Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Crit Rev Oncol Hematol ; : 104461, 2024 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-39103129

RESUMEN

The treatment of thoracic tumors with ionizing radiation can cause radiation-induced lung injury (RILI), which includes radiation pneumonitis and radiation-induced pulmonary fibrosis. Preventing RILI is crucial for controlling tumor growth and improving quality of life. However, the serious adverse effects of traditional RILI treatment methods remain a major obstacle, necessitating the development of novel treatment options that are both safe and effective. This review summarizes the molecular mechanisms of RILI and explores novel treatment options, including natural compounds, gene therapy, nanomaterials, and mesenchymal stem cells. These recent experimental approaches show potential as effective prevention and treatment options for RILI in clinical practice.

2.
Cancer Med ; 13(15): e7456, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39118481

RESUMEN

OBJECTIVES: High-grade gliomas (HGGs) are highly malignant, aggressive, and have a high incidence and mortality rate. The aim of this study was to investigate survival outcomes and prognostic factors in patients with HGGs. METHODS: In this retrospective study, a total of 159 patients with histologically confirmed HGGs were included. The recruitment period was from January 2011 to December 2019. We evaluated patient demographic data, tumor characteristics, treatment methods, immunocytochemistry results, overall survival (OS) time, and progression-free survival (PFS) time using Kaplan-<>Meier survival analysis with log-rank testing. Additionally, we employed Cox regression analysis to identify independent factors associated with survival outcomes. RESULTS: Kaplan-Meier survival analysis revealed that the 1-, 2-, and 5-years OS rates were 81.8%, 50.3%, and 12.6%, respectively. Similarly, the 1-, 2-, and 5-years PFS rates were 50.9%, 22.4%, and 3.1%, respectively. The median OS duration was 35.0 months. The univariate analysis indicated that postoperative pathological classification, grade, and age were significantly associated with patient outcomes (p < 0.01). Among the patients, 147 received concurrent chemoradiotherapy, while 12 did not. The immunohistochemical markers of ki-67, MGMT, IDH1R132H, and p53 demonstrated statistically significant differences in their prognostic impact (p = 0.001, p = 0.020, p = 0.003, and p = 0.021, respectively). In conclusion, we found that grades, age, pathological classification, ki-67, MGMT, and IDH1R132H expression were statistically significantly associated with PFS (p < 0.01, p = 0.004, p = 0.003, p = 0.001, p = 0.036, and p = 0.028). Additionally, immunohistochemical expressions of TRIB3 and AURKA were significantly higher in patients with shorter survival (p = 0.015 and p = 0.023). CONCLUSIONS: Tumor grade and the use of concurrent chemoradiotherapy after surgery were independent prognostic factors that significantly influenced patient survival. Additionally, tumor grade and MGMT expression were found to be independent factors affecting progression-free survival (PFS). Notably, the expression of TRIB3 and AURKA was higher in patients with poor survival outcomes.


Asunto(s)
Neoplasias Encefálicas , Glioma , Clasificación del Tumor , Humanos , Femenino , Masculino , Glioma/mortalidad , Glioma/patología , Glioma/terapia , Glioma/metabolismo , Estudios Retrospectivos , Persona de Mediana Edad , Adulto , Pronóstico , Anciano , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/metabolismo , Quimioradioterapia , Adulto Joven , Estimación de Kaplan-Meier , Biomarcadores de Tumor/metabolismo , Supervivencia sin Progresión , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Metilasas de Modificación del ADN/genética , Metilasas de Modificación del ADN/metabolismo , Análisis Multivariante , Proteínas Supresoras de Tumor/metabolismo , Tasa de Supervivencia , Adolescente , Enzimas Reparadoras del ADN/metabolismo , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/análisis
3.
Redox Biol ; 69: 103030, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38181705

