Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 24
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
J Agric Food Chem ; 72(31): 17356-17367, 2024 Aug 07.
Article de Anglais | MEDLINE | ID: mdl-39042602

RÉSUMÉ

Inhibition of oxidative stress and ferroptosis is currently considered to be a promising therapeutic approach for neurodegenerative diseases. Herpotrichones, a class of compounds derived from insect symbionts, have shown potential for neuroprotective activity with low toxicity. However, the specific mechanisms through which herpotrichones exert their neuroprotective effects remain to be fully elucidated. In this study, the natural [4 + 2] adducts herpotrichone A (He-A) and its new analogues were isolated from the isopod-associated fungus Herpotrichia sp. SF09 and exhibited significantly protective effects in H2O2-, 6-OHDA-, and RSL3-stimulated PC12 cells and LPS-stimulated BV-2 cells. Moreover, He-A was able to relieve ferroptotic cell death in RSL3-stimulated PC12 cells and 6-OHDA-induced zebrafish larvae. Interestingly, He-A can activate antioxidant elements and modulate the SLC7A11 pathway without capturing oxidic free radical and chelating iron. These findings highlight He-A as a novel hit that protects against ferroptosis-like neuronal damage in the treatment of neurodegenerative diseases.


Sujet(s)
Ferroptose , Neuroprotecteurs , Stress oxydatif , Danio zébré , Animaux , Ferroptose/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/composition chimique , Rats , Stress oxydatif/effets des médicaments et des substances chimiques , Cellules PC12 , Isopoda/effets des médicaments et des substances chimiques , Isopoda/composition chimique , Humains , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Souris , Maladies neurodégénératives/traitement médicamenteux , Maladies neurodégénératives/métabolisme , Larve/effets des médicaments et des substances chimiques , Larve/croissance et développement
2.
Chemosphere ; 359: 142252, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38735493

RÉSUMÉ

Organophosphates, a widely used group of pesticides, can cause severe toxicity in human beings and other non-target organisms. Liver, being the primary site for xenobiotic metabolism, is extremely vulnerable to xenobiotic-induced toxicity. Considering the numerous vital functions performed by the liver, including xenobiotic detoxification, protecting this organ from the ubiquitous pesticides in our food and environment is essential for maintaining homeostasis. In this study, we have investigated the impact of the organophosphate pesticide, Chlorpyrifos (CPF), on zebrafish liver at a concentration (300 µg/L) which is environmentally realistic. We have also demonstrated the role of dietary supplementation of α-tocopherol or Vitamin E (Vit E) (500 mg/kg feed) in mitigating pesticide-induced liver toxicity. Mechanistically, we showed that Vit E resulted in significant elevation of the Nrf2 and its downstream antioxidant enzyme activities and gene expressions, especially that of GST and GPx, resulting in reduction of CPF-induced intracellular lipid ROS and hepatic LPO. Further interrogation, such as analysis of GSH: GSSG ratio, intracellular iron concentration, iron metabolizing genes, mitochondrial dysfunction etc. revealed that CPF induces ferroptosis which can be reversed by Vit E supplementation. Ultimately, reduced concentration of CPF in zebrafish serum and flesh highlighted the role of Vit E in ameliorating CPF toxicity.


Sujet(s)
Chlorpyriphos , Ferroptose , Glutathion , Hépatocytes , Fer , Peroxydation lipidique , Vitamine E , Danio zébré , Animaux , Chlorpyriphos/toxicité , Vitamine E/pharmacologie , Vitamine E/métabolisme , Fer/métabolisme , Peroxydation lipidique/effets des médicaments et des substances chimiques , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Glutathion/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Antioxydants/métabolisme , Facteur-2 apparenté à NF-E2/métabolisme
3.
Heliyon ; 10(4): e26014, 2024 Feb 29.
Article de Anglais | MEDLINE | ID: mdl-38434050

RÉSUMÉ

Neuroblastoma, predominantly afflicting young individuals, is characterized as an embryonal tumor, with poor prognosis primarily attributed to chemoresistance. This study delved into the impact of tripartite motif (TRIM) 59, an E3 ligase, on neuroblastoma development and chemosensitivity through mediating ferroptosis and the involvement of the tumor suppressor p53. Clinical samples were assessed for TRIM59 and p53 levels to explore their correlation with neuroblastoma differentiation. In neuroblastoma cells, modulation of TRIM59 expression, either through overexpression or knockdown, was coupled with doxorubicin hydrochloride (DOX) or ferrostatin-1 (Fer-1) therapy. In vivo assessments examined the influence of TRIM59 knockdown on neuroblastoma chemosensitivity to DOX. Co-immunoprecipitation and ubiquitination assays investigated the association between TRIM59 and p53. Proliferation was gauged with Cell Counting Kit-8, lipid reactive oxygen species (ROS) were assessed via flow cytometry, and protein levels were determined by Western blotting. TRIM59 expression was inversely correlated with neuroblastoma differentiation and positively linked to cell proliferation in response to DOX. Moreover, TRIM59 impeded lipid ROS generation and ferroptosis by directly interacting with p53, promoting its ubiquitination and degradation in DOX-exposed neuroblastoma cells. Fer-1 countered the impact of TRIM59 knockdown on neuroblastoma, while TRIM59 knockdown enhanced the therapeutic efficacy of DOX in xenograph mice. This study underscores TRIM59 as an oncogene in neuroblastoma, fostering growth and chemoresistance by suppressing ferroptosis through p53 ubiquitination and degradation. TRIM59 emerges as a potential strategy for neuroblastoma therapy.

4.
Eur J Med Chem ; 268: 116250, 2024 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-38417218

RÉSUMÉ

Ferritinophagy is a cellular process to release redox-active iron. Excessive activation of ferritinophagy ultimately results in ferroptosis characterized by ROS accumulation which plays important roles in the development and progression of cancer. Sinomenine, a main bioactive alkaloid from the traditional Chinese medicine Sinomenum acutum, inhibits the proliferation of cancer cells by promoting ROS production. Herein, new compounds were designed and synthesized through the stepwise optimization of sinomenine. Among them, D3-3 induced the production of lipid ROS, and significantly promoted colorectal cancer cells to release the ferrous ion in an autophagy-dependent manner. Moreover, D3-3 enhanced the interaction of FTH1-NCOA4, indicating the activation of ferritinophagy. In vivo experiments showed that D3-3 restrained tumor growth and promoted lipid peroxidation in the HCT-116 xenograft model. These findings demonstrated that D3-3 is an inducer of ferritinophagy, eventually triggering ferroptosis. Compound D3-3, as the first molecule to be definitively demonstrated to induce ferritinophagy, is worth further evaluation as a promising drug candidate in the treatment of colorectal cancer.


Sujet(s)
Tumeurs colorectales , Ferritines , Morphinanes , Humains , Espèces réactives de l'oxygène/métabolisme , Fer/métabolisme , Autophagie , Tumeurs colorectales/traitement médicamenteux
5.
Cells ; 12(20)2023 10 23.
Article de Anglais | MEDLINE | ID: mdl-37887353

RÉSUMÉ

Pancreatic cancer is characterized by a poor prognosis, with its five-year survival rate lower than that of any other cancer type. Gemcitabine, a standard treatment for pancreatic cancer, often has poor outcomes for patients as a result of chemoresistance. Therefore, novel therapeutic targets must be identified to overcome gemcitabine resistance. Here, we found that SLC38A5, a glutamine transporter, is more highly overexpressed in gemcitabine-resistant patients than in gemcitabine-sensitive patients. Furthermore, the deletion of SLC38A5 decreased the proliferation and migration of gemcitabine-resistant PDAC cells. We also found that the inhibition of SLC38A5 triggered the ferroptosis signaling pathway via RNA sequencing. Also, silencing SLC38A5 induced mitochondrial dysfunction and reduced glutamine uptake and glutathione (GSH) levels, and downregulated the expressions of GSH-related genes NRF2 and GPX4. The blockade of glutamine uptake negatively modulated the mTOR-SREBP1-SCD1 signaling pathway. Therefore, suppression of SLC38A5 triggers ferroptosis via two pathways that regulate lipid ROS levels. Similarly, we observed that knockdown of SLC38A5 restored gemcitabine sensitivity by hindering tumor growth and metastasis in the orthotopic mouse model. Altogether, our results demonstrate that SLC38A5 could be a novel target to overcome gemcitabine resistance in PDAC therapy.


Sujet(s)
Systèmes de transport d'acides aminés neutres , Ferroptose , Tumeurs du pancréas , Animaux , Souris , Humains , , Désoxycytidine/pharmacologie , Désoxycytidine/usage thérapeutique , Glutamine , Résistance aux médicaments antinéoplasiques , Lignée cellulaire tumorale , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas
6.
Eur J Med Chem ; 261: 115829, 2023 Dec 05.
Article de Anglais | MEDLINE | ID: mdl-37801824

RÉSUMÉ

Glutathione peroxidase 4 (GPX4) is an essential antioxidant enzyme that negatively regulates ferroptosis. To exploit novel GPX4 inhibitors, we designed and synthesized 32 indirubin derivatives. Compound 31 exhibited the strongest antitumor activity against HCT-116 cells (IC50 = 0.49 ± 0.02 µM). Further studies suggested that 31 could induce ferroptosis in colon cancer cells and its cytotoxic activity could be reversed by ferroptosis inhibitors. Mechanism research showed that 31 promoted the degradation of GPX4, causing the accumulation of lipid ROS to induce ferroptosis. Animal experiments also proved that 31 could inhibit the growth of colon cancer cells in vivo and reduce the expression of GPX4 in tumor tissues. These results indicated that compound 31 had potential as a novel ferroptosis inducer agent for colon cancer.


Sujet(s)
Tumeurs du côlon , Ferroptose , Animaux , Glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Tumeurs du côlon/traitement médicamenteux
7.
Cell Biol Toxicol ; 39(2): 467-481, 2023 04.
Article de Anglais | MEDLINE | ID: mdl-34978008

RÉSUMÉ

Exosomal miRNAs activates hepatic stellate cell (HSC) and promote fibrosis. miR-222 was found to be increased in hepatitis B virus (HBV)-infected hepatocytes, and ferroptosis was reported to ameliorate liver fibrosis (LF). Although miR-222 and ferroptosis have been implicated in LF, the association between miR-222 and ferroptosis and how they coordinate to regulate LF are still not explicit. This study investigates the roles of miR-222 and transferrin receptor (TFRC) in LF. Lipid reactive oxygen species (ROS) level was analyzed by flow cytometry. FerroOrange staining was used to measure intracellular iron level. Luciferase reporter assay was adopted to confirm the binding of miR-222 and TFRC. Real-time quantitative PCR and immunoblots were applied to analyze gene and protein expression. The results showed that supplementation of exosomes derived from HBV-infected LO2 cells remarkably enhanced LX-2 cell activation, evidenced by elevated hydroxyprolin (Hyp) secretion and α-SMA and COL1A2 expression. miR-222 was significantly increased in HBV-Exo. Overexpressing miR-222 upregulated cell viability, secretion of Hpy, and expression of α-SMA and COL1A2, which were all blocked by overexpression of TFRC. Further study showed that TFRC was a target of miR-222, and miR-222 promoted LX-2 cell activation through suppressing TFRC-induced ferroptosis in LX-2 cells. Exosomal miR-222 derived from HBV-infected hepatocytes promoted LF through inhibiting TFRC and TFRC-induced ferroptosis. This study emphasizes the significance of miR-222/TFRC axis in LF and suggests new insights in clinical decision making while treating LF. Exosomes derived from HBV-infected LO2 cells promote LX-2 cell activation and liver fibrosis in mouse Exosomal miR-222 derived from HBV-infected LO2 cells promotes LX-2 cell activation TFRC is a target of miR-222 and inhibits LX-2 cell activation induced by miR-222 miR-222 promotes LX-2 cell activation through inhibiting TFRC-induced ferroptosis.


Sujet(s)
Exosomes , microARN , Animaux , Souris , Virus de l'hépatite B/génétique , Virus de l'hépatite B/métabolisme , Exosomes/génétique , Exosomes/métabolisme , Hépatocytes/métabolisme , Cirrhose du foie/génétique , Cirrhose du foie/métabolisme , microARN/génétique , microARN/métabolisme , Cellules étoilées du foie/métabolisme , Cellules étoilées du foie/anatomopathologie , Récepteurs à la transferrine/métabolisme
8.
ACS Appl Mater Interfaces ; 14(34): 38497-38505, 2022 Aug 31.
Article de Anglais | MEDLINE | ID: mdl-35977115

RÉSUMÉ

Photodynamic therapy (PDT) has been extensively explored as a noninvasive cancer treatment modality. However, the dilemma of tumor hypoxia and short half-life of singlet oxygen (1O2) severely restrict the therapeutic efficacy of PDT. Herein, we develop a facile three-in-one PDT nanoamplifier (AA@PPa/Hemin NPs) assembled by pyropheophorbide a (PPa), hemin, and arachidonic acid (AA). Interestingly, AA not only acts as an enabler to facilitate the assembly of PPa and hemin in the construction of ternary hybrid nanoassemblies but also acts as a lipid reactive oxygen species (ROS) amplifier for robust PDT. In tumor cells, hemin plays the role of a catalase-like catalyst that accelerates the production of oxygen (O2) from hydrogen peroxide (H2O2), significantly alleviating tumor hypoxia. Under laser irradiation, vast amounts of 1O2 generated by PPa trigger the peroxidation of AA to produce large amounts of cytotoxic lipid ROS, immensely amplifying the efficiency of PDT by promptly eliciting cellular oxidative stress. As expected, AA@PPa/Hemin NPs exert potent antitumor activity in a 4T1 breast-tumor-bearing BALB/c mice xenograft model. Such a cascade nanohybrid amplifier provides a novel codelivery platform for accurate and effective PDT of cancer.


Sujet(s)
Nanoparticules , Photothérapie dynamique , Animaux , Lignée cellulaire tumorale , Hémine , Humains , Peroxyde d'hydrogène , Lipides , Souris , Souris de lignée BALB C , Oxygène , Photosensibilisants/pharmacologie , Photosensibilisants/usage thérapeutique , Espèces réactives de l'oxygène
9.
Front Oncol ; 12: 858480, 2022.
Article de Anglais | MEDLINE | ID: mdl-35898880

RÉSUMÉ

Neuroblastomas are the main extracranial tumors that affect children, while glioblastomas are the most lethal brain tumors, with a median survival time of less than 12 months, and the prognosis of these tumors is poor due to multidrug resistance. Thus, the development of new therapies for the treatment of these types of tumors is urgently needed. In this context, a new type of cell death with strong antitumor potential, called ferroptosis, has recently been described. Ferroptosis is molecularly, morphologically and biochemically different from the other types of cell death described to date because it continues in the absence of classical effectors of apoptosis and does not require the necroptotic machinery. In contrast, ferroptosis has been defined as an iron-dependent form of cell death that is inhibited by glutathione peroxidase 4 (GPX4) activity. Interestingly, ferroptosis can be induced pharmacologically, with potential antitumor activity in vivo and eventual application prospects in translational medicine. Here, we summarize the main pathways of pharmacological ferroptosis induction in tumor cells known to date, along with the limitations of, perspectives on and possible applications of this in the treatment of these tumors.

10.
Biomed Pharmacother ; 153: 113279, 2022 Sep.
Article de Anglais | MEDLINE | ID: mdl-35738177

RÉSUMÉ

Heart disease is the leading cause of death worldwide. Cardiomyopathy is a disease characterized by the heart muscle damage, resulting heart in a structurally and functionally change, as well as heart failure and sudden cardiac death. The key pathogenic factor of cardiomyopathy is the loss of cardiomyocytes, but the related molecular mechanisms remain unclear. Ferroptosis is a newly discovered regulated form of cell death, characterized by iron accumulation and lipid peroxidation during cell death. Recent studies have shown that ferroptosis plays an important regulatory roles in the occurrence and development of many heart diseases such as myocardial ischemia/reperfusion injury, cardiomyopathy and heart failure. However, the systemic association of ferroptosis and cardiomyopathy remains largely unknown and needs to be elucidated. In this review, we provide an overview of the molecular mechanisms of ferroptosis and its role in individual cardiomyopathies, highlight that targeting ferroptosis maybe a potential therapeutic strategy for cardiomyopathy therapy in the future.


Sujet(s)
Cardiomyopathies , Ferroptose , Défaillance cardiaque , Cardiomyopathies/métabolisme , Défaillance cardiaque/métabolisme , Humains , Peroxydation lipidique , Myocytes cardiaques/métabolisme
11.
Acta Pharmacol Sin ; 43(11): 2977-2992, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-35581292

RÉSUMÉ

Glioblastoma (GBM), a malignant brain tumor, is a world-wide health problem because of its poor prognosis and high rates of recurrence and mortality. Apolipoprotein C1 (APOC1) is the smallest of apolipoproteins, implicated in many diseases. Recent studies have shown that APOC1 promotes tumorigenesis and development of several types of cancer. In this study we investigated the role of APOC1 in GBM tumorigenesis. Using in silico assays we showed that APOC1 was highly expressed in GBM tissues and its expression was closely related to GBM progression. We showed that APOC1 protein expression was markedly increased in four GBM cell lines (U251, U138, A172 and U87) compared to the normal brain glia cell lines (HEB, HA1800). In U251 cells, overexpression of APOC1 promoted cell proliferation, migration, invasion and colony information, which was reversed by APOC1 knockdown. APOC1 knockdown also markedly inhibited the growth of GBM xenografts in the ventricle of nude mice. We further demonstrated that APOC1 reduced ferroptosis by inhibiting KEAP1, promoting nuclear translocation of NRF2 and increasing expression of HO-1 and NQO1 in GBM cells. APOC1 also induced ferroptosis resistance by increasing cystathionine beta-synthase (CBS) expression, which promoted trans-sulfuration and increased GSH synthesis, ultimately leading to an increase in glutathione peroxidase-4 (GPX4). Thus, APOC1 plays a key role in GBM tumorigenesis, conferring resistance to ferroptosis, and may be a promising therapeutic target for GBM.


Sujet(s)
Apolipoprotéine C-I , Ferroptose , Glioblastome , Protéine-1 de type kelch associée à ECH , Facteur-2 apparenté à NF-E2 , Animaux , Humains , Souris , Apolipoprotéine C-I/métabolisme , Carcinogenèse/métabolisme , Lignée cellulaire tumorale , Transformation cellulaire néoplasique , Cystathionine beta-synthase/métabolisme , Régulation de l'expression des gènes tumoraux , Glioblastome/métabolisme , Protéine-1 de type kelch associée à ECH/métabolisme , Souris nude , Facteur-2 apparenté à NF-E2/métabolisme
12.
Mol Cell Biol ; 42(6): e0052221, 2022 06 16.
Article de Anglais | MEDLINE | ID: mdl-35499331

RÉSUMÉ

Ferroptosis is a form of regulated cell death resulting predominantly from catastrophic accumulation of lipid reactive oxygen species (ROS). While the antioxidant systems that counter ferroptosis have been well characterized, the mechanism underlying ferroptosis-associated accumulation of lipid ROS remains unclear. In this study, we demonstrated that protein disulfide isomerase (PDI) is a novel mediator of ferroptosis, which is responsible for the accumulation of lipid ROS and ultimately ferroptosis in MDA-MB-231 human breast cancer cells. Treatment with erastin led to a significant increase in inducible nitric oxide synthase (iNOS)-mediated nitric oxide production, which contributes to the accumulation of the death-inducing cellular lipid ROS. Small interfering RNA (siRNA)-mediated PDI knockdown or pharmacological inhibition of PDI's isomerase activity with cystamine strongly suppressed iNOS dimerization and its catalytic activation, subsequently prevented lipid ROS accumulation, and conferred strong protection against erastin-induced ferroptosis. Remarkably, PDI knockdown in MDA-MB-231 cells also largely abrogated the protective effect of cystamine against erastin-induced ferroptotic cell death. Together, these experimental observations demonstrate a noncanonical role of PDI in ferroptosis, which may serve as a potential therapeutic target for ferroptosis-related diseases.


Sujet(s)
Tumeurs du sein , Ferroptose , Tumeurs du sein/génétique , Cystamine , Femelle , Humains , Lipides , Pipérazines , Protein Disulfide-Isomerases/génétique , Petit ARN interférent/génétique , Espèces réactives de l'oxygène/métabolisme
13.
Food Chem Toxicol ; 164: 113046, 2022 Jun.
Article de Anglais | MEDLINE | ID: mdl-35447293

RÉSUMÉ

Exposure to Di (2-ethylhexyl) phthalate (DEHP) has been associated with toxic effects of the reproductive system. However, the exact mechanism remains to be elucidated. In this study we explored the testicular toxicity induced by DEHP, and the probable molecular mechanism in the process. In vivo, the results demonstrated that DEHP affected testosterone levels and blood-testosterone barrier (BTB) integrity and caused ferroptosis. We further demonstrated that DEHP up-regulated the expression of p38α, p-p38α, p53, p-p53, SAT1, ALOX15. This view has also been confirmed in TM4 cells. After pre-treatment with fer-1 or si-MAPK14, the expression of either p53, p-p53, SAT1 and ALOX15 up-regulated by MEHP was inhibited in vitro. Interestingly, p38α can prevent the accumulation of lipid ROS, and the production of lipid ROS in turn promoted the expression of p38α, thus forming a feedback loop during the ferroptosis. In this process, a vicious cycle consisting of p38α, p53, SAT1, ALOX15, lipid ROS was involved. This study provides new mechanistic insights into DEHP-induced toxicity of the reproductive system.


Sujet(s)
Phtalate de bis[2-éthylhexyle] , Ferroptose , Phtalate de bis[2-éthylhexyle]/métabolisme , Phtalate de bis[2-éthylhexyle]/toxicité , Humains , Lipides , Mâle , Espèces réactives de l'oxygène/métabolisme , Testicule/métabolisme , Testostérone/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
14.
Cancer Sci ; 113(7): 2272-2287, 2022 Jul.
Article de Anglais | MEDLINE | ID: mdl-35466502

RÉSUMÉ

The tyrosine kinase inhibitor lenvatinib is used to treat advanced hepatocellular carcinoma (HCC). Ferroptosis is a type of cell death characterized by the iron-dependent accumulation of lethal lipid reactive oxygen species (ROS). Nuclear factor erythroid-derived 2-like 2 (Nrf2) protects HCC cells against ferroptosis. However, the mechanism of lenvatinib-induced cytotoxicity and the relationships between lenvatinib resistance and Nrf2 are unclear. Thus, we investigated the relationship between lenvatinib and ferroptosis and clarified the involvement of Nrf2 in lenvatinib-induced cytotoxicity. Cell viability, lipid ROS levels, and protein expression were measured using Hep3B and HuH7 cells treated with lenvatinib or erastin. We examined these variables after silencing fibroblast growth factor receptor-4 (FGFR4) or Nrf2 and overexpressing-Nrf2. We immunohistochemically evaluated FGFR4 expression in recurrent lesions after resection and clarified the relationship between FGFR4 expression and lenvatinib efficacy. Lenvatinib suppressed system Xc - (xCT) and glutathione peroxidase 4 (GPX4) expression. Inhibition of the cystine import activity of xCT and GPX4 resulted in the accumulation of lipid ROS. Silencing-FGFR4 suppressed xCT and GPX4 expression and increased lipid ROS levels. Nrf2-silenced HCC cells displayed sensitivity to lenvatinib and high lipid ROS levels. In contrast, Nrf2-overexpressing HCC cells displayed resistance to lenvatinib and low lipid ROS levels. The efficacy of lenvatinib was significantly lower in recurrent HCC lesions with low-FGFR4 expression than in those with high-FGFR4 expression. Patients with FGFR4-positive HCC displayed significantly longer progression-free survival than those with FGFR4-negative HCC. Lenvatinib induced ferroptosis by inhibiting FGFR4. Nrf2 is involved in the sensitivity of HCC to lenvatinib.


Sujet(s)
Carcinome hépatocellulaire , Ferroptose , Facteur de croissance fibroblastique de type 4 , Tumeurs du foie , Phénylurées , Quinoléines , Carcinome hépatocellulaire/anatomopathologie , Facteur de croissance fibroblastique de type 4/antagonistes et inhibiteurs , Humains , Lipides , Tumeurs du foie/anatomopathologie , Facteur-2 apparenté à NF-E2/métabolisme , Phénylurées/pharmacologie , Quinoléines/pharmacologie , Espèces réactives de l'oxygène/métabolisme
15.
Plant J ; 110(5): 1370-1381, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35306710

RÉSUMÉ

Inorganic phosphate (Pi) is essential for plant growth. However, Pi is often limiting in soil. Hence, plants have established several mechanisms of response to Pi starvation. One of the important mechanisms is Pi recycling, which includes membrane lipid remodeling and plastid DNA degradation via catabolic enzymes. However, the involvement of other degradation systems in Pi recycling remains unclear. Autophagy, a system for degradation of intracellular components, contributes to recycling of some nutrients, such as nitrogen, carbon, and zinc, under starvation. In the present study, we found that autophagy-deficient mutants depleted Pi early and exhibited severe leaf growth defects under Pi starvation. The main cargo of autophagy induced by early Pi depleted conditions was the endoplasmic reticulum (ER), indicating that ER-phagy, a type of autophagy that selectively degrades the ER, is involved in the response to the early phase of Pi starvation for contribution to Pi recycling. This ER-phagy was suppressed in an INOSITOL-REQUIRING ENZYME 1 double mutant, ire1a ire1b, in which ER stress responses are defective, suggesting that the early Pi starvation induced ER-phagy is induced by ER stress. Furthermore, iron limitation and inhibition of lipid-reactive oxygen species accumulation suppressed the ER-phagy. Interestingly, membrane lipid remodeling, a response to late Pi starvation, was accelerated in the ire1a ire1b under early Pi-depleted conditions. Our findings reveal the existence of two different phases of responses to Pi starvation (i.e. early and late) and indicate that ER stress-mediated ER-phagy is involved in Pi recycling in the early phase to suppress acceleration of the late phase.


Sujet(s)
Stress du réticulum endoplasmique , Fer , Autophagie/physiologie , Réticulum endoplasmique/métabolisme , Stress du réticulum endoplasmique/physiologie , Fer/métabolisme , Lipides membranaires/métabolisme
16.
Mol Ther ; 29(7): 2185-2208, 2021 07 07.
Article de Anglais | MEDLINE | ID: mdl-33794363

RÉSUMÉ

Ferroptosis is an iron- and lipid reactive oxygen species (ROS)-dependent form of programmed cell death that is distinct from other forms of regulatory cell death at the morphological, biological, and genetic levels. Emerging evidence suggests critical roles for ferroptosis in cell metabolism, the redox status, and various diseases, such as cancers, nervous system diseases, and ischemia-reperfusion injury, with ferroptosis-related proteins. Ferroptosis is inhibited in diverse cancer types and functions as a dynamic tumor suppressor in cancer development, indicating that the regulation of ferroptosis can be utilized as an interventional target for tumor treatment. Small molecules and nanomaterials that reprogram cancer cells to undergo ferroptosis are considered effective drugs for cancer therapy. Here, we systematically summarize the molecular basis of ferroptosis, the suppressive effect of ferroptosis on tumors, the effect of ferroptosis on cellular metabolism and the tumor microenvironment (TME), and ferroptosis-inducing agents for tumor therapeutics. An understanding of the latest progress in ferroptosis could provide references for proposing new potential targets for the treatment of cancers.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Ferroptose , Tumeurs/traitement médicamenteux , Microenvironnement tumoral , Animaux , Humains , Tumeurs/immunologie , Tumeurs/anatomopathologie
17.
Cell Cycle ; 19(19): 2411-2425, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32816618

RÉSUMÉ

Human skin melanoma is one of the most aggressive and difficult to treat human malignancies, with an increasing incidence over the years. While the resection of the early diagnosed primary tumor remains the best clinical approach, advanced/metastatic melanoma still remains with a poor prognosis. Indeed, although enormous progress in the therapeutic treatment of human tumors has been made in recent years, patients affected by metastatic melanoma are still poorly affected by these clinical advances. Therefore, new valuable therapeutic approaches are urgently needed, to design and define effective treatments to consistently increase the overall survival rate of patients affected by this malignancy. In this review we summarize the main signaling pathways studied to kill human skin melanoma, and introduce the ferroptotic cell death as a new pathway to be explored to eradicate this tumor.


Sujet(s)
Ferroptose , Mélanome/secondaire , Tumeurs cutanées/anatomopathologie , Animaux , Antinéoplasiques/usage thérapeutique , Ferroptose/effets des médicaments et des substances chimiques , Humains , Mélanome/traitement médicamenteux , Mélanome/métabolisme , Thérapie moléculaire ciblée , Espèces réactives de l'oxygène/métabolisme , Transduction du signal , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/métabolisme
18.
Cell Signal ; 72: 109633, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32283255

RÉSUMÉ

Ferroptosis is a regulated form of cell death characterized by the iron-dependent accumulation of lipid hydroperoxides. Ceruloplasmin (CP) is a glycoprotein that plays an essential role in iron homeostasis. However, whether CP regulates ferroptosis has not been reported. Here, we show that CP suppresses ferroptosis by regulating iron homeostasis in hepatocellular carcinoma (HCC) cells. Depletion of CP promoted erastin- and RSL3-induced ferroptotic cell death and resulted in the accumulation of intracellular ferrous iron (Fe2+) and lipid reactive oxygen species (ROS). Moreover, overexpression of CP suppressed erastin- and RSL3-induced ferroptosis in HCC cells. In addition, a novel frameshift mutation (c.1192-1196del, p.leu398serfs) of CP gene newly identified in patients with iron accumulation and neurodegenerative diseases lost its ability to regulate iron homeostasis and thus failed to participate in the regulation of ferroptosis. Collectively, these data suggest that CP plays an indispensable role in ferroptosis by regulating iron metabolism and indicate a potential therapeutic approach for hepatocellular carcinoma.


Sujet(s)
Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Céruloplasmine/métabolisme , Ferroptose , Homéostasie , Fer/métabolisme , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Antigènes CD/métabolisme , Carbolines/pharmacologie , Transporteurs de cations/métabolisme , Lignée cellulaire tumorale , Ferroptose/effets des médicaments et des substances chimiques , Mutation avec décalage du cadre de lecture/génétique , Homéostasie/effets des médicaments et des substances chimiques , Humains , Modèles biologiques , Pipérazines/pharmacologie , Récepteurs à la transferrine/métabolisme
19.
Liver Int ; 40(6): 1378-1394, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32145145

RÉSUMÉ

BACKGROUND: NASH is one of the fastest growing liver diseases that leads to severe steatosis, inflammation and ultimately liver injury. However, the pathophysiological mechanisms of NASH remain unclear and pharmacological treatment against the disease is unavailable currently. Ferroptosis is a non-apoptotic form of cell death induced by iron-dependent lipid peroxidation. Since NASH progression is accompanied by massive lipid accumulation, which generates lipotoxic species, we investigated the role of ferroptosis in NASH progression. METHOD: Mice were fed on MCD-diet to mimic NASH progression and gene expression in liver was analysed by RNA-seq. The occurrence of hepatic ferroptosis was measured by lipid ROS level, electron microscopy and in vivo PI staining. The beneficial effects of ferroptosis inhibitors on NASH was evaluated by liver pathology analysis. The mechanism of lipid ROS induced lipid droplets accumulation was investigated by in vitro cell culture. RESULTS: RNA-seq analysis suggested that elevated arachidonic acid metabolism promotes ferroptosis in MCD-diet fed mouse livers, which was further demonstrated by lipid ROS accumulation, morphological change of mitochondria and increased cell death. Iron accumulation was detected in the liver and the serum of MCD-fed mice. Scavenging of ferroptosis-linked lipid peroxides reduced lipid accumulation both in vivo and in vitro. Importantly, ferroptosis inhibitors alleviated MCD-diet induced inflammation, fibrogenesis and liver injury. Finally, lipid ROS promotes liver steatosis by boosting lipid droplets formation. CONCLUSION: Our results demonstrate an important role of ferroptosis in the progression of MCD-diet induced NASH and suggest that ferroptosis may serve as a therapeutic target for NASH treatment.


Sujet(s)
Carence en choline , Ferroptose , Stéatose hépatique non alcoolique , Animaux , Choline , Régime alimentaire , Foie , Méthionine , Souris , Souris de lignée C57BL , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/étiologie
20.
Cell Rep ; 29(9): 2731-2744.e4, 2019 Nov 26.
Article de Anglais | MEDLINE | ID: mdl-31775041

RÉSUMÉ

Aerobic organisms need to maintain cellular redox homeostasis. Glutathione peroxidase-4 (Gpx4) has the unique ability to protect cells against lipid peroxidation. Here, we show that Gpx4 is absolutely required to prevent ferroptosis during development, maintenance, and responses of innate-like B cells, namely, the B1 and marginal zone (MZ) B cells. In contrast, Gpx4 is dispensable for the development, germinal center reactions, and antibody responses of follicular B2 cells. Mechanistically, we show increased lipid metabolism and sensitivity to lipid peroxidation and ferroptosis in B1 and MZ B cells compared to follicular B2 cells, consistent with the requirement of Gpx4 in innate-like B cells. This high sensitivity to ferroptosis of innate-like B cells may be used to therapeutically target Gpx4 in certain forms of B cell malignancies involving B1 cells.


Sujet(s)
Lymphocytes B/métabolisme , Protéines du cytosquelette/métabolisme , Ferroptose/effets des médicaments et des substances chimiques , Glutathione peroxidase/usage thérapeutique , Peroxydation lipidique/effets des médicaments et des substances chimiques , Humains
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE