Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Protein Cell ; 2024 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-38430542

RESUMO

Ferroptosis has been recognized as a unique cell death modality driven by excessive lipid peroxidation and unbalanced cellular metabolism. In this study, we established a protein interaction landscape for ferroptosis pathways through proteomic analyses, and identified choline/ethanolamine phosphotransferase 1 (CEPT1) as a lysophosphatidylcholine acyltransferase 3 (LPCAT3)-interacting protein that regulates LPCAT3 protein stability. In contrast to its known role in promoting phospholipid synthesis, we showed that CEPT1 suppresses ferroptosis potentially by interacting with phospholipases and breaking down certain pro-ferroptotic polyunsaturated fatty acid (PUFA)-containing phospholipids. Together, our study reveals a previously unrecognized role of CEPT1 in suppressing ferroptosis.

2.
Cancer Discov ; 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38552003

RESUMO

Resistance to poly (ADP-ribose) polymerase inhibitors (PARPi) limits the therapeutic efficacy of PARP inhibition in treating breast cancer susceptibility gene 1 (BRCA1)-deficient cancers. Here we reveal that BRCA1 has a dual role in regulating ferroptosis. BRCA1 promotes the transcription of voltage-dependent anion channel 3 (VDAC3) and glutathione peroxidase 4 (GPX4); consequently, BRCA1 deficiency promotes cellular resistance to erastin-induced ferroptosis but sensitizes cancer cells to ferroptosis induced by GPX4 inhibitors (GPX4i). In addition, nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy and defective GPX4 induction unleash potent ferroptosis in BRCA1-deficient cancer cells upon PARPi and GPX4i co-treatment. Finally, we show that xenograft tumors derived from BRCA1-mutant breast cancer patients with PARPi resistance exhibit decreased GPX4 expression and high sensitivity to PARP and GPX4 co-inhibition. Our results show that BRCA1 deficiency induces a ferroptosis vulnerability to PARP and GPX4 co-inhibition and inform a therapeutic strategy for overcoming PARPi resistance in BRCA1-deficient cancers.

3.
PLoS Biol ; 21(11): e3002379, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37944100

RESUMO

[This corrects the article DOI: 10.1371/journal.pbio.3000324.].

5.
Nat Commun ; 14(1): 3673, 2023 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-37339981

RESUMO

The cystine transporter solute carrier family 7 member 11 (SLC7A11; also called xCT) protects cancer cells from oxidative stress and is overexpressed in many cancers. Here we report a surprising finding that, whereas moderate overexpression of SLC7A11 is beneficial for cancer cells treated with H2O2, a common oxidative stress inducer, its high overexpression dramatically increases H2O2-induced cell death. Mechanistically, high cystine uptake in cancer cells with high overexpression of SLC7A11 in combination with H2O2 treatment results in toxic buildup of intracellular cystine and other disulfide molecules, NADPH depletion, redox system collapse, and rapid cell death (likely disulfidptosis). We further show that high overexpression of SLC7A11 promotes tumor growth but suppresses tumor metastasis, likely because metastasizing cancer cells with high expression of SLC7A11 are particularly susceptible to oxidative stress. Our findings reveal that SLC7A11 expression level dictates cancer cells' sensitivity to oxidative stress and suggests a context-dependent role for SLC7A11 in tumor biology.


Assuntos
Cistina , Neoplasias , Cistina/metabolismo , Linhagem Celular Tumoral , Peróxido de Hidrogênio/farmacologia , Peróxido de Hidrogênio/metabolismo , Estresse Oxidativo , Dissulfetos/metabolismo , Sistema y+ de Transporte de Aminoácidos/genética , Sistema y+ de Transporte de Aminoácidos/metabolismo , Neoplasias/genética
6.
Nat Cell Biol ; 25(3): 404-414, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36747082

RESUMO

SLC7A11-mediated cystine uptake suppresses ferroptosis yet promotes cell death under glucose starvation; the nature of the latter cell death remains unknown. Here we show that aberrant accumulation of intracellular disulfides in SLC7A11high cells under glucose starvation induces a previously uncharacterized form of cell death distinct from apoptosis and ferroptosis. We term this cell death disulfidptosis. Chemical proteomics and cell biological analyses showed that glucose starvation in SLC7A11high cells induces aberrant disulfide bonds in actin cytoskeleton proteins and F-actin collapse in a SLC7A11-dependent manner. CRISPR screens and functional studies revealed that inactivation of the WAVE regulatory complex (which promotes actin polymerization and lamellipodia formation) suppresses disulfidptosis, whereas constitutive activation of Rac promotes disulfidptosis. We further show that glucose transporter inhibitors induce disulfidptosis in SLC7A11high cancer cells and suppress SLC7A11high tumour growth. Our results reveal that the susceptibility of the actin cytoskeleton to disulfide stress mediates disulfidptosis and suggest a therapeutic strategy to target disulfidptosis in cancer treatment.


Assuntos
Dissulfetos , Neoplasias , Humanos , Neoplasias/metabolismo , Apoptose , Citoesqueleto de Actina/metabolismo , Glucose/metabolismo
7.
Life Metab ; 2(6)2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38179338

RESUMO

The lack of a reliable and specific marker for ferroptosis has hindered the advancement of treatments related to this cell death mechanism toward clinical application. A recent study published in Molecular Cell has identified hyperoxidized peroxiredoxin 3 (PRDX3) as a promising marker for ferroptosis, opening up new avenues for monitoring and targeting ferroptosis in disease treatment.

8.
Water Sci Technol ; 86(3): 555-567, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35960836

RESUMO

Sulfidated nano zero-valent iron supported by activated carbon (S-nZVI/AC) composites were synthesized via liquid phase reduction method, and then they were used for Cr(VI) elimination. Characterization results showed that Fe0 was the main component, besides, iron oxides and iron sulfides were also detected. Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) results showed that S-nZVI nanoparticles were homogeneously distributed on the surfaces of AC. The influences of S/Fe ratio, C/Fe ratio, pH value, reaction temperature and co-existed ions (Cl-, SO42-, PO43- and NO3-) on Cr(VI) removal performances were investigated. Furthermore, the corresponding mechanisms were also discussed. The S-nZVI/AC composites exhibited good aging-resistance performances that Cr(VI) removal efficiency still maintained at 83.1% after being sealed in water for seven days, and they also had satisfying cycling stabilities that Cr(VI) removal efficiency only decreased less than 10% after four cycles. The good performances of S-nZVI/AC composites for Cr(VI) removal are attributed to the protection effect of iron sulfides and immobilization effect of AC, making S-nZVI/AC as a promising candidate for Cr(VI) elimination in effluents.


Assuntos
Poluentes Químicos da Água , Adsorção , Cromo/química , Ferro/química , Sulfetos , Poluentes Químicos da Água/química
9.
Nat Commun ; 13(1): 2206, 2022 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-35459868

RESUMO

Targeting ferroptosis, a unique cell death modality triggered by unrestricted lipid peroxidation, in cancer therapy is hindered by our incomplete understanding of ferroptosis mechanisms under specific cancer genetic contexts. KEAP1 (kelch-like ECH associated protein 1) is frequently mutated or inactivated in lung cancers, and KEAP1 mutant lung cancers are refractory to most therapies, including radiotherapy. In this study, we identify ferroptosis suppressor protein 1 (FSP1, also known as AIFM2) as a transcriptional target of nuclear factor erythroid 2-related factor 2 (NRF2) and reveal that the ubiquinone (CoQ)-FSP1 axis mediates ferroptosis- and radiation- resistance in KEAP1 deficient lung cancer cells. We further show that pharmacological inhibition of the CoQ-FSP1 axis sensitizes KEAP1 deficient lung cancer cells or patient-derived xenograft tumors to radiation through inducing ferroptosis. Together, our study identifies CoQ-FSP1 as a key downstream effector of KEAP1-NRF2 pathway and as a potential therapeutic target for treating KEAP1 mutant lung cancers.


Assuntos
Proteínas Reguladoras de Apoptose , Ferroptose , Neoplasias Pulmonares , Proteínas Mitocondriais , Ubiquinona , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Ferroptose/genética , Humanos , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Peroxidação de Lipídeos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/radioterapia , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Ubiquinona/genética , Ubiquinona/metabolismo
11.
Nature ; 593(7860): 586-590, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33981038

RESUMO

Ferroptosis, a form of regulated cell death that is induced by excessive lipid peroxidation, is a key tumour suppression mechanism1-4. Glutathione peroxidase 4 (GPX4)5,6 and ferroptosis suppressor protein 1 (FSP1)7,8 constitute two major ferroptosis defence systems. Here we show that treatment of cancer cells with GPX4 inhibitors results in acute depletion of N-carbamoyl-L-aspartate, a pyrimidine biosynthesis intermediate, with concomitant accumulation of uridine. Supplementation with dihydroorotate or orotate-the substrate and product of dihydroorotate dehydrogenase (DHODH)-attenuates or potentiates ferroptosis induced by inhibition of GPX4, respectively, and these effects are particularly pronounced in cancer cells with low expression of GPX4 (GPX4low). Inactivation of DHODH induces extensive mitochondrial lipid peroxidation and ferroptosis in GPX4low cancer cells, and synergizes with ferroptosis inducers to induce these effects in GPX4high cancer cells. Mechanistically, DHODH operates in parallel to mitochondrial GPX4 (but independently of cytosolic GPX4 or FSP1) to inhibit ferroptosis in the mitochondrial inner membrane by reducing ubiquinone to ubiquinol (a radical-trapping antioxidant with anti-ferroptosis activity). The DHODH inhibitor brequinar selectively suppresses GPX4low tumour growth by inducing ferroptosis, whereas combined treatment with brequinar and sulfasalazine, an FDA-approved drug with ferroptosis-inducing activity, synergistically induces ferroptosis and suppresses GPX4high tumour growth. Our results identify a DHODH-mediated ferroptosis defence mechanism in mitochondria and suggest a therapeutic strategy of targeting ferroptosis in cancer treatment.


Assuntos
Di-Hidro-Orotato Desidrogenase/metabolismo , Ferroptose , Mitocôndrias/metabolismo , Neoplasias/enzimologia , Animais , Compostos de Bifenilo/farmacologia , Linhagem Celular Tumoral , Di-Hidro-Orotato Desidrogenase/genética , Feminino , Deleção de Genes , Humanos , Peroxidação de Lipídeos , Metabolômica , Camundongos Nus , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/antagonistas & inibidores , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Mol Cell Oncol ; 8(3): 1919006, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34027051

RESUMO

Ferroptosis is a cell death mechanism triggered by lipid peroxidation. Our recent study linked cyst(e)ine availability with glutathione peroxidase 4 (GPX4) protein synthesis and ferroptosis mitigation via a Rag-mechanistic target of rapamycin complex 1 (mTORC1) axis, and proposed that co-targeting mTORC1 and ferroptosis is a promising strategy for cancer therapy.

13.
Protein Cell ; 12(11): 836-857, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-33891303

RESUMO

Ferroptosis, an iron-dependent form of regulated cell death driven by peroxidative damages of polyunsaturated-fatty-acid-containing phospholipids in cellular membranes, has recently been revealed to play an important role in radiotherapy-induced cell death and tumor suppression, and to mediate the synergy between radiotherapy and immunotherapy. In this review, we summarize known as well as putative mechanisms underlying the crosstalk between radiotherapy and ferroptosis, discuss the interactions between ferroptosis and other forms of regulated cell death induced by radiotherapy, and explore combination therapeutic strategies targeting ferroptosis in radiotherapy and immunotherapy. This review will provide important frameworks for future investigations of ferroptosis in cancer therapy.


Assuntos
Ferroptose/imunologia , Imunoterapia , Neoplasias , Radioterapia , Humanos , Neoplasias/imunologia , Neoplasias/terapia
14.
Oncogene ; 40(20): 3533-3547, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33927351

RESUMO

Ferroptosis, a form of regulated cell death triggered by lipid peroxidation, was recently identified as an important mechanism in radiotherapy (RT)-mediated tumor suppression and radioresistance, although the exact genetic contexts in which to target ferroptosis in RT remains to be defined. p53 is the most commonly mutated gene in human cancers and a major effector to RT. Here, we identify ferroptosis as a critical mechanism to mediate p53 function in tumor radiosensitivity. Mechanistically, RT-mediated p53 activation antagonizes RT-induced SLC7A11 expression and represses glutathione synthesis, thereby promoting RT-induced lipid peroxidation and ferroptosis. p53 deficiency promotes radioresistance in cancer cells or tumors at least partly through SLC7A11-mediated ferroptosis inhibition. Ferroptosis inducers (FINs) that inhibit SLC7A11 exert significant radiosensitizing effects in tumor organoids and patient-derived xenografts with p53 mutation or deficiency. Finally, we show that RT-induced ferroptosis correlates with p53 activation and better clinical outcomes to RT in cancer patients. Together, our study uncovers a previously unappreciated role of ferroptosis in p53-mediated radiosensitization and suggest using FINs in combination with RT to treat p53-mutant cancers.


Assuntos
Neoplasias/metabolismo , Neoplasias/radioterapia , Proteína Supressora de Tumor p53/metabolismo , Sistema y+ de Transporte de Aminoácidos/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Ferroptose , Humanos , Peroxidação de Lipídeos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias/patologia , Tolerância a Radiação , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...