RESUMEN

Ferroptosis is a type of programmed cell death resulting from iron overload-dependent lipid peroxidation, and could be promoted by activating transcription factor 3 (ATF3). SIRT1 is an enzyme accounting for removing acetylated lysine residues from target proteins by consuming NAD+, but its role remains elusive in ferroptosis and activating ATF3. In this study, we found SIRT1 was activated during the process of RSL3-induced glioma cell ferroptosis. Moreover, the glioma cell death was aggravated by SIRT1 activator SRT2183, but suppressed by SIRT inhibitor EX527 or when SIRT1 was silenced with siRNA. These indicated SIRT1 sensitized glioma cells to ferroptosis. Furthermore, we found SIRT1 promoted RSL3-induced expressional upregulation and nuclear translocation of ATF3. Silence of ATF3 with siRNA attenuated RSL3-induced increases of ferrous iron and lipid peroxidation, downregulation of SLC7A11 and GPX4 and depletion of cysteine and GSH. Thus, SIRT1 promoted glioma cell ferroptosis by inducting ATF3 activation. Mechanistically, ATF3 activation was reinforced when RSL3-induced decline of NAD+ was aggravated by FK866 that could inhibit NAD + synthesis via salvage pathway, but suppressed when intracellular NAD+ was maintained at higher level by supplement of exogenous NAD+. Notably, the NAD + decline caused by RSL3 was enhanced when SIRT1 was further activated by SRT2183, but attenuated when SIRT1 activation was inhibited by EX527. These indicated SIRT1 promoted ATF3 activation via consumption of NAD+. Finally, we found RSL3 activated SIRT1 by inducing reactive oxygen species-dependent upregulation of AROS. Together, our study revealed SIRT1 activated by AROS sensitizes glioma cells to ferroptosis via activation of ATF3-dependent inhibition of SLC7A11 and GPX4.


Asunto(s)
Ferroptosis , Glioma , Humanos , NAD , Factor de Transcripción Activador 3/genética , Línea Celular Tumoral , Sirtuina 1/genética , Glioma/genética , Glioma/metabolismo , ARN Interferente Pequeño
4.
Biochim Biophys Acta Mol Basis Dis ; 1870(2): 166925, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38084873

RESUMEN

Pancreatic adenocarcinoma (PAAD), a common digestive malignant tumor, presents high mortality rates and limited treatment methods. Currently, chemotherapy remains the main therapy method for patients with PAAD. As a classical chemotherapy drug, cisplatin (DDP) is limited by dose-related toxicity in patients with PAAD. In this study, we demonstrated that TGM2 may be a treatment and prognosis marker in pancreatic cancer patients. Co-treatment of low dose of DDP and GK921, a transglutaminase (TGM2) inhibitor, is capable of synergistically inhibiting the PAAD cell viability and proliferation in vitro and in vivo. Based on in vitro study, GK921 inhibited the epithelial-to-mesenchymal transition (EMT) induced by TGM2 as well as aggravated cell cycle arrest and apoptosis resulted from DDP, making pancreatic cancer cells more sensible to DDP. Our results showed that GK921 increased the protein levels regarding E-cadherin as well as decreased the protein level regarding Snail2, N-cadherin, which indicated that GK921 inhibited EMT in pancreatic cancer cells. Snail2 overexpression inhibited GK921/DDP-induced cell apoptosis, as well as mitigated the GK921/DDP-caused cell death and the EMT inhibition. In vivo studies also found GK921/DDP combination can further inhibit the growth of PAAD without significantly side effects. To sum up, we showed that GK921 increased PAAD cells sensitivity to DDP via inhibiting EMT. As revealed, DDP/GK921 co-treatment could promisingly serve for treating PAAD patients.


Asunto(s)
Adenocarcinoma , Neoplasias Pancreáticas , Humanos , Cisplatino/farmacología , Adenocarcinoma/tratamiento farmacológico , Resistencia a Antineoplásicos , Línea Celular Tumoral , Neoplasias Pancreáticas/tratamiento farmacológico
6.
Acta Pharmacol Sin ; 44(10): 2125-2138, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37277492

RESUMEN

Parthanatos is a type of programmed cell death dependent on hyper-activation of poly (ADP-ribose) polymerase 1 (PARP-1). SIRT1 is a highly conserved nuclear deacetylase and often acts as an inhibitor of parthanatos by deacetylation of PARP1. Our previous study showed that deoxypodophyllotoxin (DPT), a natural compound isolated from the traditional herb Anthriscus sylvestris, triggered glioma cell death via parthanatos. In this study, we investigated the role of SIRT1 in DPT-induced human glioma cell parthanatos. We showed that DPT (450 nmol/L) activated both PARP1 and SIRT1, and induced parthanatos in U87 and U251 glioma cells. Activation of SIRT1 with SRT2183 (10 µmol/L) enhanced, while inhibition of SIRT1 with EX527 (200 µmol/L) or knockdown of SIRT1 attenuated DPT-induced PARP1 activation and glioma cell death. We demonstrated that DPT (450 nmol/L) significantly decreased intracellular NAD+ levels in U87 and U251 cells. Further decrease of NAD+ levels with FK866 (100 µmol/L) aggravated, but supplement of NAD+ (0.5, 2 mmol/L) attenuated DPT-induced PARP1 activation. We found that NAD+ depletion enhanced PARP1 activation via two ways: one was aggravating ROS-dependent DNA DSBs by upregulation of NADPH oxidase 2 (NOX2); the other was reinforcing PARP1 acetylation via increase of N-acetyltransferase 10 (NAT10) expression. We found that SIRT1 activity was improved when being phosphorylated by JNK at Ser27, the activated SIRT1 in reverse aggravated JNK activation via upregulating ROS-related ASK1 signaling, thus forming a positive feedback between JNK and SIRT1. Taken together, SIRT1 activated by JNK contributed to DPT-induced human glioma cell parthanatos via initiation of NAD+ depletion-dependent upregulation of NOX2 and NAT10.


Asunto(s)
Glioma , Parthanatos , Sirtuina 1 , Humanos , Glioma/tratamiento farmacológico , Acetiltransferasas N-Terminal/genética , Acetiltransferasas N-Terminal/metabolismo , NAD/metabolismo , NADPH Oxidasa 2/metabolismo , Parthanatos/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Sirtuina 1/metabolismo , Regulación hacia Arriba
7.
Acta Pharmacol Sin ; 44(9): 1906-1919, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37186123

RESUMEN

Parthanatos is a type of programmed cell death initiated by over-activated poly (ADP-ribose) polymerase 1 (PARP1). Nuclear translocation of apoptosis inducing factor (AIF) is a prominent feature of parthanatos. But it remains unclear how activated nuclear PARP1 induces mitochondrial AIF translocation into nuclei. Evidence has shown that deoxypodophyllotoxin (DPT) induces parthanatos in glioma cells via induction of excessive ROS. In this study we explored the downstream signal of activated PARP1 to induce nuclear translocation of AIF in DPT-triggered glioma cell parthanatos. We showed that treatment with DPT (450 nM) induced PARP1 over-activation and Tax1 binding protein 1 (TAX1BP1) distribution to mitochondria in human U87, U251 and U118 glioma cells. PARP1 activation promoted TAX1BP1 distribution to mitochondria by depleting nicotinamide adenine dinucleotide (NAD+). Knockdown of TAX1BP1 with siRNA not only inhibited TAX1BP1 accumulation in mitochondria, but also alleviated nuclear translocation of AIF and glioma cell death. We demonstrated that TAX1BP1 enhanced the activity of respiratory chain complex I not only by upregulating the expression of ND1, ND2, NDUFS2 and NDUFS4, but also promoting their assemblies into complex I. The activated respiratory complex I generated more superoxide to cause mitochondrial depolarization and nuclear translocation of AIF, while the increased mitochondrial superoxide reversely reinforced PARP1 activation by inducing ROS-dependent DNA double strand breaks. In mice bearing human U87 tumor xenograft, administration of DPT (10 mg· kg-1 ·d-1, i.p., for 8 days) markedly inhibited the tumor growth accompanied by NAD+ depletion, TAX1BP1 distribution to mitochondria, AIF distribution to nuclei as well as DNA DSBs and PARP1 activation in tumor tissues. Taken together, these data suggest that TAX1BP1 acts as a downstream signal of activated PARP1 to trigger nuclear translocation of AIF by activation of mitochondrial respiratory chain complex I.


Asunto(s)
Glioma , Parthanatos , Humanos , Ratones , Animales , Factor Inductor de la Apoptosis/genética , Superóxidos/metabolismo , Especies Reactivas de Oxígeno/metabolismo , NAD/metabolismo , Transporte de Electrón , Complejo I de Transporte de Electrón , Glioma/metabolismo , Proteínas de Neoplasias/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo
8.
Mol Med Rep ; 27(3)2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36799163

RESUMEN

Maltol, a chemical isolated from ginseng root, has shown treatment effects on several pathological processes including osteoarthritis, diabetic peripheral neuropathy and liver fibrosis. Nevertheless, its effect on ischemia­induced neuron death remains elusive. In the present study, the treatment effect of maltol on ischemia­induced neuron damage was investigated by using oxygen and glucose deprivation (OGD) model in SH­SY5Y cells. In vitro studies revealed that maltol protected SH­SY5Y cells against OGD­induced chromatinolysis by inhibiting two reactive oxygen species (ROS)­regulated pathways. One was DNA double­strand breaks and the other was nuclear translocation of apoptosis inducing factor. Mechanistically, maltol not only inhibited OGD­induced depletion of glutathione and cysteine by maintaining cystine/glutamate antiporter (xCT) level, but also abrogated OGD­induced catalase downregulation. Meanwhile, maltol also alleviated OGD­induced inactivation of mTOR by attenuating OGD­induced depletion of adenosine triphosphate and pyruvate and downregulation of pyruvate kinase M2, indicating that maltol inhibited the glycolysis dysfunction caused by OGD. Considering that activated mammalian target of the rapamycin (mTOR) could lead to enhanced xCT expression and decreased catalase degradation by autophagy, these findings indicated that maltol attenuated OGD­induced ROS via inhibition of mTOR inactivation by maintaining pyruvate level. Taken together, it was demonstrated that maltol prevented OGD­induced chromatinolysis in SH­SY5Y cells via inhibiting pyruvate depletion.


Asunto(s)
Neuroblastoma , Oxígeno , Humanos , Oxígeno/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Ácido Pirúvico , Glucosa/metabolismo , Catalasa , Serina-Treonina Quinasas TOR/metabolismo
9.
Sci Rep ; 12(1): 14675, 2022 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-36038617

RESUMEN

Shikonin induces glioma cell death via necroptosis, a caspase-independent programmed cell death pathway that is chiefly regulated by receptor-interacting serine/threonine protein kinase1 (RIP1) and 3 (RIP3). Chromatinolysis is considered as one of the key events leading to cell death during necroptosis. It is usually accompanied with nuclear translocation of AIF and formation of γ-H2AX. Cyclophilin A (CypA) is reported to participate in the nuclear translocation of AIF during apoptosis. However, it remains unclear whether CypA contributes to necroptosis and regulation of chromatinolysis. In this study, our results revealed for the first time that shikonin promoted time-dependent CypA activation, which contributed to nuclear translocation of AIF and γ-H2AX formation. In vitro studies showed that knockdown of CypA by siRNA or inhibition of CypA by its specific inhibitor, cyclosporine A (CsA), not only significantly mitigated shikonin-induced glioma cell death, but also prevented chromatinolysis. Mechanistically, activated CypA targeted mitochondria and triggered mitochondrial superoxide overproduction, which then promoted AIF translocation from mitochondria into the nucleus by depolarizing the mitochondria and intensified the formation of γ-H2AX by promoting intracellular accumulation of ROS. Additionally, the CypA in the nucleus can form DNA degradation complexes with AIF and γ-H2AX, which also promote the execution of chromatinolysis. Thus, we demonstrate that CypA contributes to shikonin-induced glioma cell necroptosis and promotion of chromatinolysis.


Asunto(s)
Glioma , Necroptosis , Apoptosis , Factor Inductor de la Apoptosis/metabolismo , Ciclofilina A/metabolismo , Glioma/metabolismo , Humanos , Naftoquinonas , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo
10.
Biochim Biophys Acta Mol Basis Dis ; 1868(12): 166529, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36041715

RESUMEN

RSL3 is a synthetic molecule that inactivates glutathione peroxidase 4 to induce ferroptosis. However, its effect on glioma stem cells (GSC) remains unclear. In this study, we found that RSL3 significantly suppressed GSC proliferation and induced their differentiation into astrocytes, which was accompanied by the downregulation of stemness-related markers, including Nestin and Sox2. Combined transcriptome and proteome analyses further revealed that RSL3 promoted GSC differentiation by suppressing transglutaminase 2 (Tgm2), but not by ferroptosis-related pathways. Tgm2 overexpression in CSC2078 cells rescued the changes in stemness-related markers and differentiation caused by RSL3, which was mediated by inhibitor of DNA binding 1 (ID1) activation. Further studies identified ID1 as a downstream signaling target of Tgm2. Blocking the phosphoinositide-3 kinase (PI3K)/Akt pathway with LY294002 suppressed PI3K, p-Akt, and ID1 levels but not Tgm2. Tgm2 overexpression abrogated the changes in PI3K, p-Akt, and ID1 levels caused by LY294002. Taken together, we demonstrate that RSL3 does not induce ferroptosis; instead, it inhibits GSC proliferation and triggers their differentiation by suppressing the Tgm2/Akt/ID1 signaling axis.


Asunto(s)
Glioma , Proteínas Proto-Oncogénicas c-akt , Diferenciación Celular , ADN , Glioma/genética , Glioma/metabolismo , Humanos , Proteína 1 Inhibidora de la Diferenciación/genética , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Nestina , Fosfatidilinositol 3-Quinasa , Fosfatidilinositoles , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Proteína Glutamina Gamma Glutamiltransferasa 2 , Proteoma , Proteínas Proto-Oncogénicas c-akt/metabolismo
11.
Biochem Biophys Res Commun ; 589: 1-8, 2022 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-34883284

RESUMEN

BNIP3 is found to eliminate cancer cells via causing mitochondrial damage and endoplasmic reticulum stress, but it remains elusive of its role in regulating DNA double strand breaks (DSBs). In this study, we find that silibinin triggers DNA DSBs, ROS accumulation and expressional upregulation of BNIP3 in glioma cells. Mitigation of ROS with antioxidant GSH significantly inhibits silibinin-induced DNA DSBs and glioma cell death. Then, we find knockdown of BNIP3 with SiRNA obviously prevents silibinin-induced DNA DSBs and ROS accumulation. Mechanistically, BNIP3 knockdown not only reverses silibinin-triggered depletion of cysteine and GSH via maintaining xCT level, but also abrogates catalase decrease. Notably, silibinin-induced dephosphorylation of mTOR is also prevented when BNIP3 is knocked down. Given that activated mTOR could promote xCT expression and inhibit autophagic degradation of catalase, our data suggest that BNIP3 contributes to silibinin-induced DNA DSBs via improving intracellular ROS by inhibition of mTOR.


Asunto(s)
Roturas del ADN de Doble Cadena , Glioma/metabolismo , Glioma/patología , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Silibina/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Sistema de Transporte de Aminoácidos y+/metabolismo , Catalasa/metabolismo , Línea Celular Tumoral , Cisteína/metabolismo , Roturas del ADN de Doble Cadena/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Glutatión/metabolismo , Humanos , Especies Reactivas de Oxígeno/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
12.
Biochem Pharmacol ; 192: 114741, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34428443

RESUMEN

The resistance to cisplatin (DDP) and dose-related toxicity are the two important obstacles in the chemotherapy of prostate cancer (PCa) patients. The present study demonstrated that cotreatment of DDP and RSL3, a type of small molecular compound which can inactivate glutathione peroxidase 4 (GPX4) and induce ferroptosis, synergistically inhibited the viability and proliferation of PCa cells in vitro and in vivo at low dose. In vitro studies revealed that RSL3 improved that sensitivity of PCa cells to DDP by producing ROS and aggravating the cell cycle arrest and apoptosis caused by DDP. Mechanistically, RSL3 could decrease the ATP and pyruvate content as well as the protein levels of HKII, PFKP, PKM2, which indicated that RSL3 induced glycolysis dysfunction in prostate cancer cells. Rescuing RSL3-induced glycolysis dysfunction by supplement of exterior sodium pyruvate not only inhibited RSL3/DDP-induced changes of apoptosis-related proteins levels, but also mitigated the cell death caused by RSL3/DDP. In vivo studies further confirmed that cotreatment of RSL3 and DDP at low dose significantly inhibited the growth of PCa with no obvious side effects. Taken together, we demonstrated that RSL3 improved the sensitivity of PCa to DDP via causing glycolysis dysfunction. Our findings indicated that DDP-based chemotherapy combined with RSL3 might provide a promising therapy for PCa.


Asunto(s)
Antineoplásicos/administración & dosificación , Carbolinas/administración & dosificación , Cisplatino/administración & dosificación , Glucólisis/efectos de los fármacos , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Animales , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Glucólisis/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Próstata/patología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
13.
Acta Pharmacol Sin ; 42(10): 1690-1702, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34112960

RESUMEN

Ferroptotic cell death is characterized by iron-dependent lipid peroxidation that is initiated by ferrous iron and H2O2 via Fenton reaction, in which the role of activating transcription factor 3 (ATF3) remains elusive. Brucine is a weak alkaline indole alkaloid extracted from the seeds of Strychnos nux-vomica, which has shown potent antitumor activity against various tumors, including glioma. In this study, we showed that brucine inhibited glioma cell growth in vitro and in vivo, which was paralleled by nuclear translocation of ATF3, lipid peroxidation, and increases of iron and H2O2. Furthermore, brucine-induced lipid peroxidation was inhibited or exacerbated when intracellular iron was chelated by deferoxamine (500 µM) or improved by ferric ammonium citrate (500 µM). Suppression of lipid peroxidation with lipophilic antioxidants ferrostatin-1 (50 µM) or liproxstatin-1 (30 µM) rescued brucine-induced glioma cell death. Moreover, knockdown of ATF3 prevented brucine-induced accumulation of iron and H2O2 and glioma cell death. We revealed that brucine induced ATF3 upregulation and translocation into nuclei via activation of ER stress. ATF3 promoted brucine-induced H2O2 accumulation via upregulating NOX4 and SOD1 to generate H2O2 on one hand, and downregulating catalase and xCT to prevent H2O2 degradation on the other hand. H2O2 then contributed to brucine-triggered iron increase and transferrin receptor upregulation, as well as lipid peroxidation. This was further verified by treating glioma cells with exogenous H2O2 alone. Moreover, H2O2 reversely exacerbated brucine-induced ER stress. Taken together, ATF3 contributes to brucine-induced glioma cell ferroptosis via increasing H2O2 and iron.


Asunto(s)
Factor de Transcripción Activador 3/metabolismo , Antineoplásicos/uso terapéutico , Ferroptosis/efectos de los fármacos , Peróxido de Hidrógeno/metabolismo , Hierro/metabolismo , Estricnina/análogos & derivados , Sistema de Transporte de Aminoácidos y+/metabolismo , Animales , Antineoplásicos/farmacología , Catalasa/metabolismo , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , NADPH Oxidasa 4/metabolismo , Neoplasias/tratamiento farmacológico , Estricnina/farmacología , Estricnina/uso terapéutico , Superóxido Dismutasa-1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Acta Pharmacol Sin ; 42(8): 1324-1337, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33879840

RESUMEN

FOXO3a (forkhead box transcription factor 3a) is involved in regulating multiple biological processes in cancer cells. BNIP3 (Bcl-2/adenovirus E1B 19-kDa-interacting protein 3) is a receptor accounting for priming damaged mitochondria for autophagic removal. In this study we investigated the role of FOXO3a in regulating the sensitivity of glioma cells to temozolomide (TMZ) and its relationship with BNIP3-mediated mitophagy. We showed that TMZ dosage-dependently inhibited the viability of human U87, U251, T98G, LN18 and rat C6 glioma cells with IC50 values of 135.75, 128.26, 142.65, 155.73 and 111.60 µM, respectively. In U87 and U251 cells, TMZ (200 µM) induced DNA double strand breaks (DSBs) and nuclear translocation of apoptosis inducing factor (AIF), which was accompanied by BNIP3-mediated mitophagy and FOXO3a accumulation in nucleus. TMZ treatment induced intracellular ROS accumulation in U87 and U251 cells via enhancing mitochondrial superoxide, which not only contributed to DNA DSBs and exacerbated mitochondrial dysfunction, but also upregulated FOXO3a expression. Knockdown of FOXO3a aggravated TMZ-induced DNA DSBs and mitochondrial damage, as well as glioma cell death. TMZ treatment not only upregulated BNIP3 and activated autophagy, but also triggered mitophagy by prompting BNIP3 translocation to mitochondria and reinforcing BNIP3 interaction with LC3BII. Inhibition of mitophagy by knocking down BNIP3 with SiRNA or blocking autophagy with 3MA or bafilomycin A1 exacerbated mitochondrial superoxide and intracellular ROS accumulation. Moreover, FOXO3a knockdown inhibited TMZ-induced BNIP3 upregulation and autophagy activation. In addition, we showed that treatment with TMZ (100 mg·kg-1·d-1, ip) for 12 days in C6 cell xenograft mice markedly inhibited tumor growth accompanied by inducing FOXO3a upregulation, oxidative stress and BNIP3-mediated mitophagy in tumor tissues. These results demonstrate that FOXO3a attenuates temozolomide-induced DNA double strand breaks in human glioma cells via promoting BNIP3-mediated mitophagy.


Asunto(s)
Antineoplásicos/uso terapéutico , Roturas del ADN de Doble Cadena/efectos de los fármacos , Proteína Forkhead Box O3/metabolismo , Glioma/metabolismo , Mitofagia/efectos de los fármacos , Temozolomida/uso terapéutico , Animales , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Glioma/tratamiento farmacológico , Humanos , Proteínas de la Membrana/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Regulación hacia Arriba/efectos de los fármacos
15.
Neuroscience ; 449: 241-250, 2020 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-33039521

RESUMEN

Parthanatos is a modality of regulated cell death initiated by poly(ADP-ribose) polymerase 1 (PARP-1) hyperactivation and characterized by apoptosis inducing factor (AIF)-dependent and microphage migration inhibitory factor (MIF)-dependent DNA degradation. It is a caspase-independent, mitochondrial-linked paradigm of cell death and has been demonstrated to be related to the pathogenesis of various nervous system diseases. An in-depth understanding of the role that parthanatos plays in the pathological processes of these diseases can provide new targets for nervous system diseases treatments. In this review, on the basis of parthanatos mechanism, the involvement of parthanatos in the pathogenesis of nervous system diseases including neurodegenerative disorders, cerebrovascular diseases, spinal cord injury and glioma will be summarized in detail.


Asunto(s)
Glioma , Parthanatos , Apoptosis , Factor Inductor de la Apoptosis/metabolismo , Muerte Celular , Humanos , Mitocondrias/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo
16.
Cell Death Dis ; 11(8): 630, 2020 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-32801360

RESUMEN

Induction of lethal autophagy has become a strategy to eliminate glioma cells, but it remains elusive whether autophagy contributes to cell death via causing mitochondria damage and nuclear translocation of apoptosis inducing factor (AIF). In this study, we find that silibinin induces AIF translocation from mitochondria to nuclei in glioma cells in vitro and in vivo, which is accompanied with autophagy activation. In vitro studies reveal that blocking autophagy with 3MA, bafilomycin A1 or by knocking down ATG5 with SiRNA inhibits silibinin-induced mitochondrial accumulation of superoxide, AIF translocation from mitochondria to nuclei and glioma cell death. Mechanistically, silibinin activates autophagy through depleting ATP by suppressing glycolysis. Then, autophagy improves intracellular H2O2 via promoting p53-mediated depletion of GSH and cysteine and downregulation of xCT. The increased H2O2 promotes silibinin-induced BNIP3 upregulation and translocation to mitochondria. Knockdown of BNIP3 with SiRNA inhibits silibinin-induced mitochondrial depolarization, accumulation of mitochondrial superoxide, and AIF translocation from mitochondria to nuclei, as well as prevents glioma cell death. Furthermore, we find that the improved H2O2 reinforces silibinin-induced glycolysis dysfunction. Collectively, autophagy contributes to silibinin-induced glioma cell death via promotion of oxidative stress-mediated BNIP3-dependent nuclear translocation of AIF.


Asunto(s)
Factor Inductor de la Apoptosis/metabolismo , Autofagia/efectos de los fármacos , Núcleo Celular/metabolismo , Glioma/metabolismo , Glioma/patología , Proteínas de la Membrana/metabolismo , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas/metabolismo , Silibina/farmacología , Animales , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Glutatión/metabolismo , Glucólisis/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Modelos Biológicos , Fosforilación/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Silibina/química , Proteína p53 Supresora de Tumor/metabolismo
17.
Oncol Lett ; 20(4): 57, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32793311

RESUMEN

Erastin is a small molecular compound that induces ferroptosis by binding to voltage-dependent anion-selective channel protein (VDAC)2, VDAC3 and solute carrier family 7 member 5 inhibiting the cystine/glutamate antiporter. However, to the best of our knowledge, the mechanism of erastin-induced breast cancer cell death remains unclear. In present study aimed to explore the underlying mechanisms of the antitumor effects of erastin on breast cancer cells. Cellular viability was assessed using an MTT assay, a lactate dehydrogenase cytotoxicity assay kit was used to determine the cell death rate, the intracellular Fe2+ levels were determined using an iron colorimetric assay kit and western blotting was used to estimate the changes of autophagy-associated proteins levels. The present study demonstrated that erastin inhibited the viability of breast cancer cells and induced breast cancer cell death in a dose-dependent manner. Additionally, autophagy was activated by erastin, as demonstrated by upregulated expression levels of autophagy-associated proteins in breast cancer cells. Bafilomycin A1, 3-methyladenine and knockdown of autophagy related (ATG)5 with small interfering RNA prevented erastin-induced breast cancer cell death and inhibited the erastin-induced changes in the expression levels of the autophagy-associated proteins beclin1, ATG5, ATG12, microtubule-associated proteins 1A/1B light chain 3B (LC3B) and P62. Furthermore, erastin-induced breast cancer cell death was inhibited by an iron chelator, deferoxamine, which inhibited the increases of erastin-induced iron levels and inhibited the erastin-induced changes in the expression levels of the autophagy-related proteins beclin1, ATG5, ATG12, LC3B and P62. In summary, erastin triggered autophagic death in breast cancer cells by increasing intracellular iron levels.

19.
Cancer Lett ; 467: 58-71, 2019 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-31560934

RESUMEN

Chromatinolysis refers to enzymatic degradation of nuclear DNA and is regarded as one of the crucial events leading to cell death. Mixed-lineage kinase domain-like protein (MLKL) has been identified as a key executor of necroptosis, but it remains unclear whether MLKL contributes to necroptosis via regulation of chromatinolysis. In this study, we find that shikonin induces MLKL activation and chromatinolysis in glioma cells in vitro and in vivo, which are accompanied with nuclear translocation of AIF and γ-H2AX formation. In vitro studies reveal that inhibition of MLKL with its specific inhibitor NSA or knockdown of MLKL with siRNA abrogates shikonin-induced glioma cell necroptosis, as well as chromatinolysis. Mechanistically, activated MLKL targets mitochondria and triggers excessive generation of mitochondrial superoxide, which promotes AIF translocation into nucleus via causing mitochondrial depolarization and aggravates γ-H2AX formation via improving intracellular accumulation of ROS. Inhibition of nuclear level of AIF by knockdown of AIF with siRNA or mitigation of γ-H2AX formation by suppressing ROS with antioxidant NAC effectively prevents shikonin-induced chromatinolysis. Then, we found that RIP3 accounts for shikonin-induced activation of MLKL, and activated MLKL reversely up-regulates the protein level of CYLD and promotes the activation of RIP1 and RIP3. Taken together, our data suggest that MLKL contributes to shikonin-induced glioma cell necroptosis via promotion of chromatinolysis, and shikonin induces a positive feedback between MLKL and its upstream signals RIP1 and RIP3.


Asunto(s)
ADN de Neoplasias/química , Glioma/tratamiento farmacológico , Naftoquinonas/administración & dosificación , Proteínas Quinasas/metabolismo , Animales , Factor Inductor de la Apoptosis/metabolismo , Línea Celular Tumoral , Fragmentación del ADN , ADN de Neoplasias/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/metabolismo , Humanos , Ratones , Mitocondrias/metabolismo , Naftoquinonas/farmacología , Necroptosis , Ratas , Especies Reactivas de Oxígeno/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Biochem Biophys Res Commun ; 518(3): 590-597, 2019 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-31445705

RESUMEN

RSL3 is a type of small molecular compound which can inactivate glutathione peroxidase 4 (GPX4) and induce ferroptosis, but its role in glioma cell death remains unclear. In this study, we found RSL3 inhibited the viabilities of glioma cells and induced glioma cell death in a dose-dependent manner. In vitro studies revealed that RSL3-induced cell death was accompanied with the changes of autophagy-associated protein levels and was alleviated by pretreatment of 3-Methyladenine, bafilomycin A1 and knockdown of ATG5 with siRNA. The ATP and pyruvate content as well as the protein levels of HKII, PFKP, PKM2 were decreased in cells treated by RSL3, indicating that RSL3 induced glycolysis dysfunction in glioma cells. Moreover, supplement of exterior sodium pyruvate, which was a final product of glycolysis, not only inhibited the changes of autophagy-associated protein levels caused by RSL3, but also prevented RSL3-induced cell death. In vivo data suggested that the inhibitory effect of RSL3 on the growth of glioma cells was associated with glycolysis dysfunction and autophagy activation. Taken together, RSL3 induced autophagic cell death in glioma cells via causing glycolysis dysfunction.


Asunto(s)
Antineoplásicos/farmacología , Muerte Celular Autofágica/efectos de los fármacos , Carbolinas/farmacología , Glioma/tratamiento farmacológico , Glucólisis/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Carbolinas/uso terapéutico , Línea Celular Tumoral , Inhibidores Enzimáticos/farmacología , Glioma/metabolismo , Glioma/patología , Glutatión Peroxidasa/antagonistas & inhibidores , Glutatión Peroxidasa/metabolismo , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA