Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Basis Dis ; 1868(1): 166287, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34626772

RESUMO

Oxidative stress and lipid peroxidation are major causes of skin injury induced by ultraviolet (UV) irradiation. Ferroptosis is a form of regulated necrosis driven by iron-dependent peroxidation of phospholipids and contributes to kinds of tissue injuries. However, it remains unclear whether the accumulation of lipid peroxides in UV irradiation-induced skin injury could lead to ferroptosis. We generated UV irradiation-induced skin injury mice model to examine the accumulation of the lipid peroxides and iron. Lipid peroxides 4-HNE, the oxidative enzyme COX2, the oxidative DNA damage biomarker 8-OHdG, and the iron level were increased in UV irradiation-induced skin. The accumulation of iron and lipid peroxidation was also observed in UVB-irradiated epidermal keratinocytes without actual ongoing ferroptotic cell death. Ferroptosis was triggered in UV-irradiated keratinocytes stimulated with ferric ammonium citrate (FAC) to mimic the iron overload. Although GPX4 protected UVB-injured keratinocytes against ferroptotic cell death resulted from dysregulation of iron metabolism and the subsequent increase of lipid ROS, keratinocytes enduring constant UVB treatment were markedly sensitized to ferroptosis. Nicotinamide mononucleotide (NMN) which is a direct and potent NAD+ precursor supplement, rescued the imbalanced NAD+/NADH ratio, recruited the production of GSH and promoted resistance to lipid peroxidation in a GPX4-dependent manner. Taken together, our data suggest that NMN recruits GSH to enhance GPX4-mediated ferroptosis defense in UV irradiation-induced skin injury and inhibits oxidative skin damage. NMN or ferroptosis inhibitor might become promising therapeutic approaches for treating oxidative stress-induced skin diseases or disorders.


Assuntos
Glutationa/genética , Ferro/metabolismo , Estresse Oxidativo/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Pele/metabolismo , 8-Hidroxi-2'-Desoxiguanosina/farmacologia , Aldeídos/farmacologia , Animais , Ciclo-Oxigenase 2/genética , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Compostos Férricos/farmacologia , Ferroptose/efeitos dos fármacos , Ferroptose/efeitos da radiação , Humanos , Queratinócitos/efeitos dos fármacos , Queratinócitos/efeitos da radiação , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos da radiação , Peróxidos Lipídicos/farmacologia , Camundongos , Mononucleotídeo de Nicotinamida/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/efeitos da radiação , Compostos de Amônio Quaternário/farmacologia , Pele/efeitos dos fármacos , Pele/lesões , Pele/patologia , Raios Ultravioleta/efeitos adversos
2.
Front Immunol ; 12: 705361, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34489957

RESUMO

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.


Assuntos
Morte Celular Imunogênica/efeitos da radiação , Alarminas/fisiologia , Animais , Antígenos de Neoplasias/imunologia , Biomarcadores , Terapia Combinada , Citocinas/fisiologia , Relação Dose-Resposta à Radiação , Ferroptose/efeitos da radiação , Proteína HMGB1/fisiologia , Humanos , Hipertermia Induzida , Camundongos , Morfolinas/uso terapêutico , Necroptose/efeitos da radiação , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/radioterapia , Piperazinas/uso terapêutico , Pirróis/uso terapêutico , Tolerância a Radiação , Radiação Ionizante
3.
Int J Radiat Biol ; 97(4): 464-473, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33464146

RESUMO

PURPOSE: Baicalein (an anti-ferroptosis drug) was recently reported to synergistically improve the survival rate of mice following a high dose of total body irradiation with anti-apoptosis and anti-necroptosis drugs. At the same time, our group has demonstrated that ferrostatin-1, a ferroptosis inhibitor, improves the survival rate of a mouse model of hematopoietic acute radiation syndrome to 60% for 150 days (p < .001). These phenomena suggest that ferroptosis inhibition can mitigate radiation damage. In this study, we continued to study the mechanisms by which ferrostatin-1 alleviated radiation-induced ferroptosis and subsequent hematopoietic acute radiation syndrome. MATERIALS AND METHODS: Male ICR mice (8-10 weeks old) were exposed to doses of 0, 8, or 10 Gy irradiated from a 137Cs source. Ferrostatin-1 was intraperitoneally injected into mice 72 h post-irradiation. Bone marrow mononuclear cells (BMMCs) and peripheral blood cells were counted. The changes in iron-related parameters, lipid metabolic enzymes, lipid peroxidation repair molecules (glutathione peroxidase 4, glutathione, and coenzyme Q10), and inflammatory factors (TNF-α, IL-6, and IL-1ß) were evaluated using biochemical or antibody techniques. RESULTS: Ferrostatin-1 increased the number of red and white blood cells, lymphocytes, and monocytes in the peripheral blood after total body irradiation in mice by mitigating the ferroptosis of BMMCs. Total body irradiation induced ferroptosis in BMMCs by increasing the iron and lipid peroxidation levels and depleting the acyl-CoA synthetase long-chain family member 4 (ASCL4), lipoxygenase 15, glutathione peroxidase 4, and glutathione levels. Ferroptotic BMMCs did not release TNF-α, IL-6, or IL-1ß at the early stage of radiation exposure. Ferrostatin-1 mitigated the lipid peroxidation of radiation-induced ferroptosis by attenuating increases in levels of hemosiderin and liable iron pool and decreases in levels of ASCL4 and glutathione peroxidase 4. CONCLUSIONS: The onset of total body irradiation-induced ferroptosis in BMMCs involved changes in iron, lipid metabolic enzymes, and anti-lipid peroxidation molecules. Ferrostatin-1 could be a potential radiation mitigation agent by acting on these targets.


Assuntos
Síndrome Aguda da Radiação/patologia , Cicloexilaminas/farmacologia , Hematopoese/efeitos dos fármacos , Fenilenodiaminas/farmacologia , Animais , Ferroptose/efeitos dos fármacos , Ferroptose/efeitos da radiação , Hematopoese/efeitos da radiação , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos da radiação , Masculino , Camundongos , Camundongos Endogâmicos ICR
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA