4 is essential for the adaptation of human glioblastoma (GB) cells to the conditions of the tumor microenvironment and is contributing to therapy resistance against chemotherapy. ATF4 induction in GB cells was modulated pharmacologically and genetically and investigated in the context of temozolomide treatment as well as glucose and oxygen deprivation. The relevance of the ISR was analyzed by cell death and metabolic measurements under conditions to approximate aspects of the GB microenvironment. ATF4 protein levels were induced by temozolomide treatment. In line, ATF4 gene suppressed GB cells (ATF4sh) displayed increased cell death and decreased survival after temozolomide treatment. Similar results were observed after treatment with the ISR inhibitor ISRIB. ATF4sh and ISRIB treated GB cells were sensitized to hypoxia-induced cell death. Our experimental study provides evidence for an important role of ATF4 for the adaptation of human GB cells to conditions of the tumor microenvironment characterized by low oxygen and nutrient availability and for the development of temozolomide resistance. Inhibiting the ISR in GB cells could therefore be a promising therapeutic approach.


Assuntos
Fator 4 Ativador da Transcrição/metabolismo , Adaptação Fisiológica , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Temozolomida/uso terapêutico , Hipóxia Tumoral , Acetamidas/farmacologia , Fator 4 Ativador da Transcrição/genética , Adaptação Fisiológica/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Cicloexilaminas/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/genética , Glutamina/metabolismo , Humanos , Consumo de Oxigênio/efeitos dos fármacos , Estresse Fisiológico/efeitos dos fármacos , Estresse Fisiológico/genética , Temozolomida/farmacologia , Hipóxia Tumoral/efeitos dos fármacos
2.
Int J Mol Med ; 45(5): 1385-1396, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32323755

RESUMO

Adenosine monophosphate (AMP)­activated protein kinase (AMPK) is a major cellular energy sensor that is activated by an increase in the AMP/adenosine triphosphate (ATP) ratio. This causes the initiation of adaptive cellular programs, leading to the inhibition of anabolic pathways and increasing ATP synthesis. AMPK indirectly inhibits mammalian target of rapamycin (mTOR) complex 1 (mTORC1), a serine/threonine kinase and central regulator of cell growth and metabolism, which integrates various growth inhibitory signals, such as the depletion of glucose, amino acids, ATP and oxygen. While neuroprotective approaches by definition focus on neurons, that are more sensitive under cell stress conditions, astrocytes play an important role in the cerebral energy homeostasis during ischemia. Therefore, the protection of astrocytic cells or other glial cells may contribute to the preservation of neuronal integrity and function. In the present study, it was thus hypothesized that a preventive induction of energy deprivation­activated signaling pathways via AMPK may protect astrocytes from hypoxia and glucose deprivation. Hypoxia­induced cell death was measured in a paradigm of hypoxia and partial glucose deprivation in vitro in the immortalized human astrocytic cell line SVG. Both the glycolysis inhibitor 2­deoxy­d­glucose (2DG) and the AMPK activator A­769662 induced the phosphorylation of AMPK, resulting in mTORC1 inhibition, as evidenced by a decrease in the phosphorylation of the target ribosomal protein S6 (RPS6). Treatment with both 2DG and A­769662 also decreased glucose consumption and lactate production. Furthermore, A­769662, but not 2DG induced an increase in oxygen consumption, possibly indicating a more efficient glucose utilization through oxidative phosphorylation. Hypoxia­induced cell death was profoundly reduced by treatment with 2DG or A­769662. On the whole, the findings of the present study demonstrate, that AMPK activation via 2DG or A­769662 protects astrocytes under hypoxic and glucose­depleted conditions.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Astrócitos/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Hipóxia/tratamento farmacológico , Substâncias Protetoras/farmacologia , Astrócitos/metabolismo , Compostos de Bifenilo , Desoxiglucose/farmacologia , Glucose/metabolismo , Glicólise/efeitos dos fármacos , Humanos , Hipóxia/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Fosforilação Oxidativa/efeitos dos fármacos , Pironas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Tiofenos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA