Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
Intervalo de año de publicación
1.
Biomed Res Int ; 2021: 8880179, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33532500

RESUMEN

Regulated necrosis (necroptosis) is crucially involved in cardiac ischaemia-reperfusion injury (MIRI). The aim of our study is to investigate whether shock wave therapy (SWT) is capable of exerting protective effects by inhibiting necroptosis during myocardial ischaemia-reperfusion (I/R) injury and the possible role of autophagy in this process. We established a hypoxia/reoxygenation (H/R) model in vitro using HL-1 cells to simulate MIRI. MTS assays and LDH cytotoxicity assay were performed to measure cell viability and cell damage. Annexin V/PI staining was used to determine apoptosis and necrosis. Western blotting was performed to assess the changes in cell signaling pathways associated with autophagy, necroptosis, and apoptosis. Reactive oxygen species (ROS) production was detected using DHE staining. Autophagosome generation and degradation (autophagic flux) were analysed using GFP and RFP tandemly tagged LC3 (tfLC3). HL-1 cells were then transfected with p62/SQSTM1 siRNA in order to analyse its role in cardioprotection. Our results revealed that SWT increased cell viability in the H/R model and decreased receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and RIPK3 expression. ROS production was also inhibited by SWT. Moreover, SWT decreased Beclin1 expression and the ratio of LC3-II/LC3-I following H/R. Simultaneously, in the tfLC3 assay, the SWT provoked a decrease in the cumulative autophagosome abundance. siRNA-mediated knockdown of p62 attenuated H/R-induced necroptosis, and SWT did not exert additive effects. Taken together, SWT ameliorated H/R injury by inhibiting necroptosis. SWT also relieved the blockade of autophagic flux in response to H/R injury. The restoration of autophagic flux by SWT might contribute to its cardioprotective effect on necroptosis following H/R injury.


Asunto(s)
Autofagia/efectos de la radiación , Hipoxia de la Célula/efectos de la radiación , Tratamiento con Ondas de Choque Extracorpóreas , Miocitos Cardíacos , Necroptosis/efectos de la radiación , Animales , Línea Celular , Supervivencia Celular/efectos de la radiación , Corazón/efectos de la radiación , Ratones , Modelos Biológicos , Daño por Reperfusión Miocárdica , Miocardio/citología , Miocitos Cardíacos/citología , Miocitos Cardíacos/efectos de la radiación
2.
Front Immunol ; 12: 705361, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34489957

RESUMEN

Immunogenic cell death (ICD) is a form of regulated cell death (RCD) induced by various stresses and produces antitumor immunity via damage-associated molecular patterns (DAMPs) release or exposure, mainly including high mobility group box 1 (HMGB1), calreticulin (CRT), adenosine triphosphate (ATP), and heat shock proteins (HSPs). Emerging evidence has suggested that ionizing radiation (IR) can induce ICD, and the dose, type, and fractionation of irradiation influence the induction of ICD. At present, IR-induced ICD is mainly verified in vitro in mice and there is few clinical evidence about it. To boost the induction of ICD by IR, some strategies have shown synergy with IR to enhance antitumor immune response, such as hyperthermia, nanoparticles, and chemotherapy. In this review, we focus on the molecular mechanisms of ICD, ICD-promoting factors associated with irradiation, the clinical evidence of ICD, and immunogenic forms of cell death. Finally, we summarize various methods of improving ICD induced by IR.


Asunto(s)
Muerte Celular Inmunogénica/efectos de la radiación , Alarminas/fisiología , Animales , Antígenos de Neoplasias/inmunología , Biomarcadores , Terapia Combinada , Citocinas/fisiología , Relación Dosis-Respuesta en la Radiación , Ferroptosis/efectos de la radiación , Proteína HMGB1/fisiología , Humanos , Hipertermia Inducida , Ratones , Morfolinas/uso terapéutico , Necroptosis/efectos de la radiación , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Neoplasias/radioterapia , Piperazinas/uso terapéutico , Pirroles/uso terapéutico , Tolerancia a Radiación , Radiación Ionizante
3.
Sci Rep ; 10(1): 18946, 2020 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-33144600

RESUMEN

Ultraviolet radiation (UVR) is a major environmental genotoxic agent. In skin, it can lead to the formation of mutagenic DNA damage. Several mechanisms are in place to prevent the conversion of these DNA damage into skin cancer-driver mutations. An important mutation prevention mechanism is the programmed cell death, which can safely dispose of the damaged cells. Apoptosis is the most studied and best characterised programmed cell death, but an increasing amount of new cell death pathways are emerging. Using different pharmacological cell death inhibitors and antioxidants, we have evaluated the implication of apoptosis, necroptosis, ferroptosis and parthanatos in UVB-induced cell death in human diploid dermal fibroblasts. Our results show that apoptosis is the only known cell death mechanism induced by UVB irradiation in fibroblasts. We also showed that lethal UVB irradiation induces a PARP-dependent drastic loss of cellular metabolic activity caused by an overused of NAD+.


Asunto(s)
Apoptosis/efectos de la radiación , Muerte Celular/efectos de la radiación , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Rayos Ultravioleta , Daño del ADN/efectos de la radiación , Ferroptosis/efectos de la radiación , Humanos , Necroptosis/efectos de la radiación , Parthanatos/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de la radiación
4.
Radiat Res ; 193(5): 435-450, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32134361

RESUMEN

Mitigation of total-body irradiation (TBI) in C57BL/6 mice by two drugs, which target apoptosis and necroptosis respectively, increases survival compared to one drug alone. Here we investigated whether the biomarker (signature)directed addition of a third anti-ferroptosis drug further mitigated TBI effects. C57BL/6NTac female mice (30-33 g) received 9.25 Gy TBI, and 24 h or later received JP4-039 (20 mg/kg), necrostatin-1 (1.65 mg/kg) and/or lipoxygenase-15 inhibitor (baicalein) (50 mg/kg) in single-, dual- or three-drug regimens. Some animals were sacrificed at days 0, 1, 2, 3, 4 or 7 postirradiation, while the majority in each group were maintained beyond 30 days. For those mice sacrificed at the early time points, femur bone marrow, intestine (ileum), lung and blood plasma were collected and analyzed for radiation-induced and mitigator-modified levels of 33 pro-inflammatory and stress response proteins. Each single mitigator administered [JP4-039 (24 h), necrostatin-1 (48 h) or baicalein (24 h)] improved survival at day 30 after TBI to 25% (P = 0.0432, 0.2816 or 0.1120, respectively) compared to 5% survival of 9.25 Gy TBI controls. Mice were administered the drug individually based on weight (mg/kg). Drug vehicles comprised 30% cyclodextrin for JP4-039 and baicalein, and 10% Cremphor-EL/10% ethanol/80% water for necrostatin-1; thus, dual-vehicle controls were also tested. The dual-drug combinations further enhanced survival: necrostatin-1 (delayed to 72 h) with baicalein 40% (P = 0.0359); JP4-039 with necrostatin-1 50% (P = 0.0062); and JP4-039 with baicalein 60% (P = 0.0064). The three-drug regimen, timed to signature directed evidence of onset after TBI of each death pathway in marrow and intestine, further increased the 30-day survival to 75% (P = 0.0002), and there was optimal normalization to preirradiation levels of inflammatory cytokine and stress response protein levels in plasma, intestine and marrow. In contrast, lung protein levels were minimally altered by 9.25 Gy TBI or mitigators over 7 days. Significantly, elevated intestinal proteins at day 7 after TBI were reduced by necrostatin-1-containing regimens; however, normalization of plasma protein levels at day 7 required the addition of JP4-039 and baicalein. These findings indicate that mitigator targeting to three distinct cell death pathways increases survival after TBI.


Asunto(s)
Apoptosis/efectos de los fármacos , Ferroptosis/efectos de los fármacos , Necroptosis/efectos de los fármacos , Protectores contra Radiación/farmacología , Irradiación Corporal Total/efectos adversos , Animales , Apoptosis/efectos de la radiación , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Médula Ósea/efectos de la radiación , Citocinas/metabolismo , Interacciones Farmacológicas , Femenino , Ferroptosis/efectos de la radiación , Íleon/efectos de los fármacos , Íleon/metabolismo , Íleon/efectos de la radiación , Ratones , Ratones Endogámicos C57BL , Necroptosis/efectos de la radiación , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/prevención & control , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Tiempo
5.
Cell Death Dis ; 11(12): 1070, 2020 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-33318476

RESUMEN

Lack of effective treatments for aggressive breast cancer is still a major global health problem. We have previously reported that photodynamic therapy using methylene blue as photosensitizer (MB-PDT) massively kills metastatic human breast cancer, marginally affecting healthy cells. In this study, we aimed to unveil the molecular mechanisms behind MB-PDT effectiveness and specificity towards tumor cells. Through lipidomics and biochemical approaches, we demonstrated that MB-PDT efficiency and specificity rely on polyunsaturated fatty acid-enriched membranes and on the better capacity to deal with photo-oxidative damage displayed by non-tumorigenic cells. We found out that, in tumorigenic cells, lysosome membrane permeabilization is accompanied by ferroptosis and/or necroptosis. Our results also pointed at a cross-talk between lysosome-dependent cell death (LDCD) and necroptosis induction after photo-oxidation, and contributed to broaden the understanding of MB-PDT-induced mechanisms and specificity in breast cancer cells. Therefore, we demonstrated that efficient approaches could be designed on the basis of lipid composition and metabolic features for hard-to-treat cancers. The results further reinforce MB-PDT as a therapeutic strategy for highly aggressive human breast cancer cells.


Asunto(s)
Neoplasias de la Mama/patología , Luz , Antioxidantes/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Carcinogénesis/efectos de los fármacos , Carcinogénesis/patología , Carcinogénesis/efectos de la radiación , Muerte Celular/efectos de los fármacos , Muerte Celular/efectos de la radiación , Línea Celular Tumoral , Femenino , Ferroptosis/efectos de los fármacos , Ferroptosis/efectos de la radiación , Humanos , Lípidos/química , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/efectos de la radiación , Azul de Metileno/farmacología , Azul de Metileno/uso terapéutico , Modelos Biológicos , Necroptosis/efectos de los fármacos , Necroptosis/efectos de la radiación , Oxidación-Reducción , Fotoquimioterapia , Neoplasias de la Mama Triple Negativas/patología
6.
J Exp Clin Cancer Res ; 38(1): 461, 2019 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-31706322

RESUMEN

BACKGROUND: Tumor cell repopulation after radiotherapy is a major cause for the tumor radioresistance and recurrence. This study aims to investigate the underlying mechanism of tumor repopulation after radiotherapy, with focus on whether and how necroptosis takes part in this process. METHODS: Necroptosis after irradiation were examined in vitro and in vivo. And the growth-promoting effect of necroptotic cells was investigated by chemical inhibitors or shRNA against necroptosis associated proteins and genes in in vitro and in vivo tumor repopulation models. Downstream relevance factors of necroptosis were identified by western blot and chemiluminescent immunoassays. Finally, the immunohistochemistry staining of identified necroptosis association growth stimulation factor was conducted in human colorectal tumor specimens to verify the relationship with clinical outcome. RESULTS: Radiation-induced necroptosis depended on activation of RIP1/RIP3/MLKL pathway, and the evidence in vitro and in vivo demonstrated that the inhibition of necroptosis attenuated growth-stimulating effects of irradiated tumor cells on living tumor reporter cells. The JNK/IL-8 were identified as downstream molecules of pMLKL during necroptosis, and inhibition of JNK, IL-8 or IL-8 receptor significantly reduced tumor repopulation after radiotherapy. Moreover, the high expression of IL-8 was associated with poor clinical prognosis in colorectal cancer patients. CONCLUSIONS: Necroptosis associated tumor repopulation after radiotherapy depended on activation of RIP1/RIP3/MLKL/JNK/IL-8 pathway. This novel pathway provided new insight into understanding the mechanism of tumor radioresistance and repopulation, and MLKL/JNK/IL-8 could be developed as promising targets for blocking tumor repopulation to enhance the efficacy of colorectal cancer radiotherapy.


Asunto(s)
Interleucina-8/metabolismo , Necroptosis , Neoplasias/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Proteínas Quinasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Animales , Apoptosis , Línea Celular Tumoral , Supervivencia Celular , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Mediciones Luminiscentes , Ratones , Imagen Molecular , Necroptosis/efectos de la radiación , Neoplasias/genética , Neoplasias/radioterapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